A 66-year-old man with abnormal thyroid function tests

Article Type
Changed
Tue, 10/01/2019 - 08:52
Display Headline
A 66-year-old man with abnormal thyroid function tests

A 66-year-old man presented to the emergency department with increasing shortness of breath and productive cough, which had begun 5 days earlier. Three years previously, he had been diagnosed with chronic obstructive pulmonary disease (COPD).

One week before the current presentation, he developed a sore throat, rhinorrhea, and nasal congestion, and the shortness of breath had started 2 days after that. Although he could speak in sentences, he was breathless even at rest. His dyspnea was associated with noisy breathing and cough productive of yellowish sputum; there was no hemoptysis. He reported fever, but he had no chills, night sweats, chest pain, or paroxysmal nocturnal dyspnea. The review of other systems was unremarkable.

His COPD was known to be mild, in Global Initiative for Chronic Obstructive Lung Disease (GOLD) grade 1, group A. His postbronchodilator ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) was less than 0.70, and his FEV1 was 84% of predicted. Apart from mild intermittent cough with white sputum, his COPD had been under good control with inhaled ipratropium 4 times daily and inhaled albuterol as needed. He said he did not have shortness of breath except when hurrying on level ground or walking up a slight hill (Modified Medical Research Council dyspnea scale grade 1; COPD Assessment Test score < 10). In the last 3 years, he had 2 exacerbations of COPD, 1 year apart, both requiring oral prednisone and antibiotic therapy.

Other relevant history included hypertension and dyslipidemia of 15-year duration, for which he was taking candesartan 16 mg twice daily and atorvastatin 20 mg daily. He was compliant with his medications.

Though he usually received an influenza vaccine every year, he did not get it the previous year. Also, 3 years previously, he received the 23-valent pneumococcal polysaccharide vaccine (PPSV23), and the year before that he received the pneumococcal conjugate vaccine (PCV13). In addition, he was immunized against herpes zoster and tetanus.

The patient had smoked 1 pack per day for the past 38 years. His primary care physician had advised him many times to quit smoking. He had enrolled in a smoking cessation program 2 years previously, in which he received varenicline in addition to behavioral counseling in the form of motivational interviewing and a telephone quit-line. Nevertheless, he continued to smoke.

He was a retired engineer. He did not drink alcohol or use illicit drugs.

PHYSICAL EXAMINATION

On physical examination, the patient was sitting up in bed, leaning forward. He was alert and oriented but was breathing rapidly and looked sick. He had no cyanosis, clubbing, pallor, or jaundice. His blood pressure was 145/90 mm Hg, heart rate 110 beats per minute and regular, respiratory rate 29 breaths per minute, and oral temperature 38.1°C (100.6°F). His oxygen saturation was 88% while breathing room air. His body mass index was 27.1 kg/m2.

His throat was mildly congested. His neck veins were flat, and there were no carotid bruits. His thyroid examination was normal, without goiter, nodules, or tenderness.

Intercostal retractions were noted around the anterolateral costal margins. He had no chest wall deformities. Chest expansion was reduced bilaterally. There was hyperresonance bilaterally. Expiratory wheezes were heard over both lungs, without crackles.

His heart had no murmurs or added sounds. There was no lower-limb edema or swelling. The rest of his physical examination was unremarkable.

alhalaseh_thyroidfunctiontests_t1.jpg
Chest radiography showed hyperinflation without infiltrates. Electrocardiography showed normal sinus rhythm, with a peaked P wave (P pulmonale) and evidence of right ventricular hypertrophy, but no ischemic changes.

Results of initial laboratory testing are shown in Table 1.

Assessment: A 66-year-old man with GOLD grade 1, group A COPD, presenting with a severe exacerbation, most likely due to viral bronchitis.

 

 

INITIAL MANAGEMENT

The patient was given oxygen 28% by Venturi mask, and his oxygen saturation went up to 90%. He was started on nebulized albuterol 2.5 mg with ipratropium bromide 500 µg every 4 hours, prednisone 40 mg orally daily for 5 days, and ceftriaxone 1 g intravenously every 24 hours. The first dose of each medication was given in the emergency department.

The patient was then admitted to a progressive care unit, where he was placed on noninvasive positive pressure ventilation, continuous cardiac monitoring, and pulse oximetry. He was started on enoxaparin 40 mg subcutaneously daily to prevent venous thromboembolism, and the oral medications he had been taking at home were continued. Because he was receiving a glucocorticoid, his blood glucose was monitored in the fasting state, 2 hours after each meal, and as needed.

Two hours after he started noninvasive positive pressure ventilation, his arterial blood gases were remeasured and showed the following results:

  • pH 7.35
  • Partial pressure of carbon dioxide (Paco2) 52 mm Hg
  • Bicarbonate 28 mmol/L
  • Partial pressure of oxygen (Pao2) 60 mm Hg
  • Oxygen saturation 90%.

HOSPITAL COURSE

On hospital day 3, his dyspnea had slightly improved. His respiratory rate was 26 to 28 breaths per minute. His oxygen saturation remained between 90% and 92%.

At 10:21 pm, his cardiac monitor showed an episode of focal atrial tachycardia at a rate of 129 beats per minute that lasted for 3 minutes and 21 seconds, terminating spontaneously. He denied any change in his clinical condition during the episode, with no chest pain, palpitation, or change in dyspnea. There was no change in his vital signs. He had another similar asymptomatic episode lasting 4 minutes and 9 seconds at 6:30 am of hospital day 4.

Because of these episodes, the attending physician ordered thyroid function tests.

THYROID FUNCTION TESTING

1. Which thyroid function test is most likely to be helpful in the assessment of this patient’s thyroid status?

  • Serum thyroid-stimulating hormone (TSH) alone
  • Serum TSH and total thyroxine (T4)
  • Serum TSH and total triiodothyronine (T3)
  • Serum TSH and free T4
  • Serum TSH and free T3

There are several tests to assess thyroid function: the serum TSH, total T4, free T4, total T3, and free T3 concentrations.1

In normal physiology, TSH from the pituitary stimulates the thyroid gland to produce and secrete T4 and T3, which in turn inhibit TSH secretion through negative feedback. A negative log-linear relation exists between serum free T4 and TSH levels.2 Thus, the serum free T4 level can remain within the normal reference range even if the TSH level is high or low. 

TSH assays can have different detection limits. A third-generation TSH assay with a detection limit of 0.01 mU/L is recommended for use in clinical practice.3

TSH testing alone. Given its superior sensitivity and specificity, serum TSH measurement is considered the best single test for assessing thyroid function in most cases.4 Nevertheless, measurement of the serum TSH level alone could be misleading in several situations, eg, hypothalamic or pituitary disorders, recent treatment of thyrotoxicosis, impaired sensitivity to thyroid hormone, and acute nonthyroidal illness.4

alhalaseh_thyroidfunctiontests_t2.jpg
Because our patient is acutely ill, measuring his serum TSH alone is not the most appropriate test of his thyroid function. Euthyroid patients who present with acute illness usually have different patterns of abnormal thyroid function test results, depending on the severity of their illness, its stage, the drugs they are receiving, and other factors. Thyroid function test abnormalities in those patients are shown in Table 2.5–7

Free vs total T4 and T3 levels

Serum total T4 includes a fraction that is bound, mainly to thyroxin-binding globulin, and a very small unbound (free) fraction. The same applies to T3. Only free thyroid hormones represent the “active” fraction available for interaction with their protein receptors in the nucleus.8 Patients with conditions that can affect the thyroid-binding protein concentrations usually have altered serum total T4 and T3 levels, whereas their free hormone concentrations remain normal. Accordingly, measurement of free hormone levels, especially free T4, is usually recommended.

Although equilibrium dialysis is the method most likely to provide an accurate serum free T4 measurement, it is not commonly used because of its limited availability and high cost. Thus, most commercial laboratories use “direct” free T4 measurement or, to a lesser degree, the free T4 index.9 However, none of the currently available free T4 tests actually measure free T4 directly; rather, they estimate it.10

Commercial laboratories can provide a direct free T3 estimate, but it may be less reliable than total T3. If serum T3 measurement is indicated, serum total T3 is usually measured. However, total T3 measurement is rarely indicated for patients with hypothyroidism because it usually remains within the normal reference range.11 Nevertheless, serum total T3 measurement could be useful in patients with T3 toxicosis and in those who are acutely ill.

Accordingly, in acutely ill hospitalized patients like ours, measuring serum TSH using a third-generation assay and free T4 is essential to assess thyroid function. Many clinicians also measure serum total T3.

 

 

CASE CONTINUED: LOW TSH, LOW-NORMAL FREE T4, LOW TOTAL T3

The attending physician ordered serum TSH, free T4, and total T3 measurements, which yielded the following:

  • TSH 0.1 mU/L (0.5–5.0)
  • Total T3 55 ng/dL (80–180)
  • Free T4 0.9 ng/dL (0.9–2.4).

2. Which best explains this patient’s abnormal thyroid test results?

  • His acute illness
  • Central hypothyroidism due to pituitary infarction
  • His albuterol therapy
  • Subclinical thyrotoxicosis
  • Hashimoto thyroiditis

Since euthyroid patients with an acute illness may have abnormal thyroid test results (Table 2),5–7 thyroid function testing is not recommended unless there is a strong indication for it, such as new-onset atrial fibrillation, atrial flutter, or focal atrial tachycardia.1 In such patients, it is important to know whether the test abnormalities represent true thyroid disorder or are the result of a nonthyroidal illness.

alhalaseh_thyroidfunctiontests_f1.jpg
%3Cp%3EFigure%201.%20Peripheral%20conversion%20of%20thyroxine%20(T%3Csub%3E4%3C%2Fsub%3E)%20to%20triiodothyronine%20(T%3Csub%3E3%3C%2Fsub%3E)%2C%20reverse%20T%3Csub%3E3%3C%2Fsub%3E%2C%20and%20diiodothyronine%20(T%3Csub%3E2%3C%2Fsub%3E)%20by%20deiodinase%20types%201%2C%202%2C%20and%203%20(D1%2C%20D2%2C%20D3)%20in%20healthy%20people%20and%20in%20patients%20with%20nonthyroidal%20illness.%3C%2Fp%3E
In healthy people, T4 is converted to T3 (the principal active hormone) by type 1 deiodinase (D1) mainly in the liver and kidneys, whereas this reaction is catalyzed by type 2 deiodinase (D2) in the hypothalamus and pituitary. Type 3 deiodinase (D3) converts T4 to reverse T3, a biologically inactive molecule.12 D1 also mediates conversion of reverse T3 to diiodothyronine (T2) (Figure 1).

alhalaseh_thyroidfunctiontests_t3.jpg
Several conditions and drugs can decrease D1 activity, resulting in low serum T3 concentrations (Table 3). In patients with nonthyroidal illness, decreased D1 activity can be observed as early as the first 24 hours after the onset of the illness and is attributed to increased inflammatory cytokines, free fatty acids, increased endogenous cortisol secretion, and use of certain drugs.13,14 In addition, the reduced D1 activity can decrease the conversion of reverse T3 to T2, resulting in elevated serum reverse T3. Increased D3 activity during an acute illness is another mechanism for elevated serum reverse T3 concentration.15

Thyroid function testing in patients with nonthyroidal illness usually shows low serum total T3, normal or low serum TSH, and normal, low, or high serum free T4. However, transient mild serum TSH elevation can be seen in some patients during the recovery period.16 These abnormalities with their mechanisms are shown in Table 2.5–7 In several commercial kits, serum direct free T4 can be falsely decreased or increased.8

THE DIFFERENTIAL DIAGNOSIS

Our patient had low serum TSH, low-normal serum direct free T4, and low serum total T3. This profile could be caused by a nonthyroidal illness, “true” central hypothyroidism, or his glucocorticoid treatment. The reason we use the term “true” in this setting is that some experts suggest that the thyroid function test abnormalities in patients with acute nonthyroidal illness represent a transient central hypothyroidism.17 The clinical presentation is key in differentiating true central hypothyroidism from nonthyroidal illness.

In addition, measuring serum cortisol may help to differentiate between the 2 states, as it would be elevated in patients with nonthyroidal illness as part of a stress response but low in patients with true central hypothyroidism, since it is usually part of combined pituitary hormone deficiency.18 Of note, some critically ill patients have low serum cortisol because of transient central adrenal insufficiency.19,20

The serum concentration of reverse T3 has been suggested as a way to differentiate between hypothyroidism (low) and nonthyroidal illness (high); however, further studies showed that it does not reliably differentiate between the conditions.21

GLUCOCORTICOIDS AND THYROID FUNCTION TESTS

By inhibiting D1, glucocorticoids can decrease peripheral conversion of T4 to T3 and thus decrease serum total T3. This effect depends on the type and dose of the glucocorticoid and the duration of therapy.

In one study,22 there was a significant reduction in serum total T3 concentration 24 hours after a single oral dose of dexamethasone 12 mg in normal participants. This effect lasted 48 hours, after which serum total T3 returned to its pretreatment level.

In another study,23 a daily oral dose of betamethasone 1.5 mg for 5 days did not significantly reduce the serum total T3 in healthy volunteers, but a daily dose of 3 mg did. This effect was more pronounced at a daily dose of 4.5 mg, whereas a dose of 6.0 mg had no further effect.

Long-term glucocorticoid therapy also decreases serum total T4 and total T3 by lowering serum thyroid-binding globulin.24

Finally, glucocorticoids can decrease TSH secretion by directly inhibiting thyrotropin-releasing hormone.25,26 However, chronic hypercortisolism, whether endogenous or exogenous, does not cause clinically central hypothyroidism, possibly because of the negative feedback mechanism of low thyroid hormones on the pituitary and the hypothalamus.27

Other drugs including dopamine, dopamine agonists, dobutamine, and somatostatin analogues can suppress serum TSH. As with glucocorticoids, these drugs do not cause clinically evident central hypothyroidism.28 Bexarotene, a retinoid X receptor ligand used in the treatment of cutaneous T-cell lymphoma, has been reported to cause clinically evident central hypothyroidism by suppressing TSH and increasing T4 clearance.29

 

 

BETA-BLOCKERS, BETA-AGONISTS AND THYROID FUNCTION

While there is general agreement that beta-adrenergic antagonists (beta-blockers) do not affect the serum TSH concentration, conflicting data have been reported concerning their effect on other thyroid function tests. This may be due to several factors, including dose, duration of therapy, the patient’s thyroid status, and differences in laboratory methodology.30

In studies of propranolol, serum total T4 concentrations did not change or were increased with daily doses of 160 mg or more in both euthyroid participants and hyperthyroid patients31–33; serum total T3 concentrations did not change or were decreased with 40 mg or more daily34; and serum reverse T3 concentrations were increased with daily doses of 80 mg or more.31 It is most likely that propranolol exerts these changes by inhibiting D1 activity in peripheral tissues.

Furthermore, a significant decrease in serum total T3 concentrations was observed in hyperthyroid patients treated with atenolol 100 mg daily, metoprolol 100 mg daily, and alprenolol 100 mg daily, but not with sotalol 80 mg daily or nadolol (up to 240 mg daily).35,36

On the other hand, beta-adrenergic agonists have not been reported to cause significant changes in thyroid function tests.37

SUBCLINICAL THYROTOXICOSIS OR HASHIMOTO THYROIDITIS?

Our patient’s thyroid function test results are more likely due to his nonthyroidal illness and glucocorticoid therapy, as there is no clinical evidence to point to a hypothalamic-pituitary disorder accounting for true central hypothyroidism.

The other options mentioned in question 2 are unlikely to explain our patient’s thyroid function test results.

Subclinical thyrotoxicosis is characterized by suppressed serum TSH, but both serum free T4 and total T3 remain within the normal reference ranges. In addition, the serum TSH level may help to differentiate between thyrotoxicosis and nonthyroidal illness. In the former, serum TSH is usually suppressed (< 0.01 mU/L), whereas in the latter it is usually low but detectable (0.05– 0.3 mU/L).38,39

Hashimoto thyroiditis is a chronic autoimmune thyroid disease characterized by diffuse lymphocytic infiltration of the thyroid gland. Almost all patients with Hashimoto thyroiditis have elevated levels of antibodies to thyroid peroxidase or thyroglobulin.40 Clinically, patients with Hashimoto thyroiditis can either be hypothyroid or have normal thyroid function, which is not the case in our patient.

CASE CONTINUED

An endocrinologist, consulted for a second opinion, agreed that the patient’s thyroid function test results were most likely due to his nonthyroidal illness and glucocorticoid therapy.

3. In view of the endocrinologist’s opinion, which should be the next step in the management of the patient’s thyroid condition?

  • Start levothyroxine (T4) therapy
  • Start liothyronine (T3) therapy
  • Start N-acetylcysteine therapy
  • Start thyrotropin-releasing hormone therapy
  • Remeasure thyroid hormones after full recovery from his acute illness

It is not clear whether the changes in thyroid hormone levels during an acute illness are a pathologic alteration for which thyroid hormone therapy may be beneficial, or a physiologic adaptation for which such therapy would not be indicated.41

However, current data argue against thyroid hormone therapy using T4 or T3 for patients with nonthyroidal illness syndrome (also called euthyroid sick syndrome).42 Indeed, several randomized controlled trials showed that thyroid hormone therapy is not beneficial in such patients and may be detrimental.41,43

Therapies other than thyroid hormone have been investigated to ameliorate thyroid hormone abnormalities in patients with nonthyroidal illness. These include N-acetylcysteine, thyrotropin-releasing hormone therapy, and nutritional support.

Some studies showed that giving N-acetyl­cysteine, an antioxidant, increased serum T3 and decreased serum reverse T3 concentrations in patients with acute myocardial infarction.44 Nevertheless, the mortality rate and length of hospitalization were not affected. Further studies are needed to know whether N-acetylcysteine therapy is beneficial for such patients.

Similarly, a study using a thyrotropin-releasing hormone analogue along with growth hormone-releasing peptide 2 showed an increase in serum TSH, T4, and T3 levels in critically ill patients.45 The benefit of this therapy has yet to be determined. On the other hand, early nutritional support was reported to prevent thyroid hormonal changes in patients postoperatively.46

Measuring thyroid hormone levels after full recovery is the most appropriate next step in our patient, as the changes in thyroid hormone concentrations subside as the acute illness resolves.47

 

 

CASE CONTINUED

The patient continued to improve. On hospital day 6, he was feeling better but still had mild respiratory distress. There had been no further episodes of arrhythmia since day 4. His blood pressure was 136/86 mm Hg, heart rate 88 beats per minute and regular, respiratory rate 18 breaths per minute, and oral temperature 37.1°C. His oxygen saturation was 92% on room air.

Before discharge, he was encouraged to quit smoking. He was offered behavioral counseling and medication therapy, but he only said that he would think about it. He was discharged on oral cefixime for 4 more days and was instructed to switch to a long-acting bronchodilator along with his other home medications and to return in 1 week to have his thyroid hormones checked.

One week later, his laboratory results were:

  • TSH 11.2 mU/L (reference range 0.5–5.0)
  • Free T4 1.2 ng/dL (0.9–2.4)
  • Total T3 92 ng/dL (80–180).

Clinically, the patient was euthyroid, and examination of his thyroid was unremarkable.

4. Based on these last test results, which statement is correct?

  • Levothyroxine therapy should be started
  • His serum TSH elevation is most likely transient
  • Thyroid ultrasonography is strongly indicated
  • A radioactive iodine uptake study should be performed
  • Measurement of thyroid-stimulating immunoglobulins is indicated

During recovery from nonthyroidal illness, some patients may have elevated serum TSH levels that are usually transient and modest (< 20 mU/L).48 Normalization of the thyroid function tests including serum TSH may take several weeks49 or months.50 However, a systematic review found that the likelihood of permanent primary hypothyroidism is high in patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness.51

Ultrasonography is useful for evaluating patients with thyroid nodules or goiter but is of little benefit for patients like ours, in whom the thyroid is normal on examination.

Similarly, a radioactive iodine uptake study is not indicated, as it is principally used to help differentiate between types of thyrotoxicosis. (Radioactive iodine is also used to treat differentiated thyroid cancer.)

Thyroid-stimulating immunoglobins are TSH receptor-stimulating antibodies that cause Graves disease. Nevertheless, measuring them is not routinely indicated for its diagnosis. However, their measurement is of significant help in the diagnosis of Graves disease if a radioactive iodine uptake study cannot be performed (as in pregnancy) and in atypical presentations such as euthyroid Graves ophthalmopathy.52 Other indications for thyroid-stimulating immunoglobin measurement are beyond the scope of the article. Our patient’s test results are not consistent with hyperthyroidism, so measuring thyroid-stimulating immunoglobins is not indicated.

CASE CONCLUSION: BETTER, BUT STILL SMOKING

The patient missed his 1-month clinic follow-up, but he visited the clinic for follow-up 3 months later. He was feeling well with no complaints. Test results including serum TSH, free T4, and total T3 were within normal ranges. His COPD was under control, with an FEV1 88% of predicted.

He was again encouraged to quit smoking and was offered drug therapy and behavioral counseling, but he declined. In addition, he was instructed to adhere to his annual influenza vaccination.

KEY POINTS

  • In patients with acute illness, it is recommended that thyroid function not be assessed unless there is a strong indication.
  • If thyroid function assessment is indicated for critically ill patients, serum TSH and free T4 concentrations should be measured. Some clinicians also measure serum total T3 level.
  • Thyroid function testing in critically ill patients usually shows low serum total T3, normal or low serum TSH, and normal or low serum free T4.
  • Many drugs can alter thyroid hormone levels.
  • Thyroid hormone therapy is not recommended for critically ill patients with low T3, low T4, or both.
  • During recovery from nonthyroidal illness, some patients may have mild elevation in serum TSH levels (< 20 mU/L).
  • Thyroid hormone levels may take several weeks or months to return to normal after the acute illness.
  • Patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness are more likely to have permanent primary hypothyroidism.
References
  1. Lamb EJ, Martin J. Thyroid function tests: often justified in the acutely ill. Ann Clin Biochem 2000; 37(pt 2):158–164. doi:10.1258/0004563001899159
  2. Spencer CA, LoPresti JS, Patel A, et al. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990; 70(2):453–460. doi:10.1210/jcem-70-2-453
  3. Ross DS, Ardisson LJ, Meskell MJ. Measurement of thyrotropin in clinical and subclinical hyperthyroidism using a new chemiluminescent assay. J Clin Endocrinol Metab 1989; 69(3):684–688. doi:10.1210/jcem-69-3-684
  4. Koulouri O, Moran C, Halsall D, Chatterjee K, Gurnell M. Pitfalls in the measurement and interpretation of thyroid function tests. Best Pract Res Clin Endocrinol Metab 2013; 27(6):745–762. doi:10.1016/j.beem.2013.10.003
  5. Lechan RM, Fekete C. Role of thyroid hormone deiodination in the hypothalamus. Thyroid 2005; 15(8):883–897. doi:10.1089/thy.2005.15.883
  6. Chopra IJ, Hershman JM, Pardridge WM, Nicoloff JT. Thyroid function in nonthyroidal ilnesses. Ann Intern Med 1983; 98(6):946–957. doi:10.7326/0003-4819-98-6-946
  7. Chopra IJ, Solomon DH, Hepner HW, Mortenstein AA. Misleadingly low free thyroxine index and usefulness of reverse triiodothyronine measurement in nonthyroidal illnesses. Ann Intern Med 1979; 90(6):905–912. doi:10.7326/0003-4819-90-6-905
  8. Pontecorvi A, Robbins J. The plasma membrane and thyroid hormone entry into cells. Trends Endocrinol Metab 1989; 1(2):90–94. pmid:18411097
  9. Hennemann G, Krenning EP. Pitfalls in the interpretation of thyroid function tests in old age and non-thyroidal illness. Horm Res 1987; 26(1–4):100–104. doi:10.1159/000180688
  10. Baloch Z, Carayon P, Conte-Devolx B, et al; Guidelines Committee, National Academy of Clinical Biochemistry. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid 2003; 13(1):3–126. doi:10.1089/105072503321086962
  11. Lum S, Nicoloff JT, Spencer CA, Kaptein EM. Peripheral tissue mechanism for maintenance of serum triiodothyronine values in a thyroxine-deficient state in man. J Clin Invest 1984; 73(2):570–575. doi:10.1172/JCI111245
  12. Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-pituitary-thyroid axis. Compr Physiol 2016; 6(3):1387–1428. doi:10.1002/cphy.c150027
  13. de Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol 2015; 225(3):R67–R81. doi:10.1530/JOE-15-0133
  14. Chopra IJ, Huang TS, Beredo A, Solomon DH, Teco GN, Mean JF. Evidence for an inhibitor of extrathyroidal conversion of thyroxine to 3, 5, 3'-triiodothyronine in sera of patients with nonthyroidal illnesses. J Clin Endocrinol Metab 1985; 60(4):666–672. doi:10.1210/jcem-60-4-666
  15. Peeters RP, Debaveye Y, Fliers E, Visser TJ. Changes within the thyroid axis during critical illness. Crit Care Clin 2006; 22(1):41–55. doi:10.1016/j.ccc.2005.08.006
  16. Spencer C, Eigen A, Shen D, et al. Specificity of sensitive assays of thyrotropin (TSH) used to screen for thyroid disease in hospitalized patients. Clin Chem 1987; 33(8):1391–1396. pmid:3301067
  17. Adler SM, Wartofsky L. The nonthyroidal illness syndrome. Endocrinol Metab Clin North Am 2007; 36(3):657–672. doi:10.1016/j.ecl.2007.04.007
  18. Persani L. Central hypothyroidism: pathogenic, diagnostic, and therapeutic challenges. J Clin Endocrinol Metab 2012; 97(9):3068–3078. doi:10.1210/jc.2012-1616
  19. Kidess AI, Caplan RH, Reynertson RH, Wickus GG, Goodnough DE. Transient corticotropin deficiency in critical illness. Mayo Clin Proc 1993; 68(5):435–441. doi:10.1016/s0025-6196(12)60188-8
  20. Lamberts SW, Bruining HA, De Jong FH. Corticosteroid therapy in severe illness. N Engl J Med 1997; 337(18):1285–1292. doi:10.1056/NEJM199710303371807
  21. Burmeister LA. Reverse T3 does not reliably differentiate hypothyroid sick syndrome from euthyroid sick syndrome. Thyroid 1995; 5(6):435–441. doi:10.1089/thy.1995.5.435
  22. Duick DS, Warren DW, Nicoloff JT, Otis CL, Croxson MS. Effect of single dose dexamethasone on the concentration of serum triiodothyronine in man. J Clin Endocrinol Metab 1974; 39(6):1151–1154. doi:10.1210/jcem-39-6-1151
  23. Gamstedt A, Järnerot G, Kågedal B. Dose related effects of betamethasone on iodothyronines and thyroid hormone-binding proteins in serum. Acta Endocrinol (Copenh) 1981; 96(4):484–490. doi:10.1530/acta.0.0960484
  24. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the “euthyroid sick syndrome.” Endocr Rev 1982; 3(2):164–217. doi:10.1210/edrv-3-2-164
  25. Wilber JF, Utiger RD. The effect of glucocorticoids on thyrotropin secretion. J Clin Invest 1969; 48(11):2096–2103. doi:10.1172/JCI106176
  26. Nicoloff JT, Fisher DA, Appleman MD Jr. The role of glucocorticoids in the regulation of thyroid function in man. J Clin Invest 1970; 49(10):1922–1929. doi:10.1172/JCI106411
  27. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med 1995; 333(25):1688–1694. doi:10.1056/NEJM199512213332507
  28. Haugen BR. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab 2009; 23(6):793–800. doi:10.1016/j.beem.2009.08.003
  29. Sherman SI, Gopal J, Haugen BR, et al. Central hypothyroidism associated with retinoid X receptor–selective ligands. N Engl J Med 1999; 340(14):1075–1079. doi:10.1056/NEJM199904083401404
  30. Murchison LE, How J, Bewsher PD. Comparison of propranolol and metoprolol in the management of hyperthyroidism. Br J Clin Pharmacol 1979; 8(6):581–587. doi:10.1111/j.1365-2125.1979.tb01048.x
  31. Faber J, Friis T, Kirkegaard C, et al. Serum T4, T3 and reverse T3 during treatment with propranolol in hyperthyroidism, L-T4 treated myxedema and in normal man. Horm Metab Res 1979; 11(1):34–36. doi:10.1055/s-0028-1092678
  32. Kristensen BO, Weeke J. Propranolol-induced increments in total and free serum thyroxine in patients with essential hypertension. Clin Pharmacol Ther 1977; 22(6):864–867. doi:10.1002/cpt1977226864
  33. Murchison LE, Bewsher PD, Chesters MI, Ferrier WR. Comparison of propranolol and practolol in the management of hyperthyroidism. Br J Clin Pharmacol 1976; 3(2):273–277. doi:10.1111/j.1365-2125.1976.tb00603.x
  34. Lotti G, Delitala G, Devilla L, Alagna S, Masala A. Reduction of plasma triiodothyronine (T3) induced by propranolol. Clin Endocrinol 1977; 6(6):405–410. doi:10.1111/j.1365-2265.1977.tb03322.x
  35. Perrild H, Hansen JM, Skovsted L, Christensen LK. Different effects of propranolol, alprenolol, sotalol, atenolol and metoprolol on serum T3 and serum rT3 in hyperthyroidism. Clin Endocrinol (Oxf) 1983; 18(2):139–142. pmid:6133659
  36. Reeves RA, From GL, Paul W, Leenen FH. Nadolol, propranolol, and thyroid hormones: evidence for a membrane-stabilizing action of propranolol. Clin Pharmacol Ther 1985; 37(2):157–161. doi:10.1038/clpt.1985.28
  37. Walker N, Jung RT, Jennings G, James WP. The effect of a beta-receptor agonist (salbutamol) on peripheral thyroid metabolism in euthyroid subjects. Horm Metab Res 1981; 13(10):590–591. doi:10.1055/s-2007-1019346
  38. Melmed S, Geola FL, Reed AW, Pekary AE, Park J, Hershman JM. A comparison of methods for assessing thyroid function in nonthyroidal illness. J Clin Endocrinol Metab 1982; 54(2):300–306. doi:10.1210/jcem-54-2-300
  39. Docter R, Krenning E, De Jong M, Hennemann G. The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism. Clin Endocrinol (Oxf) 1993; 39(5):499–518. pmid:8252737
  40. Mariotti S, Caturegli P, Piccolo P, Barbesino G, Pinchera A. Antithyroid peroxidase autoantibodies in thyroid diseases. J Clin Endocrinol Metab 1990; 71(3):661–669. doi:10.1210/jcem-71-3-661
  41. De Groot LJ. Non-thyroidal illness syndrome is a manifestation of hypothalamic-pituitary dysfunction, and in view of current evidence, should be treated with appropriate replacement therapies. Crit Care Clin 2006; 22(1):57–86. doi:10.1016/j.ccc.2005.10.001
  42. Jonklaas J, Bianco AC, Bauer AJ, et al; American Thyroid Association Task Force on Thyroid Hormone Replacement. Guidelines for the treatment of hypothyroidism: prepared by the American Thyroid Association Task Force on Thyroid Hormone Replacement. Thyroid 2014; 24(12):1670–1751. doi:10.1089/thy.2014.0028
  43. Kaptein EM, Beale E, Chan LS. Thyroid hormone therapy for obesity and nonthyroidal illnesses: a systematic review. J Clin Endocrinol Metab 2009; 94(10):3663–3675. doi:10.1210/jc.2009-0899
  44. Vidart J, Wajner SM, Leite RS, et al. N-acetylcysteine administration prevents nonthyroidal illness syndrome in patients with acute myocardial infarction: a randomized clinical trial. J Clin Endocrinol Metab 2014; 99(12):4537–4545. doi:10.1210/jc.2014-2192
  45. Van den Berghe G, Wouters P, Weekers F, et al. Reactivation of pituitary hormone release and metabolic improvement by infusion of growth hormone-releasing peptide and thyrotropin-releasing hormone in patients with protracted critical illness. J Clin Endocrinol Metab 1999; 84(4):1311–1323. doi:10.1210/jcem.84.4.5636
  46. Langouche L, Vander Perre S, Marques M, et al. Impact of early nutrient restriction during critical illness on the nonthyroidal illness syndrome and its relation with outcome: a randomized, controlled clinical study. J Clin Endocrinol Metab 2013; 98(3):1006–1013. doi:10.1210/jc.2012-2809
  47. Economidou F, Douka E, Tzanela M, Nanas S, Kotanidou A. Thyroid function during critical illness. Hormones (Athens) 2011; 10(2):117–124. doi:10.14310/horm.2002.1301
  48. Hamblin PS, Dyer SA, Mohr VS, et al. Relationship between thyrotropin and thyroxine changes during recovery from severe hypothyroxinemia of critical illness. J Clin Endocrinol Metab 1986; 62(4):717–722. doi:10.1210/jcem-62-4-717
  49. Iglesias P, Diez JJ. Thyroid dysfunction and kidney disease. Eur J Endocrinol 2009; 160(4):503–515. doi:10.1530/EJE-08-0837
  50. Spencer CA. Clinical utility and cost-effectiveness of sensitive thyrotropin assays in ambulatory and hospitalized patients. Mayo Clin Proc 1988; 63(12):1214–1222. doi:10.1016/s0025-6196(12)65408-1
  51. Attia J, Margetts P, Guyatt G. Diagnosis of thyroid disease in hospitalized patients: a systematic review. Arch Intern Med 1999; 159(7):658–665. pmid:10218744
  52. Barbesino G, Tomer Y. Clinical review: clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab 2013; 98(6):2247–2255. doi:10.1210/jc.2012-4309
Article PDF
Author and Disclosure Information

Yazan N. Alhalaseh, MD
Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan

Zaid A. Abdulelah, MD
Istishari Hospital, Amman, Jordan

Ahmad O. Armouti, MD
King Hussein Medical Center, Amman, Jordan

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

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

Issue
Cleveland Clinic Journal of Medicine - 86(10)
Publications
Topics
Page Number
666-675
Legacy Keywords
thyroid, thyroid function tests, hypothyroid, chronic obstructive pulmonary disease, COPD, thyroid-stimulating hormone, TSH, thyroxine, T4, triiodothyronine, T3, reverse T3, deiodinase, D1, euthyroid sick syndrome, nonthyroidal illness syndrome, Yazan Alhalaseh, Zaid Abdulelah, Ahmad Armouti, Ayman Zayed
Sections
Author and Disclosure Information

Yazan N. Alhalaseh, MD
Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan

Zaid A. Abdulelah, MD
Istishari Hospital, Amman, Jordan

Ahmad O. Armouti, MD
King Hussein Medical Center, Amman, Jordan

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

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

Author and Disclosure Information

Yazan N. Alhalaseh, MD
Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan

Zaid A. Abdulelah, MD
Istishari Hospital, Amman, Jordan

Ahmad O. Armouti, MD
King Hussein Medical Center, Amman, Jordan

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

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

Article PDF
Article PDF
Related Articles

A 66-year-old man presented to the emergency department with increasing shortness of breath and productive cough, which had begun 5 days earlier. Three years previously, he had been diagnosed with chronic obstructive pulmonary disease (COPD).

One week before the current presentation, he developed a sore throat, rhinorrhea, and nasal congestion, and the shortness of breath had started 2 days after that. Although he could speak in sentences, he was breathless even at rest. His dyspnea was associated with noisy breathing and cough productive of yellowish sputum; there was no hemoptysis. He reported fever, but he had no chills, night sweats, chest pain, or paroxysmal nocturnal dyspnea. The review of other systems was unremarkable.

His COPD was known to be mild, in Global Initiative for Chronic Obstructive Lung Disease (GOLD) grade 1, group A. His postbronchodilator ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) was less than 0.70, and his FEV1 was 84% of predicted. Apart from mild intermittent cough with white sputum, his COPD had been under good control with inhaled ipratropium 4 times daily and inhaled albuterol as needed. He said he did not have shortness of breath except when hurrying on level ground or walking up a slight hill (Modified Medical Research Council dyspnea scale grade 1; COPD Assessment Test score < 10). In the last 3 years, he had 2 exacerbations of COPD, 1 year apart, both requiring oral prednisone and antibiotic therapy.

Other relevant history included hypertension and dyslipidemia of 15-year duration, for which he was taking candesartan 16 mg twice daily and atorvastatin 20 mg daily. He was compliant with his medications.

Though he usually received an influenza vaccine every year, he did not get it the previous year. Also, 3 years previously, he received the 23-valent pneumococcal polysaccharide vaccine (PPSV23), and the year before that he received the pneumococcal conjugate vaccine (PCV13). In addition, he was immunized against herpes zoster and tetanus.

The patient had smoked 1 pack per day for the past 38 years. His primary care physician had advised him many times to quit smoking. He had enrolled in a smoking cessation program 2 years previously, in which he received varenicline in addition to behavioral counseling in the form of motivational interviewing and a telephone quit-line. Nevertheless, he continued to smoke.

He was a retired engineer. He did not drink alcohol or use illicit drugs.

PHYSICAL EXAMINATION

On physical examination, the patient was sitting up in bed, leaning forward. He was alert and oriented but was breathing rapidly and looked sick. He had no cyanosis, clubbing, pallor, or jaundice. His blood pressure was 145/90 mm Hg, heart rate 110 beats per minute and regular, respiratory rate 29 breaths per minute, and oral temperature 38.1°C (100.6°F). His oxygen saturation was 88% while breathing room air. His body mass index was 27.1 kg/m2.

His throat was mildly congested. His neck veins were flat, and there were no carotid bruits. His thyroid examination was normal, without goiter, nodules, or tenderness.

Intercostal retractions were noted around the anterolateral costal margins. He had no chest wall deformities. Chest expansion was reduced bilaterally. There was hyperresonance bilaterally. Expiratory wheezes were heard over both lungs, without crackles.

His heart had no murmurs or added sounds. There was no lower-limb edema or swelling. The rest of his physical examination was unremarkable.

alhalaseh_thyroidfunctiontests_t1.jpg
Chest radiography showed hyperinflation without infiltrates. Electrocardiography showed normal sinus rhythm, with a peaked P wave (P pulmonale) and evidence of right ventricular hypertrophy, but no ischemic changes.

Results of initial laboratory testing are shown in Table 1.

Assessment: A 66-year-old man with GOLD grade 1, group A COPD, presenting with a severe exacerbation, most likely due to viral bronchitis.

 

 

INITIAL MANAGEMENT

The patient was given oxygen 28% by Venturi mask, and his oxygen saturation went up to 90%. He was started on nebulized albuterol 2.5 mg with ipratropium bromide 500 µg every 4 hours, prednisone 40 mg orally daily for 5 days, and ceftriaxone 1 g intravenously every 24 hours. The first dose of each medication was given in the emergency department.

The patient was then admitted to a progressive care unit, where he was placed on noninvasive positive pressure ventilation, continuous cardiac monitoring, and pulse oximetry. He was started on enoxaparin 40 mg subcutaneously daily to prevent venous thromboembolism, and the oral medications he had been taking at home were continued. Because he was receiving a glucocorticoid, his blood glucose was monitored in the fasting state, 2 hours after each meal, and as needed.

Two hours after he started noninvasive positive pressure ventilation, his arterial blood gases were remeasured and showed the following results:

  • pH 7.35
  • Partial pressure of carbon dioxide (Paco2) 52 mm Hg
  • Bicarbonate 28 mmol/L
  • Partial pressure of oxygen (Pao2) 60 mm Hg
  • Oxygen saturation 90%.

HOSPITAL COURSE

On hospital day 3, his dyspnea had slightly improved. His respiratory rate was 26 to 28 breaths per minute. His oxygen saturation remained between 90% and 92%.

At 10:21 pm, his cardiac monitor showed an episode of focal atrial tachycardia at a rate of 129 beats per minute that lasted for 3 minutes and 21 seconds, terminating spontaneously. He denied any change in his clinical condition during the episode, with no chest pain, palpitation, or change in dyspnea. There was no change in his vital signs. He had another similar asymptomatic episode lasting 4 minutes and 9 seconds at 6:30 am of hospital day 4.

Because of these episodes, the attending physician ordered thyroid function tests.

THYROID FUNCTION TESTING

1. Which thyroid function test is most likely to be helpful in the assessment of this patient’s thyroid status?

  • Serum thyroid-stimulating hormone (TSH) alone
  • Serum TSH and total thyroxine (T4)
  • Serum TSH and total triiodothyronine (T3)
  • Serum TSH and free T4
  • Serum TSH and free T3

There are several tests to assess thyroid function: the serum TSH, total T4, free T4, total T3, and free T3 concentrations.1

In normal physiology, TSH from the pituitary stimulates the thyroid gland to produce and secrete T4 and T3, which in turn inhibit TSH secretion through negative feedback. A negative log-linear relation exists between serum free T4 and TSH levels.2 Thus, the serum free T4 level can remain within the normal reference range even if the TSH level is high or low. 

TSH assays can have different detection limits. A third-generation TSH assay with a detection limit of 0.01 mU/L is recommended for use in clinical practice.3

TSH testing alone. Given its superior sensitivity and specificity, serum TSH measurement is considered the best single test for assessing thyroid function in most cases.4 Nevertheless, measurement of the serum TSH level alone could be misleading in several situations, eg, hypothalamic or pituitary disorders, recent treatment of thyrotoxicosis, impaired sensitivity to thyroid hormone, and acute nonthyroidal illness.4

alhalaseh_thyroidfunctiontests_t2.jpg
Because our patient is acutely ill, measuring his serum TSH alone is not the most appropriate test of his thyroid function. Euthyroid patients who present with acute illness usually have different patterns of abnormal thyroid function test results, depending on the severity of their illness, its stage, the drugs they are receiving, and other factors. Thyroid function test abnormalities in those patients are shown in Table 2.5–7

Free vs total T4 and T3 levels

Serum total T4 includes a fraction that is bound, mainly to thyroxin-binding globulin, and a very small unbound (free) fraction. The same applies to T3. Only free thyroid hormones represent the “active” fraction available for interaction with their protein receptors in the nucleus.8 Patients with conditions that can affect the thyroid-binding protein concentrations usually have altered serum total T4 and T3 levels, whereas their free hormone concentrations remain normal. Accordingly, measurement of free hormone levels, especially free T4, is usually recommended.

Although equilibrium dialysis is the method most likely to provide an accurate serum free T4 measurement, it is not commonly used because of its limited availability and high cost. Thus, most commercial laboratories use “direct” free T4 measurement or, to a lesser degree, the free T4 index.9 However, none of the currently available free T4 tests actually measure free T4 directly; rather, they estimate it.10

Commercial laboratories can provide a direct free T3 estimate, but it may be less reliable than total T3. If serum T3 measurement is indicated, serum total T3 is usually measured. However, total T3 measurement is rarely indicated for patients with hypothyroidism because it usually remains within the normal reference range.11 Nevertheless, serum total T3 measurement could be useful in patients with T3 toxicosis and in those who are acutely ill.

Accordingly, in acutely ill hospitalized patients like ours, measuring serum TSH using a third-generation assay and free T4 is essential to assess thyroid function. Many clinicians also measure serum total T3.

 

 

CASE CONTINUED: LOW TSH, LOW-NORMAL FREE T4, LOW TOTAL T3

The attending physician ordered serum TSH, free T4, and total T3 measurements, which yielded the following:

  • TSH 0.1 mU/L (0.5–5.0)
  • Total T3 55 ng/dL (80–180)
  • Free T4 0.9 ng/dL (0.9–2.4).

2. Which best explains this patient’s abnormal thyroid test results?

  • His acute illness
  • Central hypothyroidism due to pituitary infarction
  • His albuterol therapy
  • Subclinical thyrotoxicosis
  • Hashimoto thyroiditis

Since euthyroid patients with an acute illness may have abnormal thyroid test results (Table 2),5–7 thyroid function testing is not recommended unless there is a strong indication for it, such as new-onset atrial fibrillation, atrial flutter, or focal atrial tachycardia.1 In such patients, it is important to know whether the test abnormalities represent true thyroid disorder or are the result of a nonthyroidal illness.

alhalaseh_thyroidfunctiontests_f1.jpg
%3Cp%3EFigure%201.%20Peripheral%20conversion%20of%20thyroxine%20(T%3Csub%3E4%3C%2Fsub%3E)%20to%20triiodothyronine%20(T%3Csub%3E3%3C%2Fsub%3E)%2C%20reverse%20T%3Csub%3E3%3C%2Fsub%3E%2C%20and%20diiodothyronine%20(T%3Csub%3E2%3C%2Fsub%3E)%20by%20deiodinase%20types%201%2C%202%2C%20and%203%20(D1%2C%20D2%2C%20D3)%20in%20healthy%20people%20and%20in%20patients%20with%20nonthyroidal%20illness.%3C%2Fp%3E
In healthy people, T4 is converted to T3 (the principal active hormone) by type 1 deiodinase (D1) mainly in the liver and kidneys, whereas this reaction is catalyzed by type 2 deiodinase (D2) in the hypothalamus and pituitary. Type 3 deiodinase (D3) converts T4 to reverse T3, a biologically inactive molecule.12 D1 also mediates conversion of reverse T3 to diiodothyronine (T2) (Figure 1).

alhalaseh_thyroidfunctiontests_t3.jpg
Several conditions and drugs can decrease D1 activity, resulting in low serum T3 concentrations (Table 3). In patients with nonthyroidal illness, decreased D1 activity can be observed as early as the first 24 hours after the onset of the illness and is attributed to increased inflammatory cytokines, free fatty acids, increased endogenous cortisol secretion, and use of certain drugs.13,14 In addition, the reduced D1 activity can decrease the conversion of reverse T3 to T2, resulting in elevated serum reverse T3. Increased D3 activity during an acute illness is another mechanism for elevated serum reverse T3 concentration.15

Thyroid function testing in patients with nonthyroidal illness usually shows low serum total T3, normal or low serum TSH, and normal, low, or high serum free T4. However, transient mild serum TSH elevation can be seen in some patients during the recovery period.16 These abnormalities with their mechanisms are shown in Table 2.5–7 In several commercial kits, serum direct free T4 can be falsely decreased or increased.8

THE DIFFERENTIAL DIAGNOSIS

Our patient had low serum TSH, low-normal serum direct free T4, and low serum total T3. This profile could be caused by a nonthyroidal illness, “true” central hypothyroidism, or his glucocorticoid treatment. The reason we use the term “true” in this setting is that some experts suggest that the thyroid function test abnormalities in patients with acute nonthyroidal illness represent a transient central hypothyroidism.17 The clinical presentation is key in differentiating true central hypothyroidism from nonthyroidal illness.

In addition, measuring serum cortisol may help to differentiate between the 2 states, as it would be elevated in patients with nonthyroidal illness as part of a stress response but low in patients with true central hypothyroidism, since it is usually part of combined pituitary hormone deficiency.18 Of note, some critically ill patients have low serum cortisol because of transient central adrenal insufficiency.19,20

The serum concentration of reverse T3 has been suggested as a way to differentiate between hypothyroidism (low) and nonthyroidal illness (high); however, further studies showed that it does not reliably differentiate between the conditions.21

GLUCOCORTICOIDS AND THYROID FUNCTION TESTS

By inhibiting D1, glucocorticoids can decrease peripheral conversion of T4 to T3 and thus decrease serum total T3. This effect depends on the type and dose of the glucocorticoid and the duration of therapy.

In one study,22 there was a significant reduction in serum total T3 concentration 24 hours after a single oral dose of dexamethasone 12 mg in normal participants. This effect lasted 48 hours, after which serum total T3 returned to its pretreatment level.

In another study,23 a daily oral dose of betamethasone 1.5 mg for 5 days did not significantly reduce the serum total T3 in healthy volunteers, but a daily dose of 3 mg did. This effect was more pronounced at a daily dose of 4.5 mg, whereas a dose of 6.0 mg had no further effect.

Long-term glucocorticoid therapy also decreases serum total T4 and total T3 by lowering serum thyroid-binding globulin.24

Finally, glucocorticoids can decrease TSH secretion by directly inhibiting thyrotropin-releasing hormone.25,26 However, chronic hypercortisolism, whether endogenous or exogenous, does not cause clinically central hypothyroidism, possibly because of the negative feedback mechanism of low thyroid hormones on the pituitary and the hypothalamus.27

Other drugs including dopamine, dopamine agonists, dobutamine, and somatostatin analogues can suppress serum TSH. As with glucocorticoids, these drugs do not cause clinically evident central hypothyroidism.28 Bexarotene, a retinoid X receptor ligand used in the treatment of cutaneous T-cell lymphoma, has been reported to cause clinically evident central hypothyroidism by suppressing TSH and increasing T4 clearance.29

 

 

BETA-BLOCKERS, BETA-AGONISTS AND THYROID FUNCTION

While there is general agreement that beta-adrenergic antagonists (beta-blockers) do not affect the serum TSH concentration, conflicting data have been reported concerning their effect on other thyroid function tests. This may be due to several factors, including dose, duration of therapy, the patient’s thyroid status, and differences in laboratory methodology.30

In studies of propranolol, serum total T4 concentrations did not change or were increased with daily doses of 160 mg or more in both euthyroid participants and hyperthyroid patients31–33; serum total T3 concentrations did not change or were decreased with 40 mg or more daily34; and serum reverse T3 concentrations were increased with daily doses of 80 mg or more.31 It is most likely that propranolol exerts these changes by inhibiting D1 activity in peripheral tissues.

Furthermore, a significant decrease in serum total T3 concentrations was observed in hyperthyroid patients treated with atenolol 100 mg daily, metoprolol 100 mg daily, and alprenolol 100 mg daily, but not with sotalol 80 mg daily or nadolol (up to 240 mg daily).35,36

On the other hand, beta-adrenergic agonists have not been reported to cause significant changes in thyroid function tests.37

SUBCLINICAL THYROTOXICOSIS OR HASHIMOTO THYROIDITIS?

Our patient’s thyroid function test results are more likely due to his nonthyroidal illness and glucocorticoid therapy, as there is no clinical evidence to point to a hypothalamic-pituitary disorder accounting for true central hypothyroidism.

The other options mentioned in question 2 are unlikely to explain our patient’s thyroid function test results.

Subclinical thyrotoxicosis is characterized by suppressed serum TSH, but both serum free T4 and total T3 remain within the normal reference ranges. In addition, the serum TSH level may help to differentiate between thyrotoxicosis and nonthyroidal illness. In the former, serum TSH is usually suppressed (< 0.01 mU/L), whereas in the latter it is usually low but detectable (0.05– 0.3 mU/L).38,39

Hashimoto thyroiditis is a chronic autoimmune thyroid disease characterized by diffuse lymphocytic infiltration of the thyroid gland. Almost all patients with Hashimoto thyroiditis have elevated levels of antibodies to thyroid peroxidase or thyroglobulin.40 Clinically, patients with Hashimoto thyroiditis can either be hypothyroid or have normal thyroid function, which is not the case in our patient.

CASE CONTINUED

An endocrinologist, consulted for a second opinion, agreed that the patient’s thyroid function test results were most likely due to his nonthyroidal illness and glucocorticoid therapy.

3. In view of the endocrinologist’s opinion, which should be the next step in the management of the patient’s thyroid condition?

  • Start levothyroxine (T4) therapy
  • Start liothyronine (T3) therapy
  • Start N-acetylcysteine therapy
  • Start thyrotropin-releasing hormone therapy
  • Remeasure thyroid hormones after full recovery from his acute illness

It is not clear whether the changes in thyroid hormone levels during an acute illness are a pathologic alteration for which thyroid hormone therapy may be beneficial, or a physiologic adaptation for which such therapy would not be indicated.41

However, current data argue against thyroid hormone therapy using T4 or T3 for patients with nonthyroidal illness syndrome (also called euthyroid sick syndrome).42 Indeed, several randomized controlled trials showed that thyroid hormone therapy is not beneficial in such patients and may be detrimental.41,43

Therapies other than thyroid hormone have been investigated to ameliorate thyroid hormone abnormalities in patients with nonthyroidal illness. These include N-acetylcysteine, thyrotropin-releasing hormone therapy, and nutritional support.

Some studies showed that giving N-acetyl­cysteine, an antioxidant, increased serum T3 and decreased serum reverse T3 concentrations in patients with acute myocardial infarction.44 Nevertheless, the mortality rate and length of hospitalization were not affected. Further studies are needed to know whether N-acetylcysteine therapy is beneficial for such patients.

Similarly, a study using a thyrotropin-releasing hormone analogue along with growth hormone-releasing peptide 2 showed an increase in serum TSH, T4, and T3 levels in critically ill patients.45 The benefit of this therapy has yet to be determined. On the other hand, early nutritional support was reported to prevent thyroid hormonal changes in patients postoperatively.46

Measuring thyroid hormone levels after full recovery is the most appropriate next step in our patient, as the changes in thyroid hormone concentrations subside as the acute illness resolves.47

 

 

CASE CONTINUED

The patient continued to improve. On hospital day 6, he was feeling better but still had mild respiratory distress. There had been no further episodes of arrhythmia since day 4. His blood pressure was 136/86 mm Hg, heart rate 88 beats per minute and regular, respiratory rate 18 breaths per minute, and oral temperature 37.1°C. His oxygen saturation was 92% on room air.

Before discharge, he was encouraged to quit smoking. He was offered behavioral counseling and medication therapy, but he only said that he would think about it. He was discharged on oral cefixime for 4 more days and was instructed to switch to a long-acting bronchodilator along with his other home medications and to return in 1 week to have his thyroid hormones checked.

One week later, his laboratory results were:

  • TSH 11.2 mU/L (reference range 0.5–5.0)
  • Free T4 1.2 ng/dL (0.9–2.4)
  • Total T3 92 ng/dL (80–180).

Clinically, the patient was euthyroid, and examination of his thyroid was unremarkable.

4. Based on these last test results, which statement is correct?

  • Levothyroxine therapy should be started
  • His serum TSH elevation is most likely transient
  • Thyroid ultrasonography is strongly indicated
  • A radioactive iodine uptake study should be performed
  • Measurement of thyroid-stimulating immunoglobulins is indicated

During recovery from nonthyroidal illness, some patients may have elevated serum TSH levels that are usually transient and modest (< 20 mU/L).48 Normalization of the thyroid function tests including serum TSH may take several weeks49 or months.50 However, a systematic review found that the likelihood of permanent primary hypothyroidism is high in patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness.51

Ultrasonography is useful for evaluating patients with thyroid nodules or goiter but is of little benefit for patients like ours, in whom the thyroid is normal on examination.

Similarly, a radioactive iodine uptake study is not indicated, as it is principally used to help differentiate between types of thyrotoxicosis. (Radioactive iodine is also used to treat differentiated thyroid cancer.)

Thyroid-stimulating immunoglobins are TSH receptor-stimulating antibodies that cause Graves disease. Nevertheless, measuring them is not routinely indicated for its diagnosis. However, their measurement is of significant help in the diagnosis of Graves disease if a radioactive iodine uptake study cannot be performed (as in pregnancy) and in atypical presentations such as euthyroid Graves ophthalmopathy.52 Other indications for thyroid-stimulating immunoglobin measurement are beyond the scope of the article. Our patient’s test results are not consistent with hyperthyroidism, so measuring thyroid-stimulating immunoglobins is not indicated.

CASE CONCLUSION: BETTER, BUT STILL SMOKING

The patient missed his 1-month clinic follow-up, but he visited the clinic for follow-up 3 months later. He was feeling well with no complaints. Test results including serum TSH, free T4, and total T3 were within normal ranges. His COPD was under control, with an FEV1 88% of predicted.

He was again encouraged to quit smoking and was offered drug therapy and behavioral counseling, but he declined. In addition, he was instructed to adhere to his annual influenza vaccination.

KEY POINTS

  • In patients with acute illness, it is recommended that thyroid function not be assessed unless there is a strong indication.
  • If thyroid function assessment is indicated for critically ill patients, serum TSH and free T4 concentrations should be measured. Some clinicians also measure serum total T3 level.
  • Thyroid function testing in critically ill patients usually shows low serum total T3, normal or low serum TSH, and normal or low serum free T4.
  • Many drugs can alter thyroid hormone levels.
  • Thyroid hormone therapy is not recommended for critically ill patients with low T3, low T4, or both.
  • During recovery from nonthyroidal illness, some patients may have mild elevation in serum TSH levels (< 20 mU/L).
  • Thyroid hormone levels may take several weeks or months to return to normal after the acute illness.
  • Patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness are more likely to have permanent primary hypothyroidism.

A 66-year-old man presented to the emergency department with increasing shortness of breath and productive cough, which had begun 5 days earlier. Three years previously, he had been diagnosed with chronic obstructive pulmonary disease (COPD).

One week before the current presentation, he developed a sore throat, rhinorrhea, and nasal congestion, and the shortness of breath had started 2 days after that. Although he could speak in sentences, he was breathless even at rest. His dyspnea was associated with noisy breathing and cough productive of yellowish sputum; there was no hemoptysis. He reported fever, but he had no chills, night sweats, chest pain, or paroxysmal nocturnal dyspnea. The review of other systems was unremarkable.

His COPD was known to be mild, in Global Initiative for Chronic Obstructive Lung Disease (GOLD) grade 1, group A. His postbronchodilator ratio of forced expiratory volume in 1 second (FEV1) to forced vital capacity (FVC) was less than 0.70, and his FEV1 was 84% of predicted. Apart from mild intermittent cough with white sputum, his COPD had been under good control with inhaled ipratropium 4 times daily and inhaled albuterol as needed. He said he did not have shortness of breath except when hurrying on level ground or walking up a slight hill (Modified Medical Research Council dyspnea scale grade 1; COPD Assessment Test score < 10). In the last 3 years, he had 2 exacerbations of COPD, 1 year apart, both requiring oral prednisone and antibiotic therapy.

Other relevant history included hypertension and dyslipidemia of 15-year duration, for which he was taking candesartan 16 mg twice daily and atorvastatin 20 mg daily. He was compliant with his medications.

Though he usually received an influenza vaccine every year, he did not get it the previous year. Also, 3 years previously, he received the 23-valent pneumococcal polysaccharide vaccine (PPSV23), and the year before that he received the pneumococcal conjugate vaccine (PCV13). In addition, he was immunized against herpes zoster and tetanus.

The patient had smoked 1 pack per day for the past 38 years. His primary care physician had advised him many times to quit smoking. He had enrolled in a smoking cessation program 2 years previously, in which he received varenicline in addition to behavioral counseling in the form of motivational interviewing and a telephone quit-line. Nevertheless, he continued to smoke.

He was a retired engineer. He did not drink alcohol or use illicit drugs.

PHYSICAL EXAMINATION

On physical examination, the patient was sitting up in bed, leaning forward. He was alert and oriented but was breathing rapidly and looked sick. He had no cyanosis, clubbing, pallor, or jaundice. His blood pressure was 145/90 mm Hg, heart rate 110 beats per minute and regular, respiratory rate 29 breaths per minute, and oral temperature 38.1°C (100.6°F). His oxygen saturation was 88% while breathing room air. His body mass index was 27.1 kg/m2.

His throat was mildly congested. His neck veins were flat, and there were no carotid bruits. His thyroid examination was normal, without goiter, nodules, or tenderness.

Intercostal retractions were noted around the anterolateral costal margins. He had no chest wall deformities. Chest expansion was reduced bilaterally. There was hyperresonance bilaterally. Expiratory wheezes were heard over both lungs, without crackles.

His heart had no murmurs or added sounds. There was no lower-limb edema or swelling. The rest of his physical examination was unremarkable.

alhalaseh_thyroidfunctiontests_t1.jpg
Chest radiography showed hyperinflation without infiltrates. Electrocardiography showed normal sinus rhythm, with a peaked P wave (P pulmonale) and evidence of right ventricular hypertrophy, but no ischemic changes.

Results of initial laboratory testing are shown in Table 1.

Assessment: A 66-year-old man with GOLD grade 1, group A COPD, presenting with a severe exacerbation, most likely due to viral bronchitis.

 

 

INITIAL MANAGEMENT

The patient was given oxygen 28% by Venturi mask, and his oxygen saturation went up to 90%. He was started on nebulized albuterol 2.5 mg with ipratropium bromide 500 µg every 4 hours, prednisone 40 mg orally daily for 5 days, and ceftriaxone 1 g intravenously every 24 hours. The first dose of each medication was given in the emergency department.

The patient was then admitted to a progressive care unit, where he was placed on noninvasive positive pressure ventilation, continuous cardiac monitoring, and pulse oximetry. He was started on enoxaparin 40 mg subcutaneously daily to prevent venous thromboembolism, and the oral medications he had been taking at home were continued. Because he was receiving a glucocorticoid, his blood glucose was monitored in the fasting state, 2 hours after each meal, and as needed.

Two hours after he started noninvasive positive pressure ventilation, his arterial blood gases were remeasured and showed the following results:

  • pH 7.35
  • Partial pressure of carbon dioxide (Paco2) 52 mm Hg
  • Bicarbonate 28 mmol/L
  • Partial pressure of oxygen (Pao2) 60 mm Hg
  • Oxygen saturation 90%.

HOSPITAL COURSE

On hospital day 3, his dyspnea had slightly improved. His respiratory rate was 26 to 28 breaths per minute. His oxygen saturation remained between 90% and 92%.

At 10:21 pm, his cardiac monitor showed an episode of focal atrial tachycardia at a rate of 129 beats per minute that lasted for 3 minutes and 21 seconds, terminating spontaneously. He denied any change in his clinical condition during the episode, with no chest pain, palpitation, or change in dyspnea. There was no change in his vital signs. He had another similar asymptomatic episode lasting 4 minutes and 9 seconds at 6:30 am of hospital day 4.

Because of these episodes, the attending physician ordered thyroid function tests.

THYROID FUNCTION TESTING

1. Which thyroid function test is most likely to be helpful in the assessment of this patient’s thyroid status?

  • Serum thyroid-stimulating hormone (TSH) alone
  • Serum TSH and total thyroxine (T4)
  • Serum TSH and total triiodothyronine (T3)
  • Serum TSH and free T4
  • Serum TSH and free T3

There are several tests to assess thyroid function: the serum TSH, total T4, free T4, total T3, and free T3 concentrations.1

In normal physiology, TSH from the pituitary stimulates the thyroid gland to produce and secrete T4 and T3, which in turn inhibit TSH secretion through negative feedback. A negative log-linear relation exists between serum free T4 and TSH levels.2 Thus, the serum free T4 level can remain within the normal reference range even if the TSH level is high or low. 

TSH assays can have different detection limits. A third-generation TSH assay with a detection limit of 0.01 mU/L is recommended for use in clinical practice.3

TSH testing alone. Given its superior sensitivity and specificity, serum TSH measurement is considered the best single test for assessing thyroid function in most cases.4 Nevertheless, measurement of the serum TSH level alone could be misleading in several situations, eg, hypothalamic or pituitary disorders, recent treatment of thyrotoxicosis, impaired sensitivity to thyroid hormone, and acute nonthyroidal illness.4

alhalaseh_thyroidfunctiontests_t2.jpg
Because our patient is acutely ill, measuring his serum TSH alone is not the most appropriate test of his thyroid function. Euthyroid patients who present with acute illness usually have different patterns of abnormal thyroid function test results, depending on the severity of their illness, its stage, the drugs they are receiving, and other factors. Thyroid function test abnormalities in those patients are shown in Table 2.5–7

Free vs total T4 and T3 levels

Serum total T4 includes a fraction that is bound, mainly to thyroxin-binding globulin, and a very small unbound (free) fraction. The same applies to T3. Only free thyroid hormones represent the “active” fraction available for interaction with their protein receptors in the nucleus.8 Patients with conditions that can affect the thyroid-binding protein concentrations usually have altered serum total T4 and T3 levels, whereas their free hormone concentrations remain normal. Accordingly, measurement of free hormone levels, especially free T4, is usually recommended.

Although equilibrium dialysis is the method most likely to provide an accurate serum free T4 measurement, it is not commonly used because of its limited availability and high cost. Thus, most commercial laboratories use “direct” free T4 measurement or, to a lesser degree, the free T4 index.9 However, none of the currently available free T4 tests actually measure free T4 directly; rather, they estimate it.10

Commercial laboratories can provide a direct free T3 estimate, but it may be less reliable than total T3. If serum T3 measurement is indicated, serum total T3 is usually measured. However, total T3 measurement is rarely indicated for patients with hypothyroidism because it usually remains within the normal reference range.11 Nevertheless, serum total T3 measurement could be useful in patients with T3 toxicosis and in those who are acutely ill.

Accordingly, in acutely ill hospitalized patients like ours, measuring serum TSH using a third-generation assay and free T4 is essential to assess thyroid function. Many clinicians also measure serum total T3.

 

 

CASE CONTINUED: LOW TSH, LOW-NORMAL FREE T4, LOW TOTAL T3

The attending physician ordered serum TSH, free T4, and total T3 measurements, which yielded the following:

  • TSH 0.1 mU/L (0.5–5.0)
  • Total T3 55 ng/dL (80–180)
  • Free T4 0.9 ng/dL (0.9–2.4).

2. Which best explains this patient’s abnormal thyroid test results?

  • His acute illness
  • Central hypothyroidism due to pituitary infarction
  • His albuterol therapy
  • Subclinical thyrotoxicosis
  • Hashimoto thyroiditis

Since euthyroid patients with an acute illness may have abnormal thyroid test results (Table 2),5–7 thyroid function testing is not recommended unless there is a strong indication for it, such as new-onset atrial fibrillation, atrial flutter, or focal atrial tachycardia.1 In such patients, it is important to know whether the test abnormalities represent true thyroid disorder or are the result of a nonthyroidal illness.

alhalaseh_thyroidfunctiontests_f1.jpg
%3Cp%3EFigure%201.%20Peripheral%20conversion%20of%20thyroxine%20(T%3Csub%3E4%3C%2Fsub%3E)%20to%20triiodothyronine%20(T%3Csub%3E3%3C%2Fsub%3E)%2C%20reverse%20T%3Csub%3E3%3C%2Fsub%3E%2C%20and%20diiodothyronine%20(T%3Csub%3E2%3C%2Fsub%3E)%20by%20deiodinase%20types%201%2C%202%2C%20and%203%20(D1%2C%20D2%2C%20D3)%20in%20healthy%20people%20and%20in%20patients%20with%20nonthyroidal%20illness.%3C%2Fp%3E
In healthy people, T4 is converted to T3 (the principal active hormone) by type 1 deiodinase (D1) mainly in the liver and kidneys, whereas this reaction is catalyzed by type 2 deiodinase (D2) in the hypothalamus and pituitary. Type 3 deiodinase (D3) converts T4 to reverse T3, a biologically inactive molecule.12 D1 also mediates conversion of reverse T3 to diiodothyronine (T2) (Figure 1).

alhalaseh_thyroidfunctiontests_t3.jpg
Several conditions and drugs can decrease D1 activity, resulting in low serum T3 concentrations (Table 3). In patients with nonthyroidal illness, decreased D1 activity can be observed as early as the first 24 hours after the onset of the illness and is attributed to increased inflammatory cytokines, free fatty acids, increased endogenous cortisol secretion, and use of certain drugs.13,14 In addition, the reduced D1 activity can decrease the conversion of reverse T3 to T2, resulting in elevated serum reverse T3. Increased D3 activity during an acute illness is another mechanism for elevated serum reverse T3 concentration.15

Thyroid function testing in patients with nonthyroidal illness usually shows low serum total T3, normal or low serum TSH, and normal, low, or high serum free T4. However, transient mild serum TSH elevation can be seen in some patients during the recovery period.16 These abnormalities with their mechanisms are shown in Table 2.5–7 In several commercial kits, serum direct free T4 can be falsely decreased or increased.8

THE DIFFERENTIAL DIAGNOSIS

Our patient had low serum TSH, low-normal serum direct free T4, and low serum total T3. This profile could be caused by a nonthyroidal illness, “true” central hypothyroidism, or his glucocorticoid treatment. The reason we use the term “true” in this setting is that some experts suggest that the thyroid function test abnormalities in patients with acute nonthyroidal illness represent a transient central hypothyroidism.17 The clinical presentation is key in differentiating true central hypothyroidism from nonthyroidal illness.

In addition, measuring serum cortisol may help to differentiate between the 2 states, as it would be elevated in patients with nonthyroidal illness as part of a stress response but low in patients with true central hypothyroidism, since it is usually part of combined pituitary hormone deficiency.18 Of note, some critically ill patients have low serum cortisol because of transient central adrenal insufficiency.19,20

The serum concentration of reverse T3 has been suggested as a way to differentiate between hypothyroidism (low) and nonthyroidal illness (high); however, further studies showed that it does not reliably differentiate between the conditions.21

GLUCOCORTICOIDS AND THYROID FUNCTION TESTS

By inhibiting D1, glucocorticoids can decrease peripheral conversion of T4 to T3 and thus decrease serum total T3. This effect depends on the type and dose of the glucocorticoid and the duration of therapy.

In one study,22 there was a significant reduction in serum total T3 concentration 24 hours after a single oral dose of dexamethasone 12 mg in normal participants. This effect lasted 48 hours, after which serum total T3 returned to its pretreatment level.

In another study,23 a daily oral dose of betamethasone 1.5 mg for 5 days did not significantly reduce the serum total T3 in healthy volunteers, but a daily dose of 3 mg did. This effect was more pronounced at a daily dose of 4.5 mg, whereas a dose of 6.0 mg had no further effect.

Long-term glucocorticoid therapy also decreases serum total T4 and total T3 by lowering serum thyroid-binding globulin.24

Finally, glucocorticoids can decrease TSH secretion by directly inhibiting thyrotropin-releasing hormone.25,26 However, chronic hypercortisolism, whether endogenous or exogenous, does not cause clinically central hypothyroidism, possibly because of the negative feedback mechanism of low thyroid hormones on the pituitary and the hypothalamus.27

Other drugs including dopamine, dopamine agonists, dobutamine, and somatostatin analogues can suppress serum TSH. As with glucocorticoids, these drugs do not cause clinically evident central hypothyroidism.28 Bexarotene, a retinoid X receptor ligand used in the treatment of cutaneous T-cell lymphoma, has been reported to cause clinically evident central hypothyroidism by suppressing TSH and increasing T4 clearance.29

 

 

BETA-BLOCKERS, BETA-AGONISTS AND THYROID FUNCTION

While there is general agreement that beta-adrenergic antagonists (beta-blockers) do not affect the serum TSH concentration, conflicting data have been reported concerning their effect on other thyroid function tests. This may be due to several factors, including dose, duration of therapy, the patient’s thyroid status, and differences in laboratory methodology.30

In studies of propranolol, serum total T4 concentrations did not change or were increased with daily doses of 160 mg or more in both euthyroid participants and hyperthyroid patients31–33; serum total T3 concentrations did not change or were decreased with 40 mg or more daily34; and serum reverse T3 concentrations were increased with daily doses of 80 mg or more.31 It is most likely that propranolol exerts these changes by inhibiting D1 activity in peripheral tissues.

Furthermore, a significant decrease in serum total T3 concentrations was observed in hyperthyroid patients treated with atenolol 100 mg daily, metoprolol 100 mg daily, and alprenolol 100 mg daily, but not with sotalol 80 mg daily or nadolol (up to 240 mg daily).35,36

On the other hand, beta-adrenergic agonists have not been reported to cause significant changes in thyroid function tests.37

SUBCLINICAL THYROTOXICOSIS OR HASHIMOTO THYROIDITIS?

Our patient’s thyroid function test results are more likely due to his nonthyroidal illness and glucocorticoid therapy, as there is no clinical evidence to point to a hypothalamic-pituitary disorder accounting for true central hypothyroidism.

The other options mentioned in question 2 are unlikely to explain our patient’s thyroid function test results.

Subclinical thyrotoxicosis is characterized by suppressed serum TSH, but both serum free T4 and total T3 remain within the normal reference ranges. In addition, the serum TSH level may help to differentiate between thyrotoxicosis and nonthyroidal illness. In the former, serum TSH is usually suppressed (< 0.01 mU/L), whereas in the latter it is usually low but detectable (0.05– 0.3 mU/L).38,39

Hashimoto thyroiditis is a chronic autoimmune thyroid disease characterized by diffuse lymphocytic infiltration of the thyroid gland. Almost all patients with Hashimoto thyroiditis have elevated levels of antibodies to thyroid peroxidase or thyroglobulin.40 Clinically, patients with Hashimoto thyroiditis can either be hypothyroid or have normal thyroid function, which is not the case in our patient.

CASE CONTINUED

An endocrinologist, consulted for a second opinion, agreed that the patient’s thyroid function test results were most likely due to his nonthyroidal illness and glucocorticoid therapy.

3. In view of the endocrinologist’s opinion, which should be the next step in the management of the patient’s thyroid condition?

  • Start levothyroxine (T4) therapy
  • Start liothyronine (T3) therapy
  • Start N-acetylcysteine therapy
  • Start thyrotropin-releasing hormone therapy
  • Remeasure thyroid hormones after full recovery from his acute illness

It is not clear whether the changes in thyroid hormone levels during an acute illness are a pathologic alteration for which thyroid hormone therapy may be beneficial, or a physiologic adaptation for which such therapy would not be indicated.41

However, current data argue against thyroid hormone therapy using T4 or T3 for patients with nonthyroidal illness syndrome (also called euthyroid sick syndrome).42 Indeed, several randomized controlled trials showed that thyroid hormone therapy is not beneficial in such patients and may be detrimental.41,43

Therapies other than thyroid hormone have been investigated to ameliorate thyroid hormone abnormalities in patients with nonthyroidal illness. These include N-acetylcysteine, thyrotropin-releasing hormone therapy, and nutritional support.

Some studies showed that giving N-acetyl­cysteine, an antioxidant, increased serum T3 and decreased serum reverse T3 concentrations in patients with acute myocardial infarction.44 Nevertheless, the mortality rate and length of hospitalization were not affected. Further studies are needed to know whether N-acetylcysteine therapy is beneficial for such patients.

Similarly, a study using a thyrotropin-releasing hormone analogue along with growth hormone-releasing peptide 2 showed an increase in serum TSH, T4, and T3 levels in critically ill patients.45 The benefit of this therapy has yet to be determined. On the other hand, early nutritional support was reported to prevent thyroid hormonal changes in patients postoperatively.46

Measuring thyroid hormone levels after full recovery is the most appropriate next step in our patient, as the changes in thyroid hormone concentrations subside as the acute illness resolves.47

 

 

CASE CONTINUED

The patient continued to improve. On hospital day 6, he was feeling better but still had mild respiratory distress. There had been no further episodes of arrhythmia since day 4. His blood pressure was 136/86 mm Hg, heart rate 88 beats per minute and regular, respiratory rate 18 breaths per minute, and oral temperature 37.1°C. His oxygen saturation was 92% on room air.

Before discharge, he was encouraged to quit smoking. He was offered behavioral counseling and medication therapy, but he only said that he would think about it. He was discharged on oral cefixime for 4 more days and was instructed to switch to a long-acting bronchodilator along with his other home medications and to return in 1 week to have his thyroid hormones checked.

One week later, his laboratory results were:

  • TSH 11.2 mU/L (reference range 0.5–5.0)
  • Free T4 1.2 ng/dL (0.9–2.4)
  • Total T3 92 ng/dL (80–180).

Clinically, the patient was euthyroid, and examination of his thyroid was unremarkable.

4. Based on these last test results, which statement is correct?

  • Levothyroxine therapy should be started
  • His serum TSH elevation is most likely transient
  • Thyroid ultrasonography is strongly indicated
  • A radioactive iodine uptake study should be performed
  • Measurement of thyroid-stimulating immunoglobulins is indicated

During recovery from nonthyroidal illness, some patients may have elevated serum TSH levels that are usually transient and modest (< 20 mU/L).48 Normalization of the thyroid function tests including serum TSH may take several weeks49 or months.50 However, a systematic review found that the likelihood of permanent primary hypothyroidism is high in patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness.51

Ultrasonography is useful for evaluating patients with thyroid nodules or goiter but is of little benefit for patients like ours, in whom the thyroid is normal on examination.

Similarly, a radioactive iodine uptake study is not indicated, as it is principally used to help differentiate between types of thyrotoxicosis. (Radioactive iodine is also used to treat differentiated thyroid cancer.)

Thyroid-stimulating immunoglobins are TSH receptor-stimulating antibodies that cause Graves disease. Nevertheless, measuring them is not routinely indicated for its diagnosis. However, their measurement is of significant help in the diagnosis of Graves disease if a radioactive iodine uptake study cannot be performed (as in pregnancy) and in atypical presentations such as euthyroid Graves ophthalmopathy.52 Other indications for thyroid-stimulating immunoglobin measurement are beyond the scope of the article. Our patient’s test results are not consistent with hyperthyroidism, so measuring thyroid-stimulating immunoglobins is not indicated.

CASE CONCLUSION: BETTER, BUT STILL SMOKING

The patient missed his 1-month clinic follow-up, but he visited the clinic for follow-up 3 months later. He was feeling well with no complaints. Test results including serum TSH, free T4, and total T3 were within normal ranges. His COPD was under control, with an FEV1 88% of predicted.

He was again encouraged to quit smoking and was offered drug therapy and behavioral counseling, but he declined. In addition, he was instructed to adhere to his annual influenza vaccination.

KEY POINTS

  • In patients with acute illness, it is recommended that thyroid function not be assessed unless there is a strong indication.
  • If thyroid function assessment is indicated for critically ill patients, serum TSH and free T4 concentrations should be measured. Some clinicians also measure serum total T3 level.
  • Thyroid function testing in critically ill patients usually shows low serum total T3, normal or low serum TSH, and normal or low serum free T4.
  • Many drugs can alter thyroid hormone levels.
  • Thyroid hormone therapy is not recommended for critically ill patients with low T3, low T4, or both.
  • During recovery from nonthyroidal illness, some patients may have mild elevation in serum TSH levels (< 20 mU/L).
  • Thyroid hormone levels may take several weeks or months to return to normal after the acute illness.
  • Patients with serum TSH levels higher than 20 mU/L during the recovery phase of their nonthyroidal illness are more likely to have permanent primary hypothyroidism.
References
  1. Lamb EJ, Martin J. Thyroid function tests: often justified in the acutely ill. Ann Clin Biochem 2000; 37(pt 2):158–164. doi:10.1258/0004563001899159
  2. Spencer CA, LoPresti JS, Patel A, et al. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990; 70(2):453–460. doi:10.1210/jcem-70-2-453
  3. Ross DS, Ardisson LJ, Meskell MJ. Measurement of thyrotropin in clinical and subclinical hyperthyroidism using a new chemiluminescent assay. J Clin Endocrinol Metab 1989; 69(3):684–688. doi:10.1210/jcem-69-3-684
  4. Koulouri O, Moran C, Halsall D, Chatterjee K, Gurnell M. Pitfalls in the measurement and interpretation of thyroid function tests. Best Pract Res Clin Endocrinol Metab 2013; 27(6):745–762. doi:10.1016/j.beem.2013.10.003
  5. Lechan RM, Fekete C. Role of thyroid hormone deiodination in the hypothalamus. Thyroid 2005; 15(8):883–897. doi:10.1089/thy.2005.15.883
  6. Chopra IJ, Hershman JM, Pardridge WM, Nicoloff JT. Thyroid function in nonthyroidal ilnesses. Ann Intern Med 1983; 98(6):946–957. doi:10.7326/0003-4819-98-6-946
  7. Chopra IJ, Solomon DH, Hepner HW, Mortenstein AA. Misleadingly low free thyroxine index and usefulness of reverse triiodothyronine measurement in nonthyroidal illnesses. Ann Intern Med 1979; 90(6):905–912. doi:10.7326/0003-4819-90-6-905
  8. Pontecorvi A, Robbins J. The plasma membrane and thyroid hormone entry into cells. Trends Endocrinol Metab 1989; 1(2):90–94. pmid:18411097
  9. Hennemann G, Krenning EP. Pitfalls in the interpretation of thyroid function tests in old age and non-thyroidal illness. Horm Res 1987; 26(1–4):100–104. doi:10.1159/000180688
  10. Baloch Z, Carayon P, Conte-Devolx B, et al; Guidelines Committee, National Academy of Clinical Biochemistry. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid 2003; 13(1):3–126. doi:10.1089/105072503321086962
  11. Lum S, Nicoloff JT, Spencer CA, Kaptein EM. Peripheral tissue mechanism for maintenance of serum triiodothyronine values in a thyroxine-deficient state in man. J Clin Invest 1984; 73(2):570–575. doi:10.1172/JCI111245
  12. Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-pituitary-thyroid axis. Compr Physiol 2016; 6(3):1387–1428. doi:10.1002/cphy.c150027
  13. de Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol 2015; 225(3):R67–R81. doi:10.1530/JOE-15-0133
  14. Chopra IJ, Huang TS, Beredo A, Solomon DH, Teco GN, Mean JF. Evidence for an inhibitor of extrathyroidal conversion of thyroxine to 3, 5, 3'-triiodothyronine in sera of patients with nonthyroidal illnesses. J Clin Endocrinol Metab 1985; 60(4):666–672. doi:10.1210/jcem-60-4-666
  15. Peeters RP, Debaveye Y, Fliers E, Visser TJ. Changes within the thyroid axis during critical illness. Crit Care Clin 2006; 22(1):41–55. doi:10.1016/j.ccc.2005.08.006
  16. Spencer C, Eigen A, Shen D, et al. Specificity of sensitive assays of thyrotropin (TSH) used to screen for thyroid disease in hospitalized patients. Clin Chem 1987; 33(8):1391–1396. pmid:3301067
  17. Adler SM, Wartofsky L. The nonthyroidal illness syndrome. Endocrinol Metab Clin North Am 2007; 36(3):657–672. doi:10.1016/j.ecl.2007.04.007
  18. Persani L. Central hypothyroidism: pathogenic, diagnostic, and therapeutic challenges. J Clin Endocrinol Metab 2012; 97(9):3068–3078. doi:10.1210/jc.2012-1616
  19. Kidess AI, Caplan RH, Reynertson RH, Wickus GG, Goodnough DE. Transient corticotropin deficiency in critical illness. Mayo Clin Proc 1993; 68(5):435–441. doi:10.1016/s0025-6196(12)60188-8
  20. Lamberts SW, Bruining HA, De Jong FH. Corticosteroid therapy in severe illness. N Engl J Med 1997; 337(18):1285–1292. doi:10.1056/NEJM199710303371807
  21. Burmeister LA. Reverse T3 does not reliably differentiate hypothyroid sick syndrome from euthyroid sick syndrome. Thyroid 1995; 5(6):435–441. doi:10.1089/thy.1995.5.435
  22. Duick DS, Warren DW, Nicoloff JT, Otis CL, Croxson MS. Effect of single dose dexamethasone on the concentration of serum triiodothyronine in man. J Clin Endocrinol Metab 1974; 39(6):1151–1154. doi:10.1210/jcem-39-6-1151
  23. Gamstedt A, Järnerot G, Kågedal B. Dose related effects of betamethasone on iodothyronines and thyroid hormone-binding proteins in serum. Acta Endocrinol (Copenh) 1981; 96(4):484–490. doi:10.1530/acta.0.0960484
  24. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the “euthyroid sick syndrome.” Endocr Rev 1982; 3(2):164–217. doi:10.1210/edrv-3-2-164
  25. Wilber JF, Utiger RD. The effect of glucocorticoids on thyrotropin secretion. J Clin Invest 1969; 48(11):2096–2103. doi:10.1172/JCI106176
  26. Nicoloff JT, Fisher DA, Appleman MD Jr. The role of glucocorticoids in the regulation of thyroid function in man. J Clin Invest 1970; 49(10):1922–1929. doi:10.1172/JCI106411
  27. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med 1995; 333(25):1688–1694. doi:10.1056/NEJM199512213332507
  28. Haugen BR. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab 2009; 23(6):793–800. doi:10.1016/j.beem.2009.08.003
  29. Sherman SI, Gopal J, Haugen BR, et al. Central hypothyroidism associated with retinoid X receptor–selective ligands. N Engl J Med 1999; 340(14):1075–1079. doi:10.1056/NEJM199904083401404
  30. Murchison LE, How J, Bewsher PD. Comparison of propranolol and metoprolol in the management of hyperthyroidism. Br J Clin Pharmacol 1979; 8(6):581–587. doi:10.1111/j.1365-2125.1979.tb01048.x
  31. Faber J, Friis T, Kirkegaard C, et al. Serum T4, T3 and reverse T3 during treatment with propranolol in hyperthyroidism, L-T4 treated myxedema and in normal man. Horm Metab Res 1979; 11(1):34–36. doi:10.1055/s-0028-1092678
  32. Kristensen BO, Weeke J. Propranolol-induced increments in total and free serum thyroxine in patients with essential hypertension. Clin Pharmacol Ther 1977; 22(6):864–867. doi:10.1002/cpt1977226864
  33. Murchison LE, Bewsher PD, Chesters MI, Ferrier WR. Comparison of propranolol and practolol in the management of hyperthyroidism. Br J Clin Pharmacol 1976; 3(2):273–277. doi:10.1111/j.1365-2125.1976.tb00603.x
  34. Lotti G, Delitala G, Devilla L, Alagna S, Masala A. Reduction of plasma triiodothyronine (T3) induced by propranolol. Clin Endocrinol 1977; 6(6):405–410. doi:10.1111/j.1365-2265.1977.tb03322.x
  35. Perrild H, Hansen JM, Skovsted L, Christensen LK. Different effects of propranolol, alprenolol, sotalol, atenolol and metoprolol on serum T3 and serum rT3 in hyperthyroidism. Clin Endocrinol (Oxf) 1983; 18(2):139–142. pmid:6133659
  36. Reeves RA, From GL, Paul W, Leenen FH. Nadolol, propranolol, and thyroid hormones: evidence for a membrane-stabilizing action of propranolol. Clin Pharmacol Ther 1985; 37(2):157–161. doi:10.1038/clpt.1985.28
  37. Walker N, Jung RT, Jennings G, James WP. The effect of a beta-receptor agonist (salbutamol) on peripheral thyroid metabolism in euthyroid subjects. Horm Metab Res 1981; 13(10):590–591. doi:10.1055/s-2007-1019346
  38. Melmed S, Geola FL, Reed AW, Pekary AE, Park J, Hershman JM. A comparison of methods for assessing thyroid function in nonthyroidal illness. J Clin Endocrinol Metab 1982; 54(2):300–306. doi:10.1210/jcem-54-2-300
  39. Docter R, Krenning E, De Jong M, Hennemann G. The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism. Clin Endocrinol (Oxf) 1993; 39(5):499–518. pmid:8252737
  40. Mariotti S, Caturegli P, Piccolo P, Barbesino G, Pinchera A. Antithyroid peroxidase autoantibodies in thyroid diseases. J Clin Endocrinol Metab 1990; 71(3):661–669. doi:10.1210/jcem-71-3-661
  41. De Groot LJ. Non-thyroidal illness syndrome is a manifestation of hypothalamic-pituitary dysfunction, and in view of current evidence, should be treated with appropriate replacement therapies. Crit Care Clin 2006; 22(1):57–86. doi:10.1016/j.ccc.2005.10.001
  42. Jonklaas J, Bianco AC, Bauer AJ, et al; American Thyroid Association Task Force on Thyroid Hormone Replacement. Guidelines for the treatment of hypothyroidism: prepared by the American Thyroid Association Task Force on Thyroid Hormone Replacement. Thyroid 2014; 24(12):1670–1751. doi:10.1089/thy.2014.0028
  43. Kaptein EM, Beale E, Chan LS. Thyroid hormone therapy for obesity and nonthyroidal illnesses: a systematic review. J Clin Endocrinol Metab 2009; 94(10):3663–3675. doi:10.1210/jc.2009-0899
  44. Vidart J, Wajner SM, Leite RS, et al. N-acetylcysteine administration prevents nonthyroidal illness syndrome in patients with acute myocardial infarction: a randomized clinical trial. J Clin Endocrinol Metab 2014; 99(12):4537–4545. doi:10.1210/jc.2014-2192
  45. Van den Berghe G, Wouters P, Weekers F, et al. Reactivation of pituitary hormone release and metabolic improvement by infusion of growth hormone-releasing peptide and thyrotropin-releasing hormone in patients with protracted critical illness. J Clin Endocrinol Metab 1999; 84(4):1311–1323. doi:10.1210/jcem.84.4.5636
  46. Langouche L, Vander Perre S, Marques M, et al. Impact of early nutrient restriction during critical illness on the nonthyroidal illness syndrome and its relation with outcome: a randomized, controlled clinical study. J Clin Endocrinol Metab 2013; 98(3):1006–1013. doi:10.1210/jc.2012-2809
  47. Economidou F, Douka E, Tzanela M, Nanas S, Kotanidou A. Thyroid function during critical illness. Hormones (Athens) 2011; 10(2):117–124. doi:10.14310/horm.2002.1301
  48. Hamblin PS, Dyer SA, Mohr VS, et al. Relationship between thyrotropin and thyroxine changes during recovery from severe hypothyroxinemia of critical illness. J Clin Endocrinol Metab 1986; 62(4):717–722. doi:10.1210/jcem-62-4-717
  49. Iglesias P, Diez JJ. Thyroid dysfunction and kidney disease. Eur J Endocrinol 2009; 160(4):503–515. doi:10.1530/EJE-08-0837
  50. Spencer CA. Clinical utility and cost-effectiveness of sensitive thyrotropin assays in ambulatory and hospitalized patients. Mayo Clin Proc 1988; 63(12):1214–1222. doi:10.1016/s0025-6196(12)65408-1
  51. Attia J, Margetts P, Guyatt G. Diagnosis of thyroid disease in hospitalized patients: a systematic review. Arch Intern Med 1999; 159(7):658–665. pmid:10218744
  52. Barbesino G, Tomer Y. Clinical review: clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab 2013; 98(6):2247–2255. doi:10.1210/jc.2012-4309
References
  1. Lamb EJ, Martin J. Thyroid function tests: often justified in the acutely ill. Ann Clin Biochem 2000; 37(pt 2):158–164. doi:10.1258/0004563001899159
  2. Spencer CA, LoPresti JS, Patel A, et al. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. J Clin Endocrinol Metab 1990; 70(2):453–460. doi:10.1210/jcem-70-2-453
  3. Ross DS, Ardisson LJ, Meskell MJ. Measurement of thyrotropin in clinical and subclinical hyperthyroidism using a new chemiluminescent assay. J Clin Endocrinol Metab 1989; 69(3):684–688. doi:10.1210/jcem-69-3-684
  4. Koulouri O, Moran C, Halsall D, Chatterjee K, Gurnell M. Pitfalls in the measurement and interpretation of thyroid function tests. Best Pract Res Clin Endocrinol Metab 2013; 27(6):745–762. doi:10.1016/j.beem.2013.10.003
  5. Lechan RM, Fekete C. Role of thyroid hormone deiodination in the hypothalamus. Thyroid 2005; 15(8):883–897. doi:10.1089/thy.2005.15.883
  6. Chopra IJ, Hershman JM, Pardridge WM, Nicoloff JT. Thyroid function in nonthyroidal ilnesses. Ann Intern Med 1983; 98(6):946–957. doi:10.7326/0003-4819-98-6-946
  7. Chopra IJ, Solomon DH, Hepner HW, Mortenstein AA. Misleadingly low free thyroxine index and usefulness of reverse triiodothyronine measurement in nonthyroidal illnesses. Ann Intern Med 1979; 90(6):905–912. doi:10.7326/0003-4819-90-6-905
  8. Pontecorvi A, Robbins J. The plasma membrane and thyroid hormone entry into cells. Trends Endocrinol Metab 1989; 1(2):90–94. pmid:18411097
  9. Hennemann G, Krenning EP. Pitfalls in the interpretation of thyroid function tests in old age and non-thyroidal illness. Horm Res 1987; 26(1–4):100–104. doi:10.1159/000180688
  10. Baloch Z, Carayon P, Conte-Devolx B, et al; Guidelines Committee, National Academy of Clinical Biochemistry. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid 2003; 13(1):3–126. doi:10.1089/105072503321086962
  11. Lum S, Nicoloff JT, Spencer CA, Kaptein EM. Peripheral tissue mechanism for maintenance of serum triiodothyronine values in a thyroxine-deficient state in man. J Clin Invest 1984; 73(2):570–575. doi:10.1172/JCI111245
  12. Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-pituitary-thyroid axis. Compr Physiol 2016; 6(3):1387–1428. doi:10.1002/cphy.c150027
  13. de Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol 2015; 225(3):R67–R81. doi:10.1530/JOE-15-0133
  14. Chopra IJ, Huang TS, Beredo A, Solomon DH, Teco GN, Mean JF. Evidence for an inhibitor of extrathyroidal conversion of thyroxine to 3, 5, 3'-triiodothyronine in sera of patients with nonthyroidal illnesses. J Clin Endocrinol Metab 1985; 60(4):666–672. doi:10.1210/jcem-60-4-666
  15. Peeters RP, Debaveye Y, Fliers E, Visser TJ. Changes within the thyroid axis during critical illness. Crit Care Clin 2006; 22(1):41–55. doi:10.1016/j.ccc.2005.08.006
  16. Spencer C, Eigen A, Shen D, et al. Specificity of sensitive assays of thyrotropin (TSH) used to screen for thyroid disease in hospitalized patients. Clin Chem 1987; 33(8):1391–1396. pmid:3301067
  17. Adler SM, Wartofsky L. The nonthyroidal illness syndrome. Endocrinol Metab Clin North Am 2007; 36(3):657–672. doi:10.1016/j.ecl.2007.04.007
  18. Persani L. Central hypothyroidism: pathogenic, diagnostic, and therapeutic challenges. J Clin Endocrinol Metab 2012; 97(9):3068–3078. doi:10.1210/jc.2012-1616
  19. Kidess AI, Caplan RH, Reynertson RH, Wickus GG, Goodnough DE. Transient corticotropin deficiency in critical illness. Mayo Clin Proc 1993; 68(5):435–441. doi:10.1016/s0025-6196(12)60188-8
  20. Lamberts SW, Bruining HA, De Jong FH. Corticosteroid therapy in severe illness. N Engl J Med 1997; 337(18):1285–1292. doi:10.1056/NEJM199710303371807
  21. Burmeister LA. Reverse T3 does not reliably differentiate hypothyroid sick syndrome from euthyroid sick syndrome. Thyroid 1995; 5(6):435–441. doi:10.1089/thy.1995.5.435
  22. Duick DS, Warren DW, Nicoloff JT, Otis CL, Croxson MS. Effect of single dose dexamethasone on the concentration of serum triiodothyronine in man. J Clin Endocrinol Metab 1974; 39(6):1151–1154. doi:10.1210/jcem-39-6-1151
  23. Gamstedt A, Järnerot G, Kågedal B. Dose related effects of betamethasone on iodothyronines and thyroid hormone-binding proteins in serum. Acta Endocrinol (Copenh) 1981; 96(4):484–490. doi:10.1530/acta.0.0960484
  24. Wartofsky L, Burman KD. Alterations in thyroid function in patients with systemic illness: the “euthyroid sick syndrome.” Endocr Rev 1982; 3(2):164–217. doi:10.1210/edrv-3-2-164
  25. Wilber JF, Utiger RD. The effect of glucocorticoids on thyrotropin secretion. J Clin Invest 1969; 48(11):2096–2103. doi:10.1172/JCI106176
  26. Nicoloff JT, Fisher DA, Appleman MD Jr. The role of glucocorticoids in the regulation of thyroid function in man. J Clin Invest 1970; 49(10):1922–1929. doi:10.1172/JCI106411
  27. Surks MI, Sievert R. Drugs and thyroid function. N Engl J Med 1995; 333(25):1688–1694. doi:10.1056/NEJM199512213332507
  28. Haugen BR. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab 2009; 23(6):793–800. doi:10.1016/j.beem.2009.08.003
  29. Sherman SI, Gopal J, Haugen BR, et al. Central hypothyroidism associated with retinoid X receptor–selective ligands. N Engl J Med 1999; 340(14):1075–1079. doi:10.1056/NEJM199904083401404
  30. Murchison LE, How J, Bewsher PD. Comparison of propranolol and metoprolol in the management of hyperthyroidism. Br J Clin Pharmacol 1979; 8(6):581–587. doi:10.1111/j.1365-2125.1979.tb01048.x
  31. Faber J, Friis T, Kirkegaard C, et al. Serum T4, T3 and reverse T3 during treatment with propranolol in hyperthyroidism, L-T4 treated myxedema and in normal man. Horm Metab Res 1979; 11(1):34–36. doi:10.1055/s-0028-1092678
  32. Kristensen BO, Weeke J. Propranolol-induced increments in total and free serum thyroxine in patients with essential hypertension. Clin Pharmacol Ther 1977; 22(6):864–867. doi:10.1002/cpt1977226864
  33. Murchison LE, Bewsher PD, Chesters MI, Ferrier WR. Comparison of propranolol and practolol in the management of hyperthyroidism. Br J Clin Pharmacol 1976; 3(2):273–277. doi:10.1111/j.1365-2125.1976.tb00603.x
  34. Lotti G, Delitala G, Devilla L, Alagna S, Masala A. Reduction of plasma triiodothyronine (T3) induced by propranolol. Clin Endocrinol 1977; 6(6):405–410. doi:10.1111/j.1365-2265.1977.tb03322.x
  35. Perrild H, Hansen JM, Skovsted L, Christensen LK. Different effects of propranolol, alprenolol, sotalol, atenolol and metoprolol on serum T3 and serum rT3 in hyperthyroidism. Clin Endocrinol (Oxf) 1983; 18(2):139–142. pmid:6133659
  36. Reeves RA, From GL, Paul W, Leenen FH. Nadolol, propranolol, and thyroid hormones: evidence for a membrane-stabilizing action of propranolol. Clin Pharmacol Ther 1985; 37(2):157–161. doi:10.1038/clpt.1985.28
  37. Walker N, Jung RT, Jennings G, James WP. The effect of a beta-receptor agonist (salbutamol) on peripheral thyroid metabolism in euthyroid subjects. Horm Metab Res 1981; 13(10):590–591. doi:10.1055/s-2007-1019346
  38. Melmed S, Geola FL, Reed AW, Pekary AE, Park J, Hershman JM. A comparison of methods for assessing thyroid function in nonthyroidal illness. J Clin Endocrinol Metab 1982; 54(2):300–306. doi:10.1210/jcem-54-2-300
  39. Docter R, Krenning E, De Jong M, Hennemann G. The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism. Clin Endocrinol (Oxf) 1993; 39(5):499–518. pmid:8252737
  40. Mariotti S, Caturegli P, Piccolo P, Barbesino G, Pinchera A. Antithyroid peroxidase autoantibodies in thyroid diseases. J Clin Endocrinol Metab 1990; 71(3):661–669. doi:10.1210/jcem-71-3-661
  41. De Groot LJ. Non-thyroidal illness syndrome is a manifestation of hypothalamic-pituitary dysfunction, and in view of current evidence, should be treated with appropriate replacement therapies. Crit Care Clin 2006; 22(1):57–86. doi:10.1016/j.ccc.2005.10.001
  42. Jonklaas J, Bianco AC, Bauer AJ, et al; American Thyroid Association Task Force on Thyroid Hormone Replacement. Guidelines for the treatment of hypothyroidism: prepared by the American Thyroid Association Task Force on Thyroid Hormone Replacement. Thyroid 2014; 24(12):1670–1751. doi:10.1089/thy.2014.0028
  43. Kaptein EM, Beale E, Chan LS. Thyroid hormone therapy for obesity and nonthyroidal illnesses: a systematic review. J Clin Endocrinol Metab 2009; 94(10):3663–3675. doi:10.1210/jc.2009-0899
  44. Vidart J, Wajner SM, Leite RS, et al. N-acetylcysteine administration prevents nonthyroidal illness syndrome in patients with acute myocardial infarction: a randomized clinical trial. J Clin Endocrinol Metab 2014; 99(12):4537–4545. doi:10.1210/jc.2014-2192
  45. Van den Berghe G, Wouters P, Weekers F, et al. Reactivation of pituitary hormone release and metabolic improvement by infusion of growth hormone-releasing peptide and thyrotropin-releasing hormone in patients with protracted critical illness. J Clin Endocrinol Metab 1999; 84(4):1311–1323. doi:10.1210/jcem.84.4.5636
  46. Langouche L, Vander Perre S, Marques M, et al. Impact of early nutrient restriction during critical illness on the nonthyroidal illness syndrome and its relation with outcome: a randomized, controlled clinical study. J Clin Endocrinol Metab 2013; 98(3):1006–1013. doi:10.1210/jc.2012-2809
  47. Economidou F, Douka E, Tzanela M, Nanas S, Kotanidou A. Thyroid function during critical illness. Hormones (Athens) 2011; 10(2):117–124. doi:10.14310/horm.2002.1301
  48. Hamblin PS, Dyer SA, Mohr VS, et al. Relationship between thyrotropin and thyroxine changes during recovery from severe hypothyroxinemia of critical illness. J Clin Endocrinol Metab 1986; 62(4):717–722. doi:10.1210/jcem-62-4-717
  49. Iglesias P, Diez JJ. Thyroid dysfunction and kidney disease. Eur J Endocrinol 2009; 160(4):503–515. doi:10.1530/EJE-08-0837
  50. Spencer CA. Clinical utility and cost-effectiveness of sensitive thyrotropin assays in ambulatory and hospitalized patients. Mayo Clin Proc 1988; 63(12):1214–1222. doi:10.1016/s0025-6196(12)65408-1
  51. Attia J, Margetts P, Guyatt G. Diagnosis of thyroid disease in hospitalized patients: a systematic review. Arch Intern Med 1999; 159(7):658–665. pmid:10218744
  52. Barbesino G, Tomer Y. Clinical review: clinical utility of TSH receptor antibodies. J Clin Endocrinol Metab 2013; 98(6):2247–2255. doi:10.1210/jc.2012-4309
Issue
Cleveland Clinic Journal of Medicine - 86(10)
Issue
Cleveland Clinic Journal of Medicine - 86(10)
Page Number
666-675
Page Number
666-675
Publications
Publications
Topics
Article Type
Display Headline
A 66-year-old man with abnormal thyroid function tests
Display Headline
A 66-year-old man with abnormal thyroid function tests
Legacy Keywords
thyroid, thyroid function tests, hypothyroid, chronic obstructive pulmonary disease, COPD, thyroid-stimulating hormone, TSH, thyroxine, T4, triiodothyronine, T3, reverse T3, deiodinase, D1, euthyroid sick syndrome, nonthyroidal illness syndrome, Yazan Alhalaseh, Zaid Abdulelah, Ahmad Armouti, Ayman Zayed
Legacy Keywords
thyroid, thyroid function tests, hypothyroid, chronic obstructive pulmonary disease, COPD, thyroid-stimulating hormone, TSH, thyroxine, T4, triiodothyronine, T3, reverse T3, deiodinase, D1, euthyroid sick syndrome, nonthyroidal illness syndrome, Yazan Alhalaseh, Zaid Abdulelah, Ahmad Armouti, Ayman Zayed
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 09/23/2019 - 15:15
Un-Gate On Date
Mon, 09/23/2019 - 15:15
Use ProPublica
CFC Schedule Remove Status
Mon, 09/23/2019 - 15:15
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Obstructive sleep apnea basics

Article Type
Changed
Tue, 09/03/2019 - 08:05
Display Headline
Obstructive sleep apnea basics

DEFINITION

Obstructive sleep apnea (OSA) occurs when there are recurrent episodes of upper airway collapse and obstruction during sleep associated with arousals with or without oxygen desaturations. The oropharynx in the back of the throat collapses during OSA events to cause arousal or oxygen desaturation or both resulting in fragmented sleep.

PREVALENCE

Studies reveal OSA is prevalent. A 2015 study in Switzerland reported 50% of men and 23% of women had at least moderate OSA.1 In 2002, the Sleep Heart Health study found that 24% of men and 9% of women have at least mild OSA.2 In the Wisconsin Sleep Study Cohort, it was reported that 10% of men and 3% of women age 30 to 49 have at least moderate OSA, while 17% of men and 9% of women age 50 to 70 have at least moderate OSA.3 OSA is highly underrecognized and it is estimated that 82% of men and 93% of women in the United States with OSA are undiagnosed.4

SYMPTOMS

vensel_rundo_osabasics_t1.jpg
There are several common sleep and daytime symptoms associated with OSA though patients vary in the number and combination of symptoms reported (Table 1).5 During sleep, snoring is one of the most common symptoms. Common daytime symptoms of OSA include excessive daytime sleepiness or fatigue. Excessive daytime sleepiness is feeling very drowsy or very sleepy at times whereas fatigue is feeling tired, low on energy, and unmotivated. Feeling unrefreshed despite getting the recommended 7 to 9 hours of sleep is also a symptom.

RISK FACTORS

The risk of OSA is influenced by unmodifiable and modifiable factors. Unmodifiable risk factors include male sex, age, and race. Genetic predisposition or a family history of OSA as well as cranial facial anatomy resulting in narrow airways may impart higher risk of OSA. Modifiable risk factors include obesity, medications that cause muscle relaxation and narrowing of the airway (opiates, benzodiazepines, alcohol), endocrine disorders (hypothyroidism, polycystic ovarian syndrome), smoking, and nasal congestion or obstruction.6

Sex

Men are at higher risk for OSA than women although once women reach menopause they have a risk similar to men. Postmenopausal women on hormone replacement therapy were found to have lower rates of OSA, suggesting that loss of hormones results in greater risk of OSA.7,8 Women also have more OSA during rapid eye movement (REM) sleep and less OSA when sleeping supine, whereas most men have OSA when sleeping supine.9,10 OSA is less severe in women compared with men of similar body mass index (BMI).11 Symptoms vary in men and women: snoring and witnessed apneas are more common in men whereas insomnia and excessive daytime sleepiness are more common in women.11 This may account for delayed diagnosis and the higher mortality in women compared with men.

Age

The risk of OSA increases with age. In a study of men 65 or older, the prevalence of moderate OSA was 23% in men younger than 72 and 30% in men older than 80.12 By comparison, the prevalence of moderate OSA in men 30 to 40 years was 10%.3 Increased risk of OSA with age may be due to age-related reduction in slow wave sleep (ie, deep sleep), which is protective against sleep-disordered breathing and airway collapse.13 Older adults are also less symptomatic, reporting less daytime sleepiness and fatigue.14

Race

The Sleep Heart Health Study found a slightly increased risk of moderate to severe OSA in blacks (20%) and American Indians (23%) compared with whites (17%).2 Another study showed the  prevalence of OSA was 30% in whites, 32% in blacks, 38% in Hispanics, and 39% in Chinese individuals.15 A higher prevalence of OSA in young blacks (≤ 25 years) compared with whites was reported,16 although another study found no differences based on race in older patients.17 These differences among racial groups may be due to variations in craniofacial anatomy.

Obesity

There is a correlation between increased risk of OSA and obesity (BMI > 30 kg/m2) and its correlates of greater waist-to-hip ratio and neck circumference.2 A 10% increase in body weight results in a sixfold increase in moderate to severe OSA and increases the apnea–hypopnea index (AHI; number of breath pauses or respiratory events per hour) by 32% whereas a 10% decrease in weight decreases the AHI by 26%.18

COMORBIDITIES

OSA is associated with a number of comorbid conditions including stroke, myocardial infarction, hypertension, hyperlipidemia, glucose intolerance, diabetes, arrhythmias including atrial fibrillation, pulmonary hypertension, congestive heart failure, and depression. Patients with moderate or severe OSA are at higher risk of these comorbid conditions.19

Patients with cardiovascular disease have a very high prevalence of OSA: hypertension (83% mild to 30% moderate to severe OSA), heart failure (55% to 12%), arrhythmias (50% to 20%), stroke (75% to 57%), and coronary heart disease (65% to 38%).20 Increased awareness and early diagnosis of OSA is critical to reducing cardiovascular disease burden.

 

 

SCREENING

vensel_rundo_osabasics_t2.jpg
Screening patients for OSA starts with a good sleep history to identify symptoms, risk factors, and comorbid conditions, as well as a physical examination for OSA-related features (Table 2). The Epworth Sleepiness Scale and STOP-BANG questionnaire are brief, effective screening tools that can inform the need for further testing.

Sleep history

A sleep history starts with determining the patient’s total sleep time, based on time to bed, time to fall asleep, and time of wake up, including any difficulty falling asleep, staying asleep, or daytime naps.

Symptoms. Daytime naps generally indicate a sleep deficit or sleep that is not refreshing. A review of sleep and daytime symptoms associated with OSA (Table 1) helps determine if excessive daytime sleepiness or unrefreshing sleep is out of proportion with the amount of sleep the patient is getting at night.

Some patients with OSA may have memory or concentration issues or feel like they have attention deficit disorder. In fact some patients are diagnosed with attention deficit disorder because of their insufficient sleep or unrefreshing sleep.

Drowsy driving is a special concern in patients with untreated OSA and sleep deprivation. Many patients have drowsy driving episodes or difficulty staying awake during long-distance driving. Caffeine use is also important information as excessive caffeine may be used to combat sleepiness during the day.

The Epworth Sleepiness Scale is a clinical screening tool that presents 8 situations for patients to consider and indicate their level of sleepiness and likelihood of falling asleep (never = 0; slight = 1; moderate = 2, high = 3).21,22 A total score ≥ 10 is considered abnormal in that the patient is excessively sleepy compared with most people.

Risk factors and comorbid conditions. OSA risk factors and comorbidities, including a BMI obesity assessment, should be reviewed with patients. Nasal congestion or mouth breathing especially at night could be due to airway obstruction increasing the risk of OSA. Family history of OSA, tobacco, alcohol use, other medical conditions, and medications should also be discussed.

Physical examination

vensel_rundo_osabasics_f1.jpg
%3Cp%3EFigure%201.%20Friedman%20palate%20positions%20(classes%201%2C%202%2C%203%2C%20and%204).%3C%2Fp%3E
Certain findings on physical examination could suggest the presence of OSA:

  • Neck circumference greater than 17 inches for men or greater than 16 inches for women
  • BMI greater than 30
  • Friedman class tongue position class 3 or greater (Figure 1)
  • Mouth features (present/enlarged tonsils, macroglossia, jaw misalignment)
  • Nasal abnormalities (turbinate hypertrophy, deviated septum).5

Patients with Friedman palate positions class 3 and 4 have a higher risk of OSA due to airway crowding during sleep when the airway naturally collapses a little and is even more restricted.

Narrow airways or oropharyngeal crowding can also be due to a swollen, enlarged, or elongated uvula; present or enlarged tonsils; or lateral wall narrowing. Alone or in combination, these features can contribute to airway obstruction.

Other signs in the mouth suggestive of obstruction are macroglossia (enlarged tongue) and tongue ridging. Tongue ridging or scalloping impressions typically occur during sleep and are caused by the tongue moving forward to open the airway and pressing against the teeth.

Retrognathia (lower jaw offset behind upper jaw) can narrow the airway and increase the risk of OSA as can a high arch palate, overbite (upper teeth forward), or overjet (upper teeth over the top of lower teeth).

A nasal examination for nasal valve collapse (ie, nostril collapses with inhalation), deviated septum, and inferior turbinate hypertrophy impart an increased risk of OSA.

Screening tools

In addition to the Epworth Sleepiness Scale, the STOP-BANG questionnaire can help determine if a patient should be tested further for OSA. The STOP-BANG questionnaire consists of 8 yes-no questions where more than 2 yes responses indicate the patient is at higher risk for moderate to severe OSA (93% sensitivity): Snore, Tired, Observed stopped breathing, high blood Pressure, BMI > 35 kg/m2, Age > 50, Neck > 15.75 inches, Gender = male).23

 

 

SLEEP STUDIES

vensel_rundo_osabasics_f2.jpg
%3Cp%3EFigure%202.%20Polysomnogram%20excerpts%20with%20normal%20sleep%2C%20obstructive%20apnea%2C%20obstructive%20hypopnea%2C%20and%20respiratory%20event-related%20arousal%20waveform%20findings.%3C%2Fp%3E
Polysomnography (PSG) is the gold standard of evaluation for OSA. The more recently availabile home sleep apnea test (HSAT) is convenient for select patients as a confirmatory test but results may underestimate the severity of sleep-related breathing disorders.

Polysomnography

vensel_rundo_osabasics_t3.jpg
PSG is a monitored, 8-hour sleep study conducted in a laboratory with an established scoring criteria for OSA-related respiratory events.24 The test can be tailored to a patient’s clinical history to determine the need for supplemental oxygen and positive airway pressure titration, detect elevated carbon dioxide (hypercapnia or hypoventilation) due to shallow breathing, and monitor for seizures or parasomnias. The PSG also records REM and nonREM sleep for REM-related sleep disorders, body position (supine and off supine), and variability in muscle tone that corresponds to the different stages of sleep (Figure 2, Table 3).

vensel_rundo_osabasics_f3.jpg
%3Cp%3EFigure%203.%20A%20hypnogram%20depicts%20the%20recorded%20levels%20of%20sleep%20over%20time.%3C%2Fp%3E
Hypnogram. A hypnogram is a type of polysomnography that illustrates the different stages of sleep over time: wake, stage 1, stage 2, and stage 3, and REM sleep (Figure 3). In a typical night’s sleep of 7 to 9 hours, patients cycle through the sleep stages 4 to 5 times. A hypnogram can also include waveforms for other parameters such as body position, respiratory events (apnea and hypopneas), microarousals, continuous positive airway pressure therapy, and oxygen saturation.

Home sleep apnea test

HSATs record 4 to 7 parameters including airflow (thermal and nasal pressure), effort (inductive ple­thysmography), and oximetry. No electroencephalogram is used, so sleep is not recorded; it is assumed the patient is sleeping for the duration of the test. As such, respiratory events are based on oxygen desaturations and reduced airflow and pressure as well as chest and abdomen effort. The raw data are edited and manually scored and reviewed by a sleep specialist.25

Although the HSAT is convenient for many patients, it underestimates the severity of sleep-related breathing disorders. HSAT is intended to confirm OSA in patients with a high likelihood of OSA based on their sleep history.26 It is ideally employed for adult patients with no major medical problems or other sleep problems who are at high risk for moderate to severe OSA based on the STOP-BANG questionnaire or those with daytime sleepiness and 2 of the 3 symptoms of snoring, witnessed apnea, or hypertension.27

A negative or inconclusive HSAT warrants a PSG to ensure the patient does not have OSA. Use of HSAT is contraindicated in patients with

  • Significant cardiopulmonary disease
  • Potential weakness due to a neuro­muscular condition
  • Awake hypoventilation or high risk for sleep-related hypoventilation (severe obesity)
  • History of stroke
  • Chronic opioid use
  • Severe insomnia
  • Symptoms of other significant sleep disorders
  • Environmental/personal factors that would preclude adequate acquisition and interpretation of data (disruptions from children, pets, other factors).27

DIAGNOSTIC CRITERIA

vensel_rundo_osabasics_t4.jpg
Results from a PSG or HSAT inform the diagnosis of OSA and the need for treatment. The current diagnostic criteria for OSA were established in 2014 by the American Academy of Sleep Medicine (Table 4).28

Respiratory events captured on a PSG or HSAT

The OSA diagnostic criteria are based on the occurrence of obstructive respiratory events recorded during sleep such as apneas, hypopneas, and respiratory event-related arousals.

vensel_rundo_osabasics_f4.jpg
%3Cp%3EFigure%204.%20Apneas%20can%20be%20obstructive%2C%20mixed%2C%20or%20central.%3C%2Fp%3E
Apneas. An apnea is a respiratory event resulting in a complete lack of airflow as measured by a greater than 90% reduction in thermal sensor for 10 or more seconds. Apneas can be obstructive, central, or mixed (Figure 4). Obstructive apneas occur when the airway is closed and respiratory effort is present in the chest and abdomen (Figure 2). In central apnea, there is no airflow and no respiratory effort, meaning the brain is not directing the body to breathe. Mixed apneas cause a lack of airflow with and without respiratory effort.

Hypopneas. A hypopnea is a respiratory event resulting in reduced airflow. The America Association of Sleep Medicine’s preferred definition is a reduction in nasal pressure of at least 30% for 10 seconds or longer with 3% or greater oxygen desaturation or an electroencephalogram arousal. Another acceptable definition is at least 30% reduction in thoracoabdominal movement or airflow with 4% or greater oxygen desaturation, which is used by the Centers for Medicare and Medicaid Services and other insurers.29,30 Hypopnea requires greater oxygen desaturation and is not dependent on arousals, which can sometimes make it more challenging to identify OSA (Figure 2).

Respiratory event-related arousals. Respiratory event-related arousals are respiratory events not meeting apnea or hypopnea criteria. They are measured as a sequence of breaths of 10 or more seconds with increasing respiratory effort or flattening of the nasal pressure waveform leading to arousal (Figure 2).29 Respiratory event-related arousals are disruptive to sleep and have many of the same consequences as apneas and hypopneas.

Severity

vensel_rundo_osabasics_t5.jpg
A diagnosis of OSA should include a measure of severity (mild, moderate, or severe) as the severity may determine if a patient with OSA is treated or not. Severity is determined by AHI, respiratory disturbance index, or respiratory event index (Table 5).29 For any of the 3 indexes, a value 5 to 14.9 is considered mild, 15 to 29.9 is considered moderate, and 30 or greater is considered severe.

SUMMARY

OSA results from airway collapse and obstruction during sleep, often causing arousal from sleep with or without oxygen desaturation. The prevalence of OSA is underestimated and it is underdiagnosed despite known risk factors and comorbid conditions. Screening for OSA with a sleep history, simple upper airway examination, and quick validated screening tool like the STOP-BANG or Epworth Sleepiness Scale aid in identifying the need for testing for OSA. A laboratory sleep study with a PSG can confirm the diagnosis and severity of OSA. HSATs are available to confirm the diagnosis of OSA in patients at high risk for moderate to severe OSA.

References
  1. Heinzer R, Vat S, Marques-Vidal P, et al. Prevalence of sleep-disordered breathing in the general population: the HypnoLaus study. Lancet Respir Med 2015; 3(4):310–318.
  2. Young T, Shahar E, Nieto FJ, et al; for the Sleep Heart Health Study Research Group. Predictors of sleep-disordered breathing in community-dwelling adults. Arch Intern Med 2002; 162(8):893–900.
  3. 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.
  4. Young T, Evans L, Finn L, Palta M. Estimation of clinically diagnosed proportion of sleep apnea syndrome in middle-aged men and women. Sleep 1997; 20(9):705–706.
  5. Epstein LJ, Kristo D, Strollo Jr, PJ, 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.
  6. Young T, Skatrud J, Peppard PE. Risk factors for obstructive sleep apnea in adults. JAMA 2004; 291(16):2013–2016.
  7. Young T, Finn L, Austin D, Peterson A. Menopausal status and sleep-disordered breathing in the Wisconsin Sleep Cohort Study. Am J Respir Crit Care Med 2003; 167(9):1181–1185.
  8. Shahar E, Redline S, Young T, et al; for the Sleep Heart Health Study Research Group. Hormone replacement therapy and sleep-disordered breathing. Am J Respir Crit Care Med 2003; 167(9):1186–1192.
  9. O’Connor C, Thornley KS, Hanly PJ. Gender differences in the polysomnographic features of obstructive sleep apnea. Am J Respir Crit Care Med 2000; 161(5):1465–1472.
  10. Collop NA, Adkins D, Phillips BA. Gender differences in sleep and sleep-disordered breathing. Clin Chest Med 2004; 25(2):257–268.
  11. Redline S, Kump K, Tishler PV, Browner I, Ferrette V. Gender differences in sleep disordered breathing in a community-based sample. Am J Respir Crit Care Med 1994; 149(3 Pt 1):722–726.
  12. Mehra R, Stone KL, Blackwell T, et al; for the Osteoporotic Fractures in Men Study. Prevalence and correlates of sleep-disordered breathing in older men: Osteoporotic Fractures in Men Sleep Study. J Am Geriatr Soc 2007; 55(9):1356–1364.
  13. Van Cauter E, Leproult R, Plat L. Age-related changes in slow wave sleep and REM sleep and relationship with growth hormone and cortisol levels in healthy men. JAMA 2000; 284(7):861–868.
  14. Groth M. Sleep apnea in the elderly. Clin Geriatr Med 2005; 21:701–712.
  15. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  16. Redline S, Tishler PV, Hans MG, Tosteson TD, Strohl KP, Spry K. Racial differences in sleep-disordered breathing in African-Americans and Caucasians. Am J Respir Crit Care Med 1997; 155(1):186–192.
  17. Song Y, Ancoli-Israel S, Lewis CE, Redline S, Harrison SL, Stone KL. The association of race/ethnicity with objectively measured sleep characteristics in older men. Behav Sleep Med 2011; 10(1):54–69.
  18. Peppard PE, Young T, Palta M, Dempsey J, Skatrud J. Longitudinal study of moderate weight change and sleep-disordered breathing. JAMA 2000; 284(23):3015–3021.
  19. Shahar E, Whitney CW, Redline S, et al. Sleep-disordered breathing and cardiovascular disease:  cross-sectional results of the Sleep Heart Health Study. Am J Respir Crit Care Med. 2001;163:19–25
  20. 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.
  21. Johns MW. Daytime sleepiness, snoring, and obstructive sleep apnea. Chest 1993; 103(1):30–36.
  22. Chervin RD, Aldrich MS. The Epworth Sleepiness Scale may not reflect objective measures of sleepiness or sleep apnea. Neurology 1999; 52(1):125–131.
  23. Chung F, Yegneswaran B, Liao P, et al. STOP questionnaire: a tool to screen patients for obstructive sleep apnea. Anesthesiology 2008; 108(5):812–821.
  24. Iber C, Ancoli-Israel S, Chesson A, Quan SF; for the American Academy of Sleep and Medicine. The ASSM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. 1st ed. Winchester, IL: American Academy of Sleep Medicine; 2007.
  25. Centers for Medicare and Medicaid Services. Medicare Learning Network. Continuous positive airway pressure (CPAP) therapy for obstructive sleep apnea (OSA). www.cms.gov/Outreach-and-Education/Medicare-Learning-Network-MLN/MLNMattersArticles/downloads/mm6048.pdf. Accessed August 19, 2019.
  26. Collop NA, Anderson WM, Boehlecke B, et al; Portable Monitoring Task Force of the American Academy of Sleep Medicine. Clinical guidelines for the use of unattended portable monitors in the diagnosis of obstructive sleep apnea in adult patients. J Clin Sleep Med 2007; 3(7):737–747.
  27. 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.
  28. Sateia MJ. International classification of sleep disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  29. AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. Version 2.5. American Academy of Sleep Medicine; April 2018.
  30. Centers for Medicare and Medicaid Services. Medicare Coverage Database. www.cms.gov/medicare-coverage-database. Accessed August 19, 2019.
Article PDF
Author and Disclosure Information

Jessica Vensel Rundo, MD, MS
Sleep Fellowship Program Director, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Jessica Vensel Rundo, MD, MS, Sleep Disorders Center, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; venselj@ccf.org

Dr. Vensel Rundo reported no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Vensel Rundo’s “Obstructive Sleep Apnea Basics” webcast released January 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Vensel Rundo.

Publications
Topics
Page Number
2-9
Legacy Keywords
Obstructive sleep apnea, OSA, apneas, hypopneas, respiratory-event related arousals, RERAs, Epworth Sleepiness Scale, ESS, STOP-BANG, polysomnogram, PSG, home sleep apnea test, HSAT, apnea-hypopnea index, AHI, respiratory disturbance index, RDI, respiratory event index, REI, Jessica vensel rundo, nancy foldvary-schaefer
Sections
Author and Disclosure Information

Jessica Vensel Rundo, MD, MS
Sleep Fellowship Program Director, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Jessica Vensel Rundo, MD, MS, Sleep Disorders Center, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; venselj@ccf.org

Dr. Vensel Rundo reported no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Vensel Rundo’s “Obstructive Sleep Apnea Basics” webcast released January 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Vensel Rundo.

Author and Disclosure Information

Jessica Vensel Rundo, MD, MS
Sleep Fellowship Program Director, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Jessica Vensel Rundo, MD, MS, Sleep Disorders Center, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; venselj@ccf.org

Dr. Vensel Rundo reported no financial interests or relationships that pose a potential conflict of interest with this article.

This article is based on Dr. Vensel Rundo’s “Obstructive Sleep Apnea Basics” webcast released January 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Vensel Rundo.

Article PDF
Article PDF
Related Articles

DEFINITION

Obstructive sleep apnea (OSA) occurs when there are recurrent episodes of upper airway collapse and obstruction during sleep associated with arousals with or without oxygen desaturations. The oropharynx in the back of the throat collapses during OSA events to cause arousal or oxygen desaturation or both resulting in fragmented sleep.

PREVALENCE

Studies reveal OSA is prevalent. A 2015 study in Switzerland reported 50% of men and 23% of women had at least moderate OSA.1 In 2002, the Sleep Heart Health study found that 24% of men and 9% of women have at least mild OSA.2 In the Wisconsin Sleep Study Cohort, it was reported that 10% of men and 3% of women age 30 to 49 have at least moderate OSA, while 17% of men and 9% of women age 50 to 70 have at least moderate OSA.3 OSA is highly underrecognized and it is estimated that 82% of men and 93% of women in the United States with OSA are undiagnosed.4

SYMPTOMS

vensel_rundo_osabasics_t1.jpg
There are several common sleep and daytime symptoms associated with OSA though patients vary in the number and combination of symptoms reported (Table 1).5 During sleep, snoring is one of the most common symptoms. Common daytime symptoms of OSA include excessive daytime sleepiness or fatigue. Excessive daytime sleepiness is feeling very drowsy or very sleepy at times whereas fatigue is feeling tired, low on energy, and unmotivated. Feeling unrefreshed despite getting the recommended 7 to 9 hours of sleep is also a symptom.

RISK FACTORS

The risk of OSA is influenced by unmodifiable and modifiable factors. Unmodifiable risk factors include male sex, age, and race. Genetic predisposition or a family history of OSA as well as cranial facial anatomy resulting in narrow airways may impart higher risk of OSA. Modifiable risk factors include obesity, medications that cause muscle relaxation and narrowing of the airway (opiates, benzodiazepines, alcohol), endocrine disorders (hypothyroidism, polycystic ovarian syndrome), smoking, and nasal congestion or obstruction.6

Sex

Men are at higher risk for OSA than women although once women reach menopause they have a risk similar to men. Postmenopausal women on hormone replacement therapy were found to have lower rates of OSA, suggesting that loss of hormones results in greater risk of OSA.7,8 Women also have more OSA during rapid eye movement (REM) sleep and less OSA when sleeping supine, whereas most men have OSA when sleeping supine.9,10 OSA is less severe in women compared with men of similar body mass index (BMI).11 Symptoms vary in men and women: snoring and witnessed apneas are more common in men whereas insomnia and excessive daytime sleepiness are more common in women.11 This may account for delayed diagnosis and the higher mortality in women compared with men.

Age

The risk of OSA increases with age. In a study of men 65 or older, the prevalence of moderate OSA was 23% in men younger than 72 and 30% in men older than 80.12 By comparison, the prevalence of moderate OSA in men 30 to 40 years was 10%.3 Increased risk of OSA with age may be due to age-related reduction in slow wave sleep (ie, deep sleep), which is protective against sleep-disordered breathing and airway collapse.13 Older adults are also less symptomatic, reporting less daytime sleepiness and fatigue.14

Race

The Sleep Heart Health Study found a slightly increased risk of moderate to severe OSA in blacks (20%) and American Indians (23%) compared with whites (17%).2 Another study showed the  prevalence of OSA was 30% in whites, 32% in blacks, 38% in Hispanics, and 39% in Chinese individuals.15 A higher prevalence of OSA in young blacks (≤ 25 years) compared with whites was reported,16 although another study found no differences based on race in older patients.17 These differences among racial groups may be due to variations in craniofacial anatomy.

Obesity

There is a correlation between increased risk of OSA and obesity (BMI > 30 kg/m2) and its correlates of greater waist-to-hip ratio and neck circumference.2 A 10% increase in body weight results in a sixfold increase in moderate to severe OSA and increases the apnea–hypopnea index (AHI; number of breath pauses or respiratory events per hour) by 32% whereas a 10% decrease in weight decreases the AHI by 26%.18

COMORBIDITIES

OSA is associated with a number of comorbid conditions including stroke, myocardial infarction, hypertension, hyperlipidemia, glucose intolerance, diabetes, arrhythmias including atrial fibrillation, pulmonary hypertension, congestive heart failure, and depression. Patients with moderate or severe OSA are at higher risk of these comorbid conditions.19

Patients with cardiovascular disease have a very high prevalence of OSA: hypertension (83% mild to 30% moderate to severe OSA), heart failure (55% to 12%), arrhythmias (50% to 20%), stroke (75% to 57%), and coronary heart disease (65% to 38%).20 Increased awareness and early diagnosis of OSA is critical to reducing cardiovascular disease burden.

 

 

SCREENING

vensel_rundo_osabasics_t2.jpg
Screening patients for OSA starts with a good sleep history to identify symptoms, risk factors, and comorbid conditions, as well as a physical examination for OSA-related features (Table 2). The Epworth Sleepiness Scale and STOP-BANG questionnaire are brief, effective screening tools that can inform the need for further testing.

Sleep history

A sleep history starts with determining the patient’s total sleep time, based on time to bed, time to fall asleep, and time of wake up, including any difficulty falling asleep, staying asleep, or daytime naps.

Symptoms. Daytime naps generally indicate a sleep deficit or sleep that is not refreshing. A review of sleep and daytime symptoms associated with OSA (Table 1) helps determine if excessive daytime sleepiness or unrefreshing sleep is out of proportion with the amount of sleep the patient is getting at night.

Some patients with OSA may have memory or concentration issues or feel like they have attention deficit disorder. In fact some patients are diagnosed with attention deficit disorder because of their insufficient sleep or unrefreshing sleep.

Drowsy driving is a special concern in patients with untreated OSA and sleep deprivation. Many patients have drowsy driving episodes or difficulty staying awake during long-distance driving. Caffeine use is also important information as excessive caffeine may be used to combat sleepiness during the day.

The Epworth Sleepiness Scale is a clinical screening tool that presents 8 situations for patients to consider and indicate their level of sleepiness and likelihood of falling asleep (never = 0; slight = 1; moderate = 2, high = 3).21,22 A total score ≥ 10 is considered abnormal in that the patient is excessively sleepy compared with most people.

Risk factors and comorbid conditions. OSA risk factors and comorbidities, including a BMI obesity assessment, should be reviewed with patients. Nasal congestion or mouth breathing especially at night could be due to airway obstruction increasing the risk of OSA. Family history of OSA, tobacco, alcohol use, other medical conditions, and medications should also be discussed.

Physical examination

vensel_rundo_osabasics_f1.jpg
%3Cp%3EFigure%201.%20Friedman%20palate%20positions%20(classes%201%2C%202%2C%203%2C%20and%204).%3C%2Fp%3E
Certain findings on physical examination could suggest the presence of OSA:

  • Neck circumference greater than 17 inches for men or greater than 16 inches for women
  • BMI greater than 30
  • Friedman class tongue position class 3 or greater (Figure 1)
  • Mouth features (present/enlarged tonsils, macroglossia, jaw misalignment)
  • Nasal abnormalities (turbinate hypertrophy, deviated septum).5

Patients with Friedman palate positions class 3 and 4 have a higher risk of OSA due to airway crowding during sleep when the airway naturally collapses a little and is even more restricted.

Narrow airways or oropharyngeal crowding can also be due to a swollen, enlarged, or elongated uvula; present or enlarged tonsils; or lateral wall narrowing. Alone or in combination, these features can contribute to airway obstruction.

Other signs in the mouth suggestive of obstruction are macroglossia (enlarged tongue) and tongue ridging. Tongue ridging or scalloping impressions typically occur during sleep and are caused by the tongue moving forward to open the airway and pressing against the teeth.

Retrognathia (lower jaw offset behind upper jaw) can narrow the airway and increase the risk of OSA as can a high arch palate, overbite (upper teeth forward), or overjet (upper teeth over the top of lower teeth).

A nasal examination for nasal valve collapse (ie, nostril collapses with inhalation), deviated septum, and inferior turbinate hypertrophy impart an increased risk of OSA.

Screening tools

In addition to the Epworth Sleepiness Scale, the STOP-BANG questionnaire can help determine if a patient should be tested further for OSA. The STOP-BANG questionnaire consists of 8 yes-no questions where more than 2 yes responses indicate the patient is at higher risk for moderate to severe OSA (93% sensitivity): Snore, Tired, Observed stopped breathing, high blood Pressure, BMI > 35 kg/m2, Age > 50, Neck > 15.75 inches, Gender = male).23

 

 

SLEEP STUDIES

vensel_rundo_osabasics_f2.jpg
%3Cp%3EFigure%202.%20Polysomnogram%20excerpts%20with%20normal%20sleep%2C%20obstructive%20apnea%2C%20obstructive%20hypopnea%2C%20and%20respiratory%20event-related%20arousal%20waveform%20findings.%3C%2Fp%3E
Polysomnography (PSG) is the gold standard of evaluation for OSA. The more recently availabile home sleep apnea test (HSAT) is convenient for select patients as a confirmatory test but results may underestimate the severity of sleep-related breathing disorders.

Polysomnography

vensel_rundo_osabasics_t3.jpg
PSG is a monitored, 8-hour sleep study conducted in a laboratory with an established scoring criteria for OSA-related respiratory events.24 The test can be tailored to a patient’s clinical history to determine the need for supplemental oxygen and positive airway pressure titration, detect elevated carbon dioxide (hypercapnia or hypoventilation) due to shallow breathing, and monitor for seizures or parasomnias. The PSG also records REM and nonREM sleep for REM-related sleep disorders, body position (supine and off supine), and variability in muscle tone that corresponds to the different stages of sleep (Figure 2, Table 3).

vensel_rundo_osabasics_f3.jpg
%3Cp%3EFigure%203.%20A%20hypnogram%20depicts%20the%20recorded%20levels%20of%20sleep%20over%20time.%3C%2Fp%3E
Hypnogram. A hypnogram is a type of polysomnography that illustrates the different stages of sleep over time: wake, stage 1, stage 2, and stage 3, and REM sleep (Figure 3). In a typical night’s sleep of 7 to 9 hours, patients cycle through the sleep stages 4 to 5 times. A hypnogram can also include waveforms for other parameters such as body position, respiratory events (apnea and hypopneas), microarousals, continuous positive airway pressure therapy, and oxygen saturation.

Home sleep apnea test

HSATs record 4 to 7 parameters including airflow (thermal and nasal pressure), effort (inductive ple­thysmography), and oximetry. No electroencephalogram is used, so sleep is not recorded; it is assumed the patient is sleeping for the duration of the test. As such, respiratory events are based on oxygen desaturations and reduced airflow and pressure as well as chest and abdomen effort. The raw data are edited and manually scored and reviewed by a sleep specialist.25

Although the HSAT is convenient for many patients, it underestimates the severity of sleep-related breathing disorders. HSAT is intended to confirm OSA in patients with a high likelihood of OSA based on their sleep history.26 It is ideally employed for adult patients with no major medical problems or other sleep problems who are at high risk for moderate to severe OSA based on the STOP-BANG questionnaire or those with daytime sleepiness and 2 of the 3 symptoms of snoring, witnessed apnea, or hypertension.27

A negative or inconclusive HSAT warrants a PSG to ensure the patient does not have OSA. Use of HSAT is contraindicated in patients with

  • Significant cardiopulmonary disease
  • Potential weakness due to a neuro­muscular condition
  • Awake hypoventilation or high risk for sleep-related hypoventilation (severe obesity)
  • History of stroke
  • Chronic opioid use
  • Severe insomnia
  • Symptoms of other significant sleep disorders
  • Environmental/personal factors that would preclude adequate acquisition and interpretation of data (disruptions from children, pets, other factors).27

DIAGNOSTIC CRITERIA

vensel_rundo_osabasics_t4.jpg
Results from a PSG or HSAT inform the diagnosis of OSA and the need for treatment. The current diagnostic criteria for OSA were established in 2014 by the American Academy of Sleep Medicine (Table 4).28

Respiratory events captured on a PSG or HSAT

The OSA diagnostic criteria are based on the occurrence of obstructive respiratory events recorded during sleep such as apneas, hypopneas, and respiratory event-related arousals.

vensel_rundo_osabasics_f4.jpg
%3Cp%3EFigure%204.%20Apneas%20can%20be%20obstructive%2C%20mixed%2C%20or%20central.%3C%2Fp%3E
Apneas. An apnea is a respiratory event resulting in a complete lack of airflow as measured by a greater than 90% reduction in thermal sensor for 10 or more seconds. Apneas can be obstructive, central, or mixed (Figure 4). Obstructive apneas occur when the airway is closed and respiratory effort is present in the chest and abdomen (Figure 2). In central apnea, there is no airflow and no respiratory effort, meaning the brain is not directing the body to breathe. Mixed apneas cause a lack of airflow with and without respiratory effort.

Hypopneas. A hypopnea is a respiratory event resulting in reduced airflow. The America Association of Sleep Medicine’s preferred definition is a reduction in nasal pressure of at least 30% for 10 seconds or longer with 3% or greater oxygen desaturation or an electroencephalogram arousal. Another acceptable definition is at least 30% reduction in thoracoabdominal movement or airflow with 4% or greater oxygen desaturation, which is used by the Centers for Medicare and Medicaid Services and other insurers.29,30 Hypopnea requires greater oxygen desaturation and is not dependent on arousals, which can sometimes make it more challenging to identify OSA (Figure 2).

Respiratory event-related arousals. Respiratory event-related arousals are respiratory events not meeting apnea or hypopnea criteria. They are measured as a sequence of breaths of 10 or more seconds with increasing respiratory effort or flattening of the nasal pressure waveform leading to arousal (Figure 2).29 Respiratory event-related arousals are disruptive to sleep and have many of the same consequences as apneas and hypopneas.

Severity

vensel_rundo_osabasics_t5.jpg
A diagnosis of OSA should include a measure of severity (mild, moderate, or severe) as the severity may determine if a patient with OSA is treated or not. Severity is determined by AHI, respiratory disturbance index, or respiratory event index (Table 5).29 For any of the 3 indexes, a value 5 to 14.9 is considered mild, 15 to 29.9 is considered moderate, and 30 or greater is considered severe.

SUMMARY

OSA results from airway collapse and obstruction during sleep, often causing arousal from sleep with or without oxygen desaturation. The prevalence of OSA is underestimated and it is underdiagnosed despite known risk factors and comorbid conditions. Screening for OSA with a sleep history, simple upper airway examination, and quick validated screening tool like the STOP-BANG or Epworth Sleepiness Scale aid in identifying the need for testing for OSA. A laboratory sleep study with a PSG can confirm the diagnosis and severity of OSA. HSATs are available to confirm the diagnosis of OSA in patients at high risk for moderate to severe OSA.

DEFINITION

Obstructive sleep apnea (OSA) occurs when there are recurrent episodes of upper airway collapse and obstruction during sleep associated with arousals with or without oxygen desaturations. The oropharynx in the back of the throat collapses during OSA events to cause arousal or oxygen desaturation or both resulting in fragmented sleep.

PREVALENCE

Studies reveal OSA is prevalent. A 2015 study in Switzerland reported 50% of men and 23% of women had at least moderate OSA.1 In 2002, the Sleep Heart Health study found that 24% of men and 9% of women have at least mild OSA.2 In the Wisconsin Sleep Study Cohort, it was reported that 10% of men and 3% of women age 30 to 49 have at least moderate OSA, while 17% of men and 9% of women age 50 to 70 have at least moderate OSA.3 OSA is highly underrecognized and it is estimated that 82% of men and 93% of women in the United States with OSA are undiagnosed.4

SYMPTOMS

vensel_rundo_osabasics_t1.jpg
There are several common sleep and daytime symptoms associated with OSA though patients vary in the number and combination of symptoms reported (Table 1).5 During sleep, snoring is one of the most common symptoms. Common daytime symptoms of OSA include excessive daytime sleepiness or fatigue. Excessive daytime sleepiness is feeling very drowsy or very sleepy at times whereas fatigue is feeling tired, low on energy, and unmotivated. Feeling unrefreshed despite getting the recommended 7 to 9 hours of sleep is also a symptom.

RISK FACTORS

The risk of OSA is influenced by unmodifiable and modifiable factors. Unmodifiable risk factors include male sex, age, and race. Genetic predisposition or a family history of OSA as well as cranial facial anatomy resulting in narrow airways may impart higher risk of OSA. Modifiable risk factors include obesity, medications that cause muscle relaxation and narrowing of the airway (opiates, benzodiazepines, alcohol), endocrine disorders (hypothyroidism, polycystic ovarian syndrome), smoking, and nasal congestion or obstruction.6

Sex

Men are at higher risk for OSA than women although once women reach menopause they have a risk similar to men. Postmenopausal women on hormone replacement therapy were found to have lower rates of OSA, suggesting that loss of hormones results in greater risk of OSA.7,8 Women also have more OSA during rapid eye movement (REM) sleep and less OSA when sleeping supine, whereas most men have OSA when sleeping supine.9,10 OSA is less severe in women compared with men of similar body mass index (BMI).11 Symptoms vary in men and women: snoring and witnessed apneas are more common in men whereas insomnia and excessive daytime sleepiness are more common in women.11 This may account for delayed diagnosis and the higher mortality in women compared with men.

Age

The risk of OSA increases with age. In a study of men 65 or older, the prevalence of moderate OSA was 23% in men younger than 72 and 30% in men older than 80.12 By comparison, the prevalence of moderate OSA in men 30 to 40 years was 10%.3 Increased risk of OSA with age may be due to age-related reduction in slow wave sleep (ie, deep sleep), which is protective against sleep-disordered breathing and airway collapse.13 Older adults are also less symptomatic, reporting less daytime sleepiness and fatigue.14

Race

The Sleep Heart Health Study found a slightly increased risk of moderate to severe OSA in blacks (20%) and American Indians (23%) compared with whites (17%).2 Another study showed the  prevalence of OSA was 30% in whites, 32% in blacks, 38% in Hispanics, and 39% in Chinese individuals.15 A higher prevalence of OSA in young blacks (≤ 25 years) compared with whites was reported,16 although another study found no differences based on race in older patients.17 These differences among racial groups may be due to variations in craniofacial anatomy.

Obesity

There is a correlation between increased risk of OSA and obesity (BMI > 30 kg/m2) and its correlates of greater waist-to-hip ratio and neck circumference.2 A 10% increase in body weight results in a sixfold increase in moderate to severe OSA and increases the apnea–hypopnea index (AHI; number of breath pauses or respiratory events per hour) by 32% whereas a 10% decrease in weight decreases the AHI by 26%.18

COMORBIDITIES

OSA is associated with a number of comorbid conditions including stroke, myocardial infarction, hypertension, hyperlipidemia, glucose intolerance, diabetes, arrhythmias including atrial fibrillation, pulmonary hypertension, congestive heart failure, and depression. Patients with moderate or severe OSA are at higher risk of these comorbid conditions.19

Patients with cardiovascular disease have a very high prevalence of OSA: hypertension (83% mild to 30% moderate to severe OSA), heart failure (55% to 12%), arrhythmias (50% to 20%), stroke (75% to 57%), and coronary heart disease (65% to 38%).20 Increased awareness and early diagnosis of OSA is critical to reducing cardiovascular disease burden.

 

 

SCREENING

vensel_rundo_osabasics_t2.jpg
Screening patients for OSA starts with a good sleep history to identify symptoms, risk factors, and comorbid conditions, as well as a physical examination for OSA-related features (Table 2). The Epworth Sleepiness Scale and STOP-BANG questionnaire are brief, effective screening tools that can inform the need for further testing.

Sleep history

A sleep history starts with determining the patient’s total sleep time, based on time to bed, time to fall asleep, and time of wake up, including any difficulty falling asleep, staying asleep, or daytime naps.

Symptoms. Daytime naps generally indicate a sleep deficit or sleep that is not refreshing. A review of sleep and daytime symptoms associated with OSA (Table 1) helps determine if excessive daytime sleepiness or unrefreshing sleep is out of proportion with the amount of sleep the patient is getting at night.

Some patients with OSA may have memory or concentration issues or feel like they have attention deficit disorder. In fact some patients are diagnosed with attention deficit disorder because of their insufficient sleep or unrefreshing sleep.

Drowsy driving is a special concern in patients with untreated OSA and sleep deprivation. Many patients have drowsy driving episodes or difficulty staying awake during long-distance driving. Caffeine use is also important information as excessive caffeine may be used to combat sleepiness during the day.

The Epworth Sleepiness Scale is a clinical screening tool that presents 8 situations for patients to consider and indicate their level of sleepiness and likelihood of falling asleep (never = 0; slight = 1; moderate = 2, high = 3).21,22 A total score ≥ 10 is considered abnormal in that the patient is excessively sleepy compared with most people.

Risk factors and comorbid conditions. OSA risk factors and comorbidities, including a BMI obesity assessment, should be reviewed with patients. Nasal congestion or mouth breathing especially at night could be due to airway obstruction increasing the risk of OSA. Family history of OSA, tobacco, alcohol use, other medical conditions, and medications should also be discussed.

Physical examination

vensel_rundo_osabasics_f1.jpg
%3Cp%3EFigure%201.%20Friedman%20palate%20positions%20(classes%201%2C%202%2C%203%2C%20and%204).%3C%2Fp%3E
Certain findings on physical examination could suggest the presence of OSA:

  • Neck circumference greater than 17 inches for men or greater than 16 inches for women
  • BMI greater than 30
  • Friedman class tongue position class 3 or greater (Figure 1)
  • Mouth features (present/enlarged tonsils, macroglossia, jaw misalignment)
  • Nasal abnormalities (turbinate hypertrophy, deviated septum).5

Patients with Friedman palate positions class 3 and 4 have a higher risk of OSA due to airway crowding during sleep when the airway naturally collapses a little and is even more restricted.

Narrow airways or oropharyngeal crowding can also be due to a swollen, enlarged, or elongated uvula; present or enlarged tonsils; or lateral wall narrowing. Alone or in combination, these features can contribute to airway obstruction.

Other signs in the mouth suggestive of obstruction are macroglossia (enlarged tongue) and tongue ridging. Tongue ridging or scalloping impressions typically occur during sleep and are caused by the tongue moving forward to open the airway and pressing against the teeth.

Retrognathia (lower jaw offset behind upper jaw) can narrow the airway and increase the risk of OSA as can a high arch palate, overbite (upper teeth forward), or overjet (upper teeth over the top of lower teeth).

A nasal examination for nasal valve collapse (ie, nostril collapses with inhalation), deviated septum, and inferior turbinate hypertrophy impart an increased risk of OSA.

Screening tools

In addition to the Epworth Sleepiness Scale, the STOP-BANG questionnaire can help determine if a patient should be tested further for OSA. The STOP-BANG questionnaire consists of 8 yes-no questions where more than 2 yes responses indicate the patient is at higher risk for moderate to severe OSA (93% sensitivity): Snore, Tired, Observed stopped breathing, high blood Pressure, BMI > 35 kg/m2, Age > 50, Neck > 15.75 inches, Gender = male).23

 

 

SLEEP STUDIES

vensel_rundo_osabasics_f2.jpg
%3Cp%3EFigure%202.%20Polysomnogram%20excerpts%20with%20normal%20sleep%2C%20obstructive%20apnea%2C%20obstructive%20hypopnea%2C%20and%20respiratory%20event-related%20arousal%20waveform%20findings.%3C%2Fp%3E
Polysomnography (PSG) is the gold standard of evaluation for OSA. The more recently availabile home sleep apnea test (HSAT) is convenient for select patients as a confirmatory test but results may underestimate the severity of sleep-related breathing disorders.

Polysomnography

vensel_rundo_osabasics_t3.jpg
PSG is a monitored, 8-hour sleep study conducted in a laboratory with an established scoring criteria for OSA-related respiratory events.24 The test can be tailored to a patient’s clinical history to determine the need for supplemental oxygen and positive airway pressure titration, detect elevated carbon dioxide (hypercapnia or hypoventilation) due to shallow breathing, and monitor for seizures or parasomnias. The PSG also records REM and nonREM sleep for REM-related sleep disorders, body position (supine and off supine), and variability in muscle tone that corresponds to the different stages of sleep (Figure 2, Table 3).

vensel_rundo_osabasics_f3.jpg
%3Cp%3EFigure%203.%20A%20hypnogram%20depicts%20the%20recorded%20levels%20of%20sleep%20over%20time.%3C%2Fp%3E
Hypnogram. A hypnogram is a type of polysomnography that illustrates the different stages of sleep over time: wake, stage 1, stage 2, and stage 3, and REM sleep (Figure 3). In a typical night’s sleep of 7 to 9 hours, patients cycle through the sleep stages 4 to 5 times. A hypnogram can also include waveforms for other parameters such as body position, respiratory events (apnea and hypopneas), microarousals, continuous positive airway pressure therapy, and oxygen saturation.

Home sleep apnea test

HSATs record 4 to 7 parameters including airflow (thermal and nasal pressure), effort (inductive ple­thysmography), and oximetry. No electroencephalogram is used, so sleep is not recorded; it is assumed the patient is sleeping for the duration of the test. As such, respiratory events are based on oxygen desaturations and reduced airflow and pressure as well as chest and abdomen effort. The raw data are edited and manually scored and reviewed by a sleep specialist.25

Although the HSAT is convenient for many patients, it underestimates the severity of sleep-related breathing disorders. HSAT is intended to confirm OSA in patients with a high likelihood of OSA based on their sleep history.26 It is ideally employed for adult patients with no major medical problems or other sleep problems who are at high risk for moderate to severe OSA based on the STOP-BANG questionnaire or those with daytime sleepiness and 2 of the 3 symptoms of snoring, witnessed apnea, or hypertension.27

A negative or inconclusive HSAT warrants a PSG to ensure the patient does not have OSA. Use of HSAT is contraindicated in patients with

  • Significant cardiopulmonary disease
  • Potential weakness due to a neuro­muscular condition
  • Awake hypoventilation or high risk for sleep-related hypoventilation (severe obesity)
  • History of stroke
  • Chronic opioid use
  • Severe insomnia
  • Symptoms of other significant sleep disorders
  • Environmental/personal factors that would preclude adequate acquisition and interpretation of data (disruptions from children, pets, other factors).27

DIAGNOSTIC CRITERIA

vensel_rundo_osabasics_t4.jpg
Results from a PSG or HSAT inform the diagnosis of OSA and the need for treatment. The current diagnostic criteria for OSA were established in 2014 by the American Academy of Sleep Medicine (Table 4).28

Respiratory events captured on a PSG or HSAT

The OSA diagnostic criteria are based on the occurrence of obstructive respiratory events recorded during sleep such as apneas, hypopneas, and respiratory event-related arousals.

vensel_rundo_osabasics_f4.jpg
%3Cp%3EFigure%204.%20Apneas%20can%20be%20obstructive%2C%20mixed%2C%20or%20central.%3C%2Fp%3E
Apneas. An apnea is a respiratory event resulting in a complete lack of airflow as measured by a greater than 90% reduction in thermal sensor for 10 or more seconds. Apneas can be obstructive, central, or mixed (Figure 4). Obstructive apneas occur when the airway is closed and respiratory effort is present in the chest and abdomen (Figure 2). In central apnea, there is no airflow and no respiratory effort, meaning the brain is not directing the body to breathe. Mixed apneas cause a lack of airflow with and without respiratory effort.

Hypopneas. A hypopnea is a respiratory event resulting in reduced airflow. The America Association of Sleep Medicine’s preferred definition is a reduction in nasal pressure of at least 30% for 10 seconds or longer with 3% or greater oxygen desaturation or an electroencephalogram arousal. Another acceptable definition is at least 30% reduction in thoracoabdominal movement or airflow with 4% or greater oxygen desaturation, which is used by the Centers for Medicare and Medicaid Services and other insurers.29,30 Hypopnea requires greater oxygen desaturation and is not dependent on arousals, which can sometimes make it more challenging to identify OSA (Figure 2).

Respiratory event-related arousals. Respiratory event-related arousals are respiratory events not meeting apnea or hypopnea criteria. They are measured as a sequence of breaths of 10 or more seconds with increasing respiratory effort or flattening of the nasal pressure waveform leading to arousal (Figure 2).29 Respiratory event-related arousals are disruptive to sleep and have many of the same consequences as apneas and hypopneas.

Severity

vensel_rundo_osabasics_t5.jpg
A diagnosis of OSA should include a measure of severity (mild, moderate, or severe) as the severity may determine if a patient with OSA is treated or not. Severity is determined by AHI, respiratory disturbance index, or respiratory event index (Table 5).29 For any of the 3 indexes, a value 5 to 14.9 is considered mild, 15 to 29.9 is considered moderate, and 30 or greater is considered severe.

SUMMARY

OSA results from airway collapse and obstruction during sleep, often causing arousal from sleep with or without oxygen desaturation. The prevalence of OSA is underestimated and it is underdiagnosed despite known risk factors and comorbid conditions. Screening for OSA with a sleep history, simple upper airway examination, and quick validated screening tool like the STOP-BANG or Epworth Sleepiness Scale aid in identifying the need for testing for OSA. A laboratory sleep study with a PSG can confirm the diagnosis and severity of OSA. HSATs are available to confirm the diagnosis of OSA in patients at high risk for moderate to severe OSA.

References
  1. Heinzer R, Vat S, Marques-Vidal P, et al. Prevalence of sleep-disordered breathing in the general population: the HypnoLaus study. Lancet Respir Med 2015; 3(4):310–318.
  2. Young T, Shahar E, Nieto FJ, et al; for the Sleep Heart Health Study Research Group. Predictors of sleep-disordered breathing in community-dwelling adults. Arch Intern Med 2002; 162(8):893–900.
  3. 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.
  4. Young T, Evans L, Finn L, Palta M. Estimation of clinically diagnosed proportion of sleep apnea syndrome in middle-aged men and women. Sleep 1997; 20(9):705–706.
  5. Epstein LJ, Kristo D, Strollo Jr, PJ, 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.
  6. Young T, Skatrud J, Peppard PE. Risk factors for obstructive sleep apnea in adults. JAMA 2004; 291(16):2013–2016.
  7. Young T, Finn L, Austin D, Peterson A. Menopausal status and sleep-disordered breathing in the Wisconsin Sleep Cohort Study. Am J Respir Crit Care Med 2003; 167(9):1181–1185.
  8. Shahar E, Redline S, Young T, et al; for the Sleep Heart Health Study Research Group. Hormone replacement therapy and sleep-disordered breathing. Am J Respir Crit Care Med 2003; 167(9):1186–1192.
  9. O’Connor C, Thornley KS, Hanly PJ. Gender differences in the polysomnographic features of obstructive sleep apnea. Am J Respir Crit Care Med 2000; 161(5):1465–1472.
  10. Collop NA, Adkins D, Phillips BA. Gender differences in sleep and sleep-disordered breathing. Clin Chest Med 2004; 25(2):257–268.
  11. Redline S, Kump K, Tishler PV, Browner I, Ferrette V. Gender differences in sleep disordered breathing in a community-based sample. Am J Respir Crit Care Med 1994; 149(3 Pt 1):722–726.
  12. Mehra R, Stone KL, Blackwell T, et al; for the Osteoporotic Fractures in Men Study. Prevalence and correlates of sleep-disordered breathing in older men: Osteoporotic Fractures in Men Sleep Study. J Am Geriatr Soc 2007; 55(9):1356–1364.
  13. Van Cauter E, Leproult R, Plat L. Age-related changes in slow wave sleep and REM sleep and relationship with growth hormone and cortisol levels in healthy men. JAMA 2000; 284(7):861–868.
  14. Groth M. Sleep apnea in the elderly. Clin Geriatr Med 2005; 21:701–712.
  15. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  16. Redline S, Tishler PV, Hans MG, Tosteson TD, Strohl KP, Spry K. Racial differences in sleep-disordered breathing in African-Americans and Caucasians. Am J Respir Crit Care Med 1997; 155(1):186–192.
  17. Song Y, Ancoli-Israel S, Lewis CE, Redline S, Harrison SL, Stone KL. The association of race/ethnicity with objectively measured sleep characteristics in older men. Behav Sleep Med 2011; 10(1):54–69.
  18. Peppard PE, Young T, Palta M, Dempsey J, Skatrud J. Longitudinal study of moderate weight change and sleep-disordered breathing. JAMA 2000; 284(23):3015–3021.
  19. Shahar E, Whitney CW, Redline S, et al. Sleep-disordered breathing and cardiovascular disease:  cross-sectional results of the Sleep Heart Health Study. Am J Respir Crit Care Med. 2001;163:19–25
  20. 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.
  21. Johns MW. Daytime sleepiness, snoring, and obstructive sleep apnea. Chest 1993; 103(1):30–36.
  22. Chervin RD, Aldrich MS. The Epworth Sleepiness Scale may not reflect objective measures of sleepiness or sleep apnea. Neurology 1999; 52(1):125–131.
  23. Chung F, Yegneswaran B, Liao P, et al. STOP questionnaire: a tool to screen patients for obstructive sleep apnea. Anesthesiology 2008; 108(5):812–821.
  24. Iber C, Ancoli-Israel S, Chesson A, Quan SF; for the American Academy of Sleep and Medicine. The ASSM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. 1st ed. Winchester, IL: American Academy of Sleep Medicine; 2007.
  25. Centers for Medicare and Medicaid Services. Medicare Learning Network. Continuous positive airway pressure (CPAP) therapy for obstructive sleep apnea (OSA). www.cms.gov/Outreach-and-Education/Medicare-Learning-Network-MLN/MLNMattersArticles/downloads/mm6048.pdf. Accessed August 19, 2019.
  26. Collop NA, Anderson WM, Boehlecke B, et al; Portable Monitoring Task Force of the American Academy of Sleep Medicine. Clinical guidelines for the use of unattended portable monitors in the diagnosis of obstructive sleep apnea in adult patients. J Clin Sleep Med 2007; 3(7):737–747.
  27. 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.
  28. Sateia MJ. International classification of sleep disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  29. AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. Version 2.5. American Academy of Sleep Medicine; April 2018.
  30. Centers for Medicare and Medicaid Services. Medicare Coverage Database. www.cms.gov/medicare-coverage-database. Accessed August 19, 2019.
References
  1. Heinzer R, Vat S, Marques-Vidal P, et al. Prevalence of sleep-disordered breathing in the general population: the HypnoLaus study. Lancet Respir Med 2015; 3(4):310–318.
  2. Young T, Shahar E, Nieto FJ, et al; for the Sleep Heart Health Study Research Group. Predictors of sleep-disordered breathing in community-dwelling adults. Arch Intern Med 2002; 162(8):893–900.
  3. 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.
  4. Young T, Evans L, Finn L, Palta M. Estimation of clinically diagnosed proportion of sleep apnea syndrome in middle-aged men and women. Sleep 1997; 20(9):705–706.
  5. Epstein LJ, Kristo D, Strollo Jr, PJ, 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.
  6. Young T, Skatrud J, Peppard PE. Risk factors for obstructive sleep apnea in adults. JAMA 2004; 291(16):2013–2016.
  7. Young T, Finn L, Austin D, Peterson A. Menopausal status and sleep-disordered breathing in the Wisconsin Sleep Cohort Study. Am J Respir Crit Care Med 2003; 167(9):1181–1185.
  8. Shahar E, Redline S, Young T, et al; for the Sleep Heart Health Study Research Group. Hormone replacement therapy and sleep-disordered breathing. Am J Respir Crit Care Med 2003; 167(9):1186–1192.
  9. O’Connor C, Thornley KS, Hanly PJ. Gender differences in the polysomnographic features of obstructive sleep apnea. Am J Respir Crit Care Med 2000; 161(5):1465–1472.
  10. Collop NA, Adkins D, Phillips BA. Gender differences in sleep and sleep-disordered breathing. Clin Chest Med 2004; 25(2):257–268.
  11. Redline S, Kump K, Tishler PV, Browner I, Ferrette V. Gender differences in sleep disordered breathing in a community-based sample. Am J Respir Crit Care Med 1994; 149(3 Pt 1):722–726.
  12. Mehra R, Stone KL, Blackwell T, et al; for the Osteoporotic Fractures in Men Study. Prevalence and correlates of sleep-disordered breathing in older men: Osteoporotic Fractures in Men Sleep Study. J Am Geriatr Soc 2007; 55(9):1356–1364.
  13. Van Cauter E, Leproult R, Plat L. Age-related changes in slow wave sleep and REM sleep and relationship with growth hormone and cortisol levels in healthy men. JAMA 2000; 284(7):861–868.
  14. Groth M. Sleep apnea in the elderly. Clin Geriatr Med 2005; 21:701–712.
  15. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  16. Redline S, Tishler PV, Hans MG, Tosteson TD, Strohl KP, Spry K. Racial differences in sleep-disordered breathing in African-Americans and Caucasians. Am J Respir Crit Care Med 1997; 155(1):186–192.
  17. Song Y, Ancoli-Israel S, Lewis CE, Redline S, Harrison SL, Stone KL. The association of race/ethnicity with objectively measured sleep characteristics in older men. Behav Sleep Med 2011; 10(1):54–69.
  18. Peppard PE, Young T, Palta M, Dempsey J, Skatrud J. Longitudinal study of moderate weight change and sleep-disordered breathing. JAMA 2000; 284(23):3015–3021.
  19. Shahar E, Whitney CW, Redline S, et al. Sleep-disordered breathing and cardiovascular disease:  cross-sectional results of the Sleep Heart Health Study. Am J Respir Crit Care Med. 2001;163:19–25
  20. 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.
  21. Johns MW. Daytime sleepiness, snoring, and obstructive sleep apnea. Chest 1993; 103(1):30–36.
  22. Chervin RD, Aldrich MS. The Epworth Sleepiness Scale may not reflect objective measures of sleepiness or sleep apnea. Neurology 1999; 52(1):125–131.
  23. Chung F, Yegneswaran B, Liao P, et al. STOP questionnaire: a tool to screen patients for obstructive sleep apnea. Anesthesiology 2008; 108(5):812–821.
  24. Iber C, Ancoli-Israel S, Chesson A, Quan SF; for the American Academy of Sleep and Medicine. The ASSM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. 1st ed. Winchester, IL: American Academy of Sleep Medicine; 2007.
  25. Centers for Medicare and Medicaid Services. Medicare Learning Network. Continuous positive airway pressure (CPAP) therapy for obstructive sleep apnea (OSA). www.cms.gov/Outreach-and-Education/Medicare-Learning-Network-MLN/MLNMattersArticles/downloads/mm6048.pdf. Accessed August 19, 2019.
  26. Collop NA, Anderson WM, Boehlecke B, et al; Portable Monitoring Task Force of the American Academy of Sleep Medicine. Clinical guidelines for the use of unattended portable monitors in the diagnosis of obstructive sleep apnea in adult patients. J Clin Sleep Med 2007; 3(7):737–747.
  27. 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.
  28. Sateia MJ. International classification of sleep disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  29. AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. Version 2.5. American Academy of Sleep Medicine; April 2018.
  30. Centers for Medicare and Medicaid Services. Medicare Coverage Database. www.cms.gov/medicare-coverage-database. Accessed August 19, 2019.
Page Number
2-9
Page Number
2-9
Publications
Publications
Topics
Article Type
Display Headline
Obstructive sleep apnea basics
Display Headline
Obstructive sleep apnea basics
Legacy Keywords
Obstructive sleep apnea, OSA, apneas, hypopneas, respiratory-event related arousals, RERAs, Epworth Sleepiness Scale, ESS, STOP-BANG, polysomnogram, PSG, home sleep apnea test, HSAT, apnea-hypopnea index, AHI, respiratory disturbance index, RDI, respiratory event index, REI, Jessica vensel rundo, nancy foldvary-schaefer
Legacy Keywords
Obstructive sleep apnea, OSA, apneas, hypopneas, respiratory-event related arousals, RERAs, Epworth Sleepiness Scale, ESS, STOP-BANG, polysomnogram, PSG, home sleep apnea test, HSAT, apnea-hypopnea index, AHI, respiratory disturbance index, RDI, respiratory event index, REI, Jessica vensel rundo, nancy foldvary-schaefer
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2019 September;86(9 suppl 1):2-9
Inside the Article

KEY POINTS

  • OSA is characterized by repeated episodes of complete or partial obstruction of the airway during sleep.
  • The prevalence of OSA is underestimated and underdiagnosed.
  • A sleep history, simple upper airway examination, and quick validated screening tool like the STOP-BANG or Epworth Sleepiness Scale aid in identifying the need for testing for OSA.
  • Polysomnogram is the gold standard for evaluation of OSA. Home sleep apnea tests can be used to confirm a diagnosis of OSA in patients at high risk for moderate to severe OSA.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 07:30
Un-Gate On Date
Wed, 08/28/2019 - 07:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 07:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Sleep apnea and the heart

Article Type
Changed
Tue, 09/03/2019 - 08:04
Display Headline
Sleep apnea and the heart

SLEEP AND CARDIOVASCULAR PHYSIOLOGY

Wakefullness and sleep, the latter comprised of non-rapid eye movement (NREM) sleep and rapid eye movement (REM) sleep, comprise our primary states of being. Sleep states oscillate between NREM and REM sleep. The first and shortest period of REM sleep typically occurs 90 to 120 minutes into the sleep cycle. Most REM sleep, including the longest period of REM sleep, occurs during the latter part of the sleep cycle.

With these sleep state changes, physiologic changes also occur, such as reduced heart rate and blood pressure because of enhanced parasympathetic tone. During REM sleep, there are also intermittent sympathetic nervous system surges. Other physiologic changes include a regular respiratory rate during NREM sleep and an irregular respiratory rate during REM sleep. Body temperature is normal during NREM sleep and poikilothermic (ie, tends to flucuate) during REM sleep. Blood pressure is reduced 10% to 15% during sleep1 and then rises, so that the highest blood pressure occurs in the morning. Data from 10 million users of activity-monitoring devices show that the heart rate changes during sleep.2 The heart rate is decreased in those who get less than 7 hours of sleep, then increases with longer sleep duration in a U-shaped distribution.

Cardiovascular events are more likely to occur at certain times of day. Myocardial infarction is more likely in the morning, with a threefold increased risk within the first 3 hours of awakening that peaks around 9 AM.3,4 Similar diurnal patterns have been observed with other cardiovascular conditions such as sudden cardiac death and ischemic episodes, with the highest risk during morning hours (6 to 9 AM).4

The reason for this morning predisposition for cardio­vascular events is unclear, but it is thought that perhaps the autonomic fluctuations that occur during REM sleep and the predominance of REM sleep in early morning may be a factor. Diurnal changes in blood pressure and cortisol levels may also contribute, as well as levels of systemic inflammatory and thrombotic markers such as plasminogen activator inhibitor 1.

Arrhythmias are also more likely to occur in a diurnal pattern. Atrial fibrillation (AF), particularly paroxysmal AF, is believed to be vagally mediated in 10% to 25% of patients.5 Therefore, for those who are predisposed, sleep may represent a period of increased risk for AF. In a study of individuals 60 years and older, the maximum duration and peak frequency of AF occurred from midnight to 2 AM.5

Recent studies have found that REM-related obstructive sleep apnea (OSA) is associated with increased cardiovascular risk. Experimental models show that REM sleep may increase the risk for compromised coronary blood flow.6 Increased heart rate corresponds to reduced coronary blood flow and thus, to decreased coronary perfusion time and less time for relaxation of the heart, increasing the risk for coronary artery disease, thrombosis, and ischemia.

SLEEP APNEA PATHOPHYSIOLOGY

The normal physiology of the sleep-heart inter­action is disrupted by sleep apnea. OSA is defined as episodes of complete or partial airway obstruction that occur during sleep with thoraco­abdominal effort. Central sleep apnea (CSA) is the cessation of breathing with no thoracoabdominal effort. The pathophysiology of the sleep-heart interaction varies for OSA and CSA.

Obstructive sleep apnea

OSA is a nocturnal physiologic stressor that is highly prevalent and underrecognized. It affects approximately 17% of the adult population, and the prevalence is increasing with the obesity epidemic. Nearly 1 in 15 individuals is estimated to be affected by at least moderate OSA.7,8 OSA is underdiagnosed particularly in minority populations.9 Data from the 2015 Multi-Ethnic Study of Atherosclerosis (MESA) showed undiagnosed moderate to severe sleep apnea in 84% to 93% of individuals,9 similar to an estimated 85% of undiagnosed cases in 2002.10

OSA is highly prevalent in individuals with underlying coronary disease11–13 and in those with cardiovascular risk factors such as diabetes, hypertension, and heart failure. The prevalence of OSA in patients with cardiovascular disease ranges from 30% (hypertension) to 60% (stroke or transient ischemic attack, arrhythmia, end-stage renal disease).14

 

 

Pathophysiology of OSA

mehra_sleepapneaandtheheart_f1.jpg
%3Cp%3EFigure%201.%20A%20polysomnogram%20showing%20autonomic%20nervous%20system%20disturbances%2C%20intermittent%20hypoxia%2C%20and%20intrathoracic%20pressure%20alterations.%3C%2Fp%3E
The pathophysiology of OSA can be observed during polysomnography, characterized by autonomic nervous system disturbances, intermittent hypoxia, and intrathoracic pressure alterations, (Figure 1). Intermittent bouts of hypoxia or oxygen-lowering occur because airflow is obstructed despite persistent thoracic and abdominal effort. Systemic inflammation and oxidative stress occur due to these intrathoracic pressure alterations, increased CO2 and reduced oxygen levels, and autonomic nervous system disturbances.

The alterations in sympathetic activation that occur during sleep in patients with OSA persist during wakefulness. Microneurographic recording of sympathetic nerve activity in the peroneal nerve reveal that the rate of sympathetic bursts doubles and the amplitude is greater in individuals with OSA compared with a control group.15

Sympathetic nerve activity, blood pressure, and heart rate were shown to increase during REM sleep in individuals with OSA on continuous positive airway pressure (CPAP) during an induced apneic event (pressure reduction from 8 cm to 6 cm of water).15

During OSA episodes, there is an increased cardiac load. Impaired diastolic function and atrial and aortic enlargement, and in particular, the thin-walled atria are very susceptible to the intra­thoracic pressure swings caused by OSA. Physiologic changes with OSA from pressure changes in the chest result in shift of the intraventricular septum, causing a reduction in cardiac output.16 With the lowering of oxygen during episodes of apnea, constriction of the pulmonary vasculature leads to elevation of pressure in the pulmonary vasculature reflected by the increase in mean pulmonary arterial pressures.17

Other studies have shown that OSA increases upregulation of markers of systemic inflammation and prothrombotic markers, the very markers that can increase cardiovascular or atherogenic risk.18–22 One example is the soluble interleukin 6 receptor, shown to be elevated in the morning relative to sleep apnea compared with the evening.20 Other biomarkers observed to be associated with sleep apnea include markers of prothrombotic potentials such as plasminogen activator inhibitor 1.19 Oxidative stress occurs because intermittent bouts of lower oxygen can lead to oxidation of serum proteins and lipids. Endothelial dysfunction has been observed as well as insulin resistance and dyslipidemia.23 Taken together, these are pathways that lead to atherogenesis and increased cardiovascular risk.

Central sleep apnea

CSA episodes are the cessation of breathing without thoracoabdominal effort, in contrast to the persistence of thoracoabdominal effort in OSA. CSA is characterized by breathing instability with highly sensitive chemoresponses and prolonged circulation time.24 This can be physiologic in some cases, as when it occurs after a very large breath or sigh and then a central apnea event occurs after the sigh. The alterations in oxygen and CO2 and the stretch of the receptors in the alveoli of the lungs initiate the Hering-Breuer inhalation reflex.

Pathophysiology of CSA

Complex pathways of medullary and aortic receptor chemosensitivity are at the root of the pathophysiology of CSA.24 With CSA there is often a relative state of hypocapnia at baseline. During sleep, there is reduction in drive, thus chemo­sensitivity can be activated so that central apnea episodes can ensue as a result of alterations in CO2 (ie, hypocapnia). Another factor that can contribute to the pathophysiology of CSA is arousal from sleep that can reduce CO2 levels and therefore perpetuate central events.

The concept of loop gain is used to understand the pathophysiology of CSA. Loop gain is a measure of the relative stability of a ventilation system and indicates the likelihood of an individual to have periodic breathing. It is calculated by the response to a disturbance divided by the disturbance itself.25 With a high loop gain, there is a more pronounced or exuberant response to the disturbance, indicating more instability in the system and increasing the tendency for irregular breathing and CSA episodes.

Hunter-Cheyne-Stokes respiration occurs with CSA and is characterized by cyclical crescendo-decrescendo respiratory effort that occurs during wakefulness and sleep without upper-airway obstruction.26,27 Unlike OSA, which is worse during REM sleep, Hunter-Cheyne-Stokes breathing in CSA is typically worse in NREM sleep, during N1 and N2 in particular.

 

 

SLEEP APNEA AND HEART FAILURE

Both OSA and CSA are prevalent in patients with heart failure and may be associated with the progression of heart failure. CSA often occurs in patients with heart failure. The pathophysiology is multi­factorial, including pulmonary congestion that results in stretch of the J receptors in the alveoli, prolonged circulation time, and increased chemosensitivity.

Complex pathways in the neuroaxis or somnogenic biomarkers of inflammation or both may be implicated in the paradoxical lack of subjective sleepiness in the presence of increased objective measures of sleepiness in systolic heart failure. One study found a relationship with one biomarker of inflammation and oxidative stress as it relates to objective symptoms of sleepiness but not subjective symptoms of sleepiness.28

Another contributing factor in the relationship between OSA and CSA in heart failure has also been described related to rostral shifts in fluid to the neck and to the pulmonary receptors in the alveoli of the lungs.29 These rostral shifts in fluids may contribute to sleep apnea with parapharyngeal edema leading to OSA and pulmonary congestion leading to CSA.

Sleep apnea is associated with increased post-discharge mortality and hospitalization readmissions in the setting of acute heart failure.30 Mortality analysis of 1,096 patients admitted for decompensated heart failure found CSA and OSA were independently associated with mortality in patients compared with patients with no or minimal sleep-disordered breathing.30

CSA has also been shown to be a predictor of readmission in patients admitted for heart failure exacerbations.31 Targeting underlying CSA may reduce readmissions in those admitted with acute decompensated heart failure. While men were identified to be at increased risk of death relative to sleep-disordered breathing based on the initial results of the Sleep Heart Health Study, a subsequent epidemiologic substudy reflective of an older age group showed that OSA was more strongly associated with left ventricular mass index, risk of heart failure, or death in women compared with men.32

Treatment

Standard therapy for treatment of OSA is CPAP. Adaptive servo-ventilation (ASV) and transvenous phrenic nerve stimulation are also available as treatment options in certain cases of CSA.

One of the first randomized controlled trials designed to assess the impact of CSA treatment on survival in patients with heart failure initially favored the control group then later the CPAP group and was terminated early based on stopping rules.33,34 While adherence to therapy was suboptimal at an average of 3.6 hours, post hoc analysis showed that patients with CSA using CPAP with effective suppression of CSA had improved survival compared with patients who did not have effective suppression using CPAP.34

ASV is mainly used for treatment of CSA. In ASV, positive airway pressure for ventilation support is provided and adjusts as apneic episodes are detected during sleep. The support provided adapts to the physiology of the patient and can deliver breaths and utilize anticyclic modes of ventilation to address crescendo-decrescendo breathing patterns observed in Hunter-Cheyne-Stokes respiration.

In the Treatment of Sleep-Disordered Breathing With Predominant Central Sleep Apnea by Adaptive Servo Ventilation in Patients With Heart Failure (SERVE-HF) trial, 1,300 patients with systolic heart failure and predominantly CSA were randomized to receive ASV vs solely standard medical management.35 The primary composite end point included all-cause mortality or unplanned admission or hospitalization for heart failure. No difference was found in the primary end point between the ASV and the control group; however, there was an unanticipated negative impact of ASV on cardiovascular outcomes in some secondary end points. Based on the secondary outcome of cardiovascular-specific mortality, clinicians were advised that ASV was contraindicated for the treatment of CSA in patients with symptomatic heart failure with a left ventricular ejection fraction less than 45%. The interpretation of this study was complicated by several methodologic limitations.36

The Cardiovascular Improvements With Minute Ventilation-Targeted Adaptive Servo-Ventilation Therapy in Heart Failure (CAT-HF) randomized controlled trial also evaluated ASV compared with standard medical management in 126 patients with heart failure.37 This trial was terminated early because of the results of the SERVE-HF trial. Compliance with therapy was suboptimal at an average of 2.7 hours per day. The composite end point did not differ between the 2 groups; however, this was likely because the study was underpowered and was terminated early. Subgroup analysis revealed that patients with heart failure with preserved ejection fraction may benefit from ASV; however, additional studies are needed to confirm these findings.

Therefore, although ASV is not indicated when there is predominantly CSA in patients with systolic heart failure, preliminary results support potential benefit in patients with OSA and preserved ejection fraction.

Another novel treatment for CSA is transvenous phrenic nerve stimulation. A device is implanted that stimulates the phrenic nerve to initiate breaths. The initial study of trans­venous phrenic nerve stimulation reported a significant reduction in the number of episodes of central apnea per hour of sleep.38,39 The apnea–hypopnea index improved overall and some types of obstructive apneic events were reduced with transvenous phrenic nerve stimulation.

A multicenter randomized control trial of trans­venous phrenic nerve stimulation found improvement in several sleep apnea indices, including central apnea, hypoxia, reduced arousals from sleep, and patient reported well-being.40 Transvenous phrenic nerve stimulation holds promise as a novel therapy for central predominant sleep apnea not only in terms of improving the degree of central apnea and sleep-disordered breathing, but also in improving functional outcomes. Longitudinal and intereventional trial data are needed to clarify the impact of transvenous phrenic nerve stimulation on long-term cardiac outcomes.

SLEEP APNEA AND ATRIAL FIBRILLATION AND STROKE

Atrial fibrillation

AF is the most common sustained cardiac arrhythmia. The number of Americans with AF is projected to increase from 2.3 million to more than 10 million by the year 2050.41 The increasing incidence and prevalence of AF is not fully explained by the aging population and established risk factors.42 Unrecognized sleep apnea, estimated to exist in 85% or more of the population, may partially account for the increasing incidence of AF.43

There are 3 types of AF, which are thought to follow a continuum: paroxysmal AF is characterized by episodes that occur intermittently; persistent AF is characterized by episodes that last longer than 7 days; chronic or permanent AF is typically characterized by AF that is ongoing over many years.44 As with sleep apnea, AF is often asymptomatic and is likely underdiagnosed.

Sleep apnea and AF share several risk factors. Obesity is a risk factor for both OSA and AF; however, a meta-analysis supported a stronger association of OSA and AF vs obesity and AF.45 Increasing age is a risk factor for both OSA and AF.46,47 Although white populations are at higher risk for AF, OSA is associated with a 58% increased risk of AF in African Americans.48 Nocturnal hypoxia has been associated with increased risk of AF in Asians.49

mehra_sleepapneaandtheheart_f2.jpg
%3Cp%3EFigure%202.%20Pathophysiologic%20pathways%20of%20obstructive%20sleep%20apnea%20and%20cardiac%20arrhythmia.%3C%2Fp%3E
In terms of pathophysiology of sleep apnea and cardiac arrhythmia, OSA increases inflammation, intrathoracic pressures, and CO2 levels. The increase in inflammation and oxidative stress is thought to alter the cardiac electrophysiology of the heart and contribute to structural remodeling of the heart that increases the risk of cardiac arrhythmia (Figure 2).50

Experimental data continue to accrue providing biologic plausibility of the relationship between sleep apnea and AF. OSA contributes to structural and electrical remodeling of the heart with evidence supporting increased fibrosis and electrical remodeling in patients with OSA compared with a control group.51 Markers of structural remodeling, such as atrial size, electrical silence, and atrial voltage conduction velocity, are altered in OSA.50

Data from the Sleep Heart Health Study show very strong associations between atrial and ventricular cardiac arrhythmias and sleep apnea with two- to fivefold higher odds of arrhythmias in patients with severe OSA compared with controls even after accounting for confounding factors such as obesity.52

A multicenter, epidemiological study of older men showed that increasing severity of sleep apnea corresponds with an increased prevalence of AF and ventricular ectopy.53 This graded dose-response relationship suggests a causal relationship between sleep apnea and AF and ventricular ectopy. There also appears to be an immediate influence of apneic events and hypopneic events as it relates to arrhythmia. A case-crossover study showed an associated 18-fold increased risk of nocturnal arrhythmia within 90 seconds of an apneic or hypopneic event.54 This association was found with paroxysms of AF and with episodes of nonsustained ventricular tachycardia.

Data from a clinic-based cohort study show an association between AF and OSA.55 Specifically, increased severity of sleep apnea was associated with an increased prevalence of AF. Increasing degree of hypoxia or oxygen-lowering was also associated with increased incidence of AF or newly identified AF identified over time.

Longitudinal examination of 2 epidemiologic studies, the Sleep Heart Health Study and Outcomes of Sleep Disorders Study in Older Men, found CSA to be predictive of AF with a two- to threefold higher odds of developing incident AF as it related to baseline CSA.56 According to these data, CSA may pose a greater risk for development of AF than OSA.

With respect to AF after cardiac surgery, patients with sleep apnea and obesity appear to be at higher risk for developing AF as measured by the apnea–hypopnea index and oxygen desaturation index.57

Treatment of sleep apnea may improve arrhythmic burden. Case-based studies have shown reduced burden and resolution of baseline arrhythmia with CPAP treatment for OSA as therapeutic pressure was achieved.58 Another case-based study involved an individual with snoring and OSA and AF at baseline.59 Several retrospective studies have shown that treatment of OSA after ablation and after cardio­version results in reduced recurrence of AF; however, large definitive clinical trials are lacking.

Stroke

Sleep apnea is a risk factor for stroke due to intermittent hypoxia-mediated elevation of oxidative stress and systemic inflammation, hypercoaguability, and impairment of cerebral autoregulation.60 However, the relationship may be bidirectional in that stroke may be a risk factor for sleep apnea in the post-stroke period. The prevalence of sleep apnea post-stroke has been reported to be up to 70%. CSA can occur in up to 26% during the post-stroke phase.61 Data are inconsistent in terms of the location and size of stroke and the risk of sleep apnea, though cerebrovascular neuronal damage to the brainstem and cortical areas are evident.62 In one study, the incidence of stroke appeared to increase with the severity of sleep apnea.63 These findings were more pronounced in men than in women; however, this study may not have captured the increased cardiovascular risk in postmenopausal women. The Outcomes of Sleep Disorders in Older Men study found that severe hypoxia increased the incidence of stroke, and that hypoxia may be a predictor of newly diagnosed stroke in older men.64 Although definitive clinical trials are underway, post-hoc propensity-score matched analysis from the Sleep Apnea Cardiovascular Endpoints (SAVE) study showed a lower stroke risk in those adherent to CPAP compared with the control group (HR=0.56, 95% CI: 0.30-0.90).65

 

 

SLEEP APNEA, CORONARY ARTERY DISEASE, AND CARIOVASCULAR MORTALITY

The association between sleep apnea and coronary artery disease and cardiovascular mortality was considered in a Spanish study of 1,500 patients followed for 10 years, which reported that CPAP therapy reduced cardiac events in patients with OSA.66 Patients with sleep apnea had an increased risk of fatal myocardial infarction or stroke. Survival of patients treated for sleep apnea approached that of patients without OSA.

In a study of a racially diverse cohort, an association of physician diagnosed sleep apnea with cardiovascular events and survival was identified.67 Diagnosed sleep apnea was estimated to confer a two- to threefold increase in various cardiovascular outcomes and all-cause mortality.

mehra_sleepapneaandtheheart_f3.jpg
%3Cp%3EFigure%203.%20Survival%20of%20patients%20with%20obstructive%20sleep%20apnea%20by%20apnea%E2%80%93hypopnea%20index.%3C%2Fp%3E
All-cause mortality data from the Sleep Heart Health Study of more than 6,000 participants showed that progressive worsening of OSA as defined by the apnea–hypopnea index resulted in poorer survival even after accounting for confounding factors (Figure 3).68 Decreased survival appeared to mostly affect men or patients under age 70.

mehra_sleepapneaandtheheart_t1.jpg
The diurnal pattern of cardiovascular physiology as it relates to sleep is thought to be cardioprotective because of reductions in blood pressure and heart rate. However, in the case of OSA, there appears to be a nocturnal vulnerability or predilection for sudden cardiac death. Patients with OSA were shown to have a higher risk of sudden nocturnal cardiac death occurring from midnight to 6 am compared with individuals without OSA and the general population (Table 1).69

The effect of treatment for sleep apnea on cardiovascular outcomes was the focus of a recent randomized controlled trial of nearly 3,000 participants with a mean follow-up of 4 years.65 Use of CPAP compared with usual care found no difference in cardiovascular outcomes. However, secondary analysis revealed a possible benefit of a lower risk of stroke with use of CPAP therapy. Several factors should be considered in interpreting these findings: ie, low adherence with CPAP therapy (3 hours), whether the study was sufficiently powered to detect a change in cardiovascular outcomes, and if the duration of follow-up was adequate. In terms of patient demographics and study generalizability, the study did not include patients with severe sleep apnea and hypoxia, and most participants were men, of Asian descent, with a mean body mass index of 28 kg/m2, and low levels of sleepiness at baseline.

References
  1. Kario K. Morning surge in blood pressure and cardiovascular risk: evidence and perspectives. Hypertension 2010; 56(5):765–773.
  2. FitBit: 150 billion data hrs shows sleep hours sweet spot, optimal health strategy. True Strange Library website. https://truestrange.com/2018/08/29/fitbit-150-billion-data-hrs-shows-sleep-hours-sweet-spot-optimal-health-strategy. Accessed August 19, 2019.
  3. Muller JE, Stone PH, Turi ZG, et al; MILIS Study Group. Circadian variation in the frequency of onset of acute myocardial infarction. N Engl J Med 1985; 313(21):1315–1322.
  4. Marler JR, Price TR, Clark GL, et al. Morning increase in onset of ischemic stroke. Stroke 1989; 20(4):473–476.
  5. Yamashita T, Murakawa Y, Hayami N, et al. Relation between aging and circadian variation of paroxysmal atrial fibrillation. Am J Cardiol 1998; 82(11):1364–1367.
  6. Kirby DA, Verrier RL. Differential effects of sleep stage on coronary hemodynamic function. Am J Physiol 1989; 256(5 Pt 2):H1378–H1383.
  7. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered breathing among middle-aged adults. N Engl J Med 1993; 328(17):1230–1235.
  8. 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.
  9. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  10. Kapur V, Strohl KP, Redline S, Iber C, O’Connor G, Nieto J. Under­diagnosis of sleep apnea syndrome in U.S. communities. Sleep Breath 2002; 6(2):49–54.
  11. Mooe T, Rabben T, Wiklund U, Franklin KA, Eriksson P. Sleep-disordered breathing in men with coronary artery disease. Chest 1996; 109(3):659–663.
  12. Schäfer H, Koehler U, Ewig S, Hasper E, Tasci S, Lüderitz B. Obstructive sleep apnea as a risk marker in coronary artery disease. Cardiology 1999; 92(2):79–84.
  13. Leung RST, Bradley TD. Sleep apnea and cardiovascular disease. Am J Respir Crit Care Med 2001; 164(12):2147–2165.
  14. Cepeda-Valery B, Acharjee S, Romero-Corral A, Pressman GS, Gami AS. Obstructive sleep apnea and acute coronary syndromes: etiology, risk, and management. Curr Cardiol Rep 2014; 16(10):535.
  15. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive sleep apnea. J Clin Invest 1995; 96(4):1897–1904.
  16. Kasai T, Bradley TD. Obstructive sleep apnea and heart failure: pathophysiologic and therapeutic implications. J Am Coll Cardiol 2011; 57(2):119–127.
  17. Sajkov D, McEvoy RD. Obstructive sleep apnea and pulmonary hypertension. Prog Cardiovasc Dis 2009; 51(5):363–370.
  18. Nadeem R, Molnar J, Madbouly EM, et al. Serum inflammatory markers in obstructive sleep apnea: a meta-analysis. J Clin Sleep Med 2013; 9(10):1003–1012.
  19. Mehra R, Xu F, Babineau DC, et al. Sleep-disordered breathing and prothrombotic biomarkers: cross-sectional results of the Cleveland Family Study. Am J Respir Crit Care Med 2010; 182(6):826–833.
  20. Mehra R, Storfer-Isser A, Kirchner HL, et al. Soluble interleukin 6 receptor: a novel marker of moderate to severe sleep-related breathing disorder. Arch Intern Med 2006; 166(16):1725–1731.
  21. Paz y Mar HL, Hazen SL, Tracy RP, et al. Effect of continuous positive airway pressure on cardiovascular biomarkers: the sleep apnea stress randomized controlled trial. Chest 2016; 150(1):80–90.
  22. Xie X, Pan L, Ren D, Du C, Guo Y. Effects of continuous positive airway pressure therapy on systemic inflammation in obstructive sleep apnea: a meta-analysis. Sleep Med 2013; 14(11):1139–1150.
  23. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005; 352(16):1685–1695.
  24. Eckert DJ, Jordan AS, Merchia P, Malhotra A. Central sleep apnea: pathophysiology and treatment. Chest 2007; 131(2):595–607.
  25. White DP. Pathogenesis of obstructive and central sleep apnea. Am J Respir Crit Care Med 2005; 172(11):1363–1370.
  26. Javaheri S. Heart failure. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th ed. Philadelphia, PA: Elsevier; 2017:1271–1285.
  27. Olson LJ, Somers VK. Treating central sleep apnea in heart failure: outcomes revisited. Circulation 2007; 115(25):3140–3142.
  28. Mehra R, Wang L, Andrews N, et al. Dissociation of objective and subjective daytime sleepiness and biomarkers of systemic inflammation in sleep-disordered breathing and systolic heart failure. J Clin Sleep Med 2017; 13(12):1411–1422.
  29. Kasai T, Floras JS, Bradley TD. Sleep apnea and cardiovascular disease: a bidirectional relationship. Circulation 2012; 126(12):1495–1510.
  30. Khayat R, Jarjoura D, Porter K, et al. Sleep disordered breathing and post-discharge mortality in patients with acute heart failure. Eur Heart J 2015; 36(23):1463–1469.
  31. Khayat R, Abraham W, Patt B, et al. Central sleep apnea is a predictor of cardiac readmission in hospitalized patients with systolic heart failure. J Card Fail 2012; 18(7):534–540.
  32. Roca GQ, Redline S, Claggett B, et al. Sex-specific association of sleep apnea severity with subclinical myocardial injury, ventricular hypertrophy, and heart failure risk in a community-dwelling cohort: the Atherosclerosis Risk in Communities–Sleep Heart Health Study. Circulation 2015; 132(14):1329–1337.
  33. Bradley TD, Logan AG, Kimoff RJ, et al; CANPAP Investigators. Continuous positive airway pressure for central sleep apnea and heart failure. N Engl J Med 2005; 353(19):2025–2033.
  34. Arzt M, Floras JS, Logan AG, et al; CANPAP Investigators. Suppression of central sleep apnea by continuous positive airway pressure and transplant-free survival in heart failure: a post hoc analysis of the Canadian Continuous Positive Airway Pressure for Patients with Central Sleep Apnea and Heart Failure Trial (CANPAP). Circulation 2007; 115(25):3173–3180.
  35. Cowie MR, Woehrle H, Wegscheider K, et al. Adaptive servo-ventilation for central sleep apnea in systolic heart failure. N Engl J Med 2015; 373(12):1095–1105.
  36. Mehra R, Gottlieb DJ. A paradigm shift in the treatment of central sleep apnea in heart failure. Chest 2015; 148(4):848–851.
  37. O’Connor CM, Whellan DJ, Fiuzat M, et al. Cardiovascular outcomes with minute ventilation-targeted adaptive servo-ventilation therapy in heart failure: the CAT-HF trial. J Am Coll Cardiol 2017; 69(12):1577–1587.
  38. Abraham WT, Jagielski D, Oldenburg O, et al; remede Pilot Study Investigators. Phrenic nerve stimulation for the treatment of central sleep apnea. JACC Heart Fail 2015; 3(5):360–369.
  39. Ponikowski P, Javaheri S, Michalkiewicz D, et al. Transvenous phrenic nerve stimulation for the treatment of central sleep apnoea in heart failure. Eur Heart J 2012; 33(7):889–894.
  40. Costanzo MR, Ponikowski P, Javaheri S, et al; remede System Pivotal Trial Study Group. Transvenous neurostimulation for central sleep apnoea: a randomised controlled trial. Lancet 2016; 388(10048):974–982.
  41. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370-2375.
  42. Wolf PA, Benjamin EJ, Belanger AJ, Kannel WB, Levy D, D’Agostino RB. Secular trends in the prevalence of atrial fibrillation: the Framingham Study. Am Heart J 1996; 131(4):790–795.
  43. Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125.
  44. Camm AJ, Kirchhof P, Lip GYH, et al; European Heart Rhythm Association; European Association for Cardio-Thoracic Surgery. Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart J 2010; 31(19):2369–2429.
  45. Trulock KM, Narayan SM, Piccini JP. Rhythm control in heart failure patients with atrial fibrillation: contemporary challenges including the role of ablation. J Am Coll Cardiol 2014; 64(7):710–721.
  46. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  47. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors In Atrial Fibrillation (ATRIA) study. JAMA 2001; 258(18):2370–2375.
  48. Kwon Y, Mehra R. Obstructive sleep apnea and atrial fibrillation: honing in on race-specific susceptibilities. J Clin Sleep Med 2018; 14(9):1459–1461.
  49. Mehra R. Sleep apnea and nocturnal cardiac arrhythmia: understanding differences across ethnicity. J Clin Sleep Med 2017; 13(11):1229–1231.
  50. May AM, Van Wagoner DR, Mehra R. OSA and cardiac arrhymogenesis: mechanistic insights. Chest 2017; 151(1):225–241.
  51. Dimitri H, Ng M, Brooks AG, et al. Atrial remodeling in obstructive sleep apnea: implications for atrial fibrillation. Heart Rhythm 2012; 9(3):321–327.
  52. Mehra R, Benjamin EJ, Shahar E, et al. Association of nocturnal arrhythmias with sleep-disordered breathing: the Sleep Heart Health Study. Am J Respir Crit Care Med 2006; 173(8):910–916.
  53. Mehra R, Stone KL, Varosy PD, et al. Nocturnal arrhythmias across a spectrum of obstructive and central sleep-disordered breathing in older men: outcomes of sleep disorders in older men (MrOS sleep) study. Arch Intern Med 2009; 169(12):1147–1155.
  54. Monahan K, Storfer-Isser A, Mehra R, et al. Triggering of nocturnal arrhythmias by sleep-disordered breathing events. J Am Coll Cardiol 2009; 54(19):1797–1804.
  55. Gami AS, Hodge DO, Herges RM, et al. Obstructive sleep apnea, obesity, and the risk of incident atrial fibrillation. J Am Coll Cardiol 2007; 49(5):565–571.
  56. May AM, Blackwell T, Stone PH, et al; MrOS Sleep (Outcomes of Sleep Disorders in Older Men) Study Group. Am J Respir Crit Care Med 2016; 193(7):783–791.
  57. Kaw R, El Zarif S, Wang L, Bena J, Blackstone EH, Mehra R. Obesity as an effect modifier in sleep-disordered breathing and postcardiac surgery atrial fibrillation. Chest 2017; 151(6):1279–1287.
  58. Walia H, Strohl KP, Mehra R. Effect of continuous positive airway pressure on an atrial arrhythmia in a patient with mild obstructive sleep apnea. J Clin Sleep Med 2011; 7(4):397–398.
  59. Walia HK, Chung MK, Ibrahim S, Mehra R. Positive airway pressure-induced conversion of atrial fibrillation to normal sinus rhythm in severe obstructive sleep apnea. J Clin Sleep Med 2016; 12(9):1301–1303.
  60. Veasey SC, Davis CW, Fenik P, et al. Long-term intermittent hypoxia in mice: protracted hypersomnolence with oxidative injury to sleep-wake brain regions. Sleep 2004; 27(2):194–201.
  61. Parra O, Arboix A, Bechich S, et al. Time course of sleep-related breathing disorders in first-ever stroke or transient ischemic attack. Am J Respir Crit Care Med 2000; 161(2I):375–380.
  62. Song TJ, Park JH, Choi K, et al. Moderate-to-severe obstructive sleep apnea is associated with cerebral small vessel disease. Sleep Med 2017; 30:36–42.
  63. 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.
  64. Stone KL, Blackwell TL, Ancoli-Israel S, et al; Osteoporotic Fractures in Men (MrOS) Study Research Group. Sleep disordered breathing and risk of stroke in older community-dwelling men. Sleep 2016; 39(3):531–540.
  65. McEvoy RD, Antic NA, Heeley E, et al; SAVE Investigators and Coordinators. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med 2016; 375(10):919–931.
  66. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardiovascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  67. Yeboah J, Redline S, Johnson C, et al. Association between sleep apnea, snoring, incident cardiovascular events and all-cause mortality in an adult population: MESA. Atherosclerosis 2011; 219(2):963–968.
  68. Punjabi NM, Caffo BS, Goodwin JL, et al. Sleep-disordered breathing and mortality: a prospective cohort study. PLoS Med 2009; 6(8):e1000132.
  69. Gami AS, Howard DE, Olson EJ, Somers VK. Day–night pattern of sudden death in obstructive sleep apnea. N Engl J Med 2005; 352(12):1206–1214.
Article PDF
Author and Disclosure Information

Reena Mehra, MD, MS
Director, Sleep Disorders Research Program, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Reena Mehra, MD, MS, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mehrar@ccf.org

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

This article is based on Dr. Mehra’s “Sleep Apnea and the Heart” webcast released February 28, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Mehra.

Publications
Topics
Page Number
10-18
Legacy Keywords
continuous positive airway pressure, CPAP, adaptive servo ventilation; phrenic nerve stimulation; sleep-heart interaction, reena mehra, nancy foldvary-schaefer
Sections
Author and Disclosure Information

Reena Mehra, MD, MS
Director, Sleep Disorders Research Program, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Reena Mehra, MD, MS, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mehrar@ccf.org

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

This article is based on Dr. Mehra’s “Sleep Apnea and the Heart” webcast released February 28, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Mehra.

Author and Disclosure Information

Reena Mehra, MD, MS
Director, Sleep Disorders Research Program, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Reena Mehra, MD, MS, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; mehrar@ccf.org

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

This article is based on Dr. Mehra’s “Sleep Apnea and the Heart” webcast released February 28, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Mehra.

Article PDF
Article PDF
Related Articles

SLEEP AND CARDIOVASCULAR PHYSIOLOGY

Wakefullness and sleep, the latter comprised of non-rapid eye movement (NREM) sleep and rapid eye movement (REM) sleep, comprise our primary states of being. Sleep states oscillate between NREM and REM sleep. The first and shortest period of REM sleep typically occurs 90 to 120 minutes into the sleep cycle. Most REM sleep, including the longest period of REM sleep, occurs during the latter part of the sleep cycle.

With these sleep state changes, physiologic changes also occur, such as reduced heart rate and blood pressure because of enhanced parasympathetic tone. During REM sleep, there are also intermittent sympathetic nervous system surges. Other physiologic changes include a regular respiratory rate during NREM sleep and an irregular respiratory rate during REM sleep. Body temperature is normal during NREM sleep and poikilothermic (ie, tends to flucuate) during REM sleep. Blood pressure is reduced 10% to 15% during sleep1 and then rises, so that the highest blood pressure occurs in the morning. Data from 10 million users of activity-monitoring devices show that the heart rate changes during sleep.2 The heart rate is decreased in those who get less than 7 hours of sleep, then increases with longer sleep duration in a U-shaped distribution.

Cardiovascular events are more likely to occur at certain times of day. Myocardial infarction is more likely in the morning, with a threefold increased risk within the first 3 hours of awakening that peaks around 9 AM.3,4 Similar diurnal patterns have been observed with other cardiovascular conditions such as sudden cardiac death and ischemic episodes, with the highest risk during morning hours (6 to 9 AM).4

The reason for this morning predisposition for cardio­vascular events is unclear, but it is thought that perhaps the autonomic fluctuations that occur during REM sleep and the predominance of REM sleep in early morning may be a factor. Diurnal changes in blood pressure and cortisol levels may also contribute, as well as levels of systemic inflammatory and thrombotic markers such as plasminogen activator inhibitor 1.

Arrhythmias are also more likely to occur in a diurnal pattern. Atrial fibrillation (AF), particularly paroxysmal AF, is believed to be vagally mediated in 10% to 25% of patients.5 Therefore, for those who are predisposed, sleep may represent a period of increased risk for AF. In a study of individuals 60 years and older, the maximum duration and peak frequency of AF occurred from midnight to 2 AM.5

Recent studies have found that REM-related obstructive sleep apnea (OSA) is associated with increased cardiovascular risk. Experimental models show that REM sleep may increase the risk for compromised coronary blood flow.6 Increased heart rate corresponds to reduced coronary blood flow and thus, to decreased coronary perfusion time and less time for relaxation of the heart, increasing the risk for coronary artery disease, thrombosis, and ischemia.

SLEEP APNEA PATHOPHYSIOLOGY

The normal physiology of the sleep-heart inter­action is disrupted by sleep apnea. OSA is defined as episodes of complete or partial airway obstruction that occur during sleep with thoraco­abdominal effort. Central sleep apnea (CSA) is the cessation of breathing with no thoracoabdominal effort. The pathophysiology of the sleep-heart interaction varies for OSA and CSA.

Obstructive sleep apnea

OSA is a nocturnal physiologic stressor that is highly prevalent and underrecognized. It affects approximately 17% of the adult population, and the prevalence is increasing with the obesity epidemic. Nearly 1 in 15 individuals is estimated to be affected by at least moderate OSA.7,8 OSA is underdiagnosed particularly in minority populations.9 Data from the 2015 Multi-Ethnic Study of Atherosclerosis (MESA) showed undiagnosed moderate to severe sleep apnea in 84% to 93% of individuals,9 similar to an estimated 85% of undiagnosed cases in 2002.10

OSA is highly prevalent in individuals with underlying coronary disease11–13 and in those with cardiovascular risk factors such as diabetes, hypertension, and heart failure. The prevalence of OSA in patients with cardiovascular disease ranges from 30% (hypertension) to 60% (stroke or transient ischemic attack, arrhythmia, end-stage renal disease).14

 

 

Pathophysiology of OSA

mehra_sleepapneaandtheheart_f1.jpg
%3Cp%3EFigure%201.%20A%20polysomnogram%20showing%20autonomic%20nervous%20system%20disturbances%2C%20intermittent%20hypoxia%2C%20and%20intrathoracic%20pressure%20alterations.%3C%2Fp%3E
The pathophysiology of OSA can be observed during polysomnography, characterized by autonomic nervous system disturbances, intermittent hypoxia, and intrathoracic pressure alterations, (Figure 1). Intermittent bouts of hypoxia or oxygen-lowering occur because airflow is obstructed despite persistent thoracic and abdominal effort. Systemic inflammation and oxidative stress occur due to these intrathoracic pressure alterations, increased CO2 and reduced oxygen levels, and autonomic nervous system disturbances.

The alterations in sympathetic activation that occur during sleep in patients with OSA persist during wakefulness. Microneurographic recording of sympathetic nerve activity in the peroneal nerve reveal that the rate of sympathetic bursts doubles and the amplitude is greater in individuals with OSA compared with a control group.15

Sympathetic nerve activity, blood pressure, and heart rate were shown to increase during REM sleep in individuals with OSA on continuous positive airway pressure (CPAP) during an induced apneic event (pressure reduction from 8 cm to 6 cm of water).15

During OSA episodes, there is an increased cardiac load. Impaired diastolic function and atrial and aortic enlargement, and in particular, the thin-walled atria are very susceptible to the intra­thoracic pressure swings caused by OSA. Physiologic changes with OSA from pressure changes in the chest result in shift of the intraventricular septum, causing a reduction in cardiac output.16 With the lowering of oxygen during episodes of apnea, constriction of the pulmonary vasculature leads to elevation of pressure in the pulmonary vasculature reflected by the increase in mean pulmonary arterial pressures.17

Other studies have shown that OSA increases upregulation of markers of systemic inflammation and prothrombotic markers, the very markers that can increase cardiovascular or atherogenic risk.18–22 One example is the soluble interleukin 6 receptor, shown to be elevated in the morning relative to sleep apnea compared with the evening.20 Other biomarkers observed to be associated with sleep apnea include markers of prothrombotic potentials such as plasminogen activator inhibitor 1.19 Oxidative stress occurs because intermittent bouts of lower oxygen can lead to oxidation of serum proteins and lipids. Endothelial dysfunction has been observed as well as insulin resistance and dyslipidemia.23 Taken together, these are pathways that lead to atherogenesis and increased cardiovascular risk.

Central sleep apnea

CSA episodes are the cessation of breathing without thoracoabdominal effort, in contrast to the persistence of thoracoabdominal effort in OSA. CSA is characterized by breathing instability with highly sensitive chemoresponses and prolonged circulation time.24 This can be physiologic in some cases, as when it occurs after a very large breath or sigh and then a central apnea event occurs after the sigh. The alterations in oxygen and CO2 and the stretch of the receptors in the alveoli of the lungs initiate the Hering-Breuer inhalation reflex.

Pathophysiology of CSA

Complex pathways of medullary and aortic receptor chemosensitivity are at the root of the pathophysiology of CSA.24 With CSA there is often a relative state of hypocapnia at baseline. During sleep, there is reduction in drive, thus chemo­sensitivity can be activated so that central apnea episodes can ensue as a result of alterations in CO2 (ie, hypocapnia). Another factor that can contribute to the pathophysiology of CSA is arousal from sleep that can reduce CO2 levels and therefore perpetuate central events.

The concept of loop gain is used to understand the pathophysiology of CSA. Loop gain is a measure of the relative stability of a ventilation system and indicates the likelihood of an individual to have periodic breathing. It is calculated by the response to a disturbance divided by the disturbance itself.25 With a high loop gain, there is a more pronounced or exuberant response to the disturbance, indicating more instability in the system and increasing the tendency for irregular breathing and CSA episodes.

Hunter-Cheyne-Stokes respiration occurs with CSA and is characterized by cyclical crescendo-decrescendo respiratory effort that occurs during wakefulness and sleep without upper-airway obstruction.26,27 Unlike OSA, which is worse during REM sleep, Hunter-Cheyne-Stokes breathing in CSA is typically worse in NREM sleep, during N1 and N2 in particular.

 

 

SLEEP APNEA AND HEART FAILURE

Both OSA and CSA are prevalent in patients with heart failure and may be associated with the progression of heart failure. CSA often occurs in patients with heart failure. The pathophysiology is multi­factorial, including pulmonary congestion that results in stretch of the J receptors in the alveoli, prolonged circulation time, and increased chemosensitivity.

Complex pathways in the neuroaxis or somnogenic biomarkers of inflammation or both may be implicated in the paradoxical lack of subjective sleepiness in the presence of increased objective measures of sleepiness in systolic heart failure. One study found a relationship with one biomarker of inflammation and oxidative stress as it relates to objective symptoms of sleepiness but not subjective symptoms of sleepiness.28

Another contributing factor in the relationship between OSA and CSA in heart failure has also been described related to rostral shifts in fluid to the neck and to the pulmonary receptors in the alveoli of the lungs.29 These rostral shifts in fluids may contribute to sleep apnea with parapharyngeal edema leading to OSA and pulmonary congestion leading to CSA.

Sleep apnea is associated with increased post-discharge mortality and hospitalization readmissions in the setting of acute heart failure.30 Mortality analysis of 1,096 patients admitted for decompensated heart failure found CSA and OSA were independently associated with mortality in patients compared with patients with no or minimal sleep-disordered breathing.30

CSA has also been shown to be a predictor of readmission in patients admitted for heart failure exacerbations.31 Targeting underlying CSA may reduce readmissions in those admitted with acute decompensated heart failure. While men were identified to be at increased risk of death relative to sleep-disordered breathing based on the initial results of the Sleep Heart Health Study, a subsequent epidemiologic substudy reflective of an older age group showed that OSA was more strongly associated with left ventricular mass index, risk of heart failure, or death in women compared with men.32

Treatment

Standard therapy for treatment of OSA is CPAP. Adaptive servo-ventilation (ASV) and transvenous phrenic nerve stimulation are also available as treatment options in certain cases of CSA.

One of the first randomized controlled trials designed to assess the impact of CSA treatment on survival in patients with heart failure initially favored the control group then later the CPAP group and was terminated early based on stopping rules.33,34 While adherence to therapy was suboptimal at an average of 3.6 hours, post hoc analysis showed that patients with CSA using CPAP with effective suppression of CSA had improved survival compared with patients who did not have effective suppression using CPAP.34

ASV is mainly used for treatment of CSA. In ASV, positive airway pressure for ventilation support is provided and adjusts as apneic episodes are detected during sleep. The support provided adapts to the physiology of the patient and can deliver breaths and utilize anticyclic modes of ventilation to address crescendo-decrescendo breathing patterns observed in Hunter-Cheyne-Stokes respiration.

In the Treatment of Sleep-Disordered Breathing With Predominant Central Sleep Apnea by Adaptive Servo Ventilation in Patients With Heart Failure (SERVE-HF) trial, 1,300 patients with systolic heart failure and predominantly CSA were randomized to receive ASV vs solely standard medical management.35 The primary composite end point included all-cause mortality or unplanned admission or hospitalization for heart failure. No difference was found in the primary end point between the ASV and the control group; however, there was an unanticipated negative impact of ASV on cardiovascular outcomes in some secondary end points. Based on the secondary outcome of cardiovascular-specific mortality, clinicians were advised that ASV was contraindicated for the treatment of CSA in patients with symptomatic heart failure with a left ventricular ejection fraction less than 45%. The interpretation of this study was complicated by several methodologic limitations.36

The Cardiovascular Improvements With Minute Ventilation-Targeted Adaptive Servo-Ventilation Therapy in Heart Failure (CAT-HF) randomized controlled trial also evaluated ASV compared with standard medical management in 126 patients with heart failure.37 This trial was terminated early because of the results of the SERVE-HF trial. Compliance with therapy was suboptimal at an average of 2.7 hours per day. The composite end point did not differ between the 2 groups; however, this was likely because the study was underpowered and was terminated early. Subgroup analysis revealed that patients with heart failure with preserved ejection fraction may benefit from ASV; however, additional studies are needed to confirm these findings.

Therefore, although ASV is not indicated when there is predominantly CSA in patients with systolic heart failure, preliminary results support potential benefit in patients with OSA and preserved ejection fraction.

Another novel treatment for CSA is transvenous phrenic nerve stimulation. A device is implanted that stimulates the phrenic nerve to initiate breaths. The initial study of trans­venous phrenic nerve stimulation reported a significant reduction in the number of episodes of central apnea per hour of sleep.38,39 The apnea–hypopnea index improved overall and some types of obstructive apneic events were reduced with transvenous phrenic nerve stimulation.

A multicenter randomized control trial of trans­venous phrenic nerve stimulation found improvement in several sleep apnea indices, including central apnea, hypoxia, reduced arousals from sleep, and patient reported well-being.40 Transvenous phrenic nerve stimulation holds promise as a novel therapy for central predominant sleep apnea not only in terms of improving the degree of central apnea and sleep-disordered breathing, but also in improving functional outcomes. Longitudinal and intereventional trial data are needed to clarify the impact of transvenous phrenic nerve stimulation on long-term cardiac outcomes.

SLEEP APNEA AND ATRIAL FIBRILLATION AND STROKE

Atrial fibrillation

AF is the most common sustained cardiac arrhythmia. The number of Americans with AF is projected to increase from 2.3 million to more than 10 million by the year 2050.41 The increasing incidence and prevalence of AF is not fully explained by the aging population and established risk factors.42 Unrecognized sleep apnea, estimated to exist in 85% or more of the population, may partially account for the increasing incidence of AF.43

There are 3 types of AF, which are thought to follow a continuum: paroxysmal AF is characterized by episodes that occur intermittently; persistent AF is characterized by episodes that last longer than 7 days; chronic or permanent AF is typically characterized by AF that is ongoing over many years.44 As with sleep apnea, AF is often asymptomatic and is likely underdiagnosed.

Sleep apnea and AF share several risk factors. Obesity is a risk factor for both OSA and AF; however, a meta-analysis supported a stronger association of OSA and AF vs obesity and AF.45 Increasing age is a risk factor for both OSA and AF.46,47 Although white populations are at higher risk for AF, OSA is associated with a 58% increased risk of AF in African Americans.48 Nocturnal hypoxia has been associated with increased risk of AF in Asians.49

mehra_sleepapneaandtheheart_f2.jpg
%3Cp%3EFigure%202.%20Pathophysiologic%20pathways%20of%20obstructive%20sleep%20apnea%20and%20cardiac%20arrhythmia.%3C%2Fp%3E
In terms of pathophysiology of sleep apnea and cardiac arrhythmia, OSA increases inflammation, intrathoracic pressures, and CO2 levels. The increase in inflammation and oxidative stress is thought to alter the cardiac electrophysiology of the heart and contribute to structural remodeling of the heart that increases the risk of cardiac arrhythmia (Figure 2).50

Experimental data continue to accrue providing biologic plausibility of the relationship between sleep apnea and AF. OSA contributes to structural and electrical remodeling of the heart with evidence supporting increased fibrosis and electrical remodeling in patients with OSA compared with a control group.51 Markers of structural remodeling, such as atrial size, electrical silence, and atrial voltage conduction velocity, are altered in OSA.50

Data from the Sleep Heart Health Study show very strong associations between atrial and ventricular cardiac arrhythmias and sleep apnea with two- to fivefold higher odds of arrhythmias in patients with severe OSA compared with controls even after accounting for confounding factors such as obesity.52

A multicenter, epidemiological study of older men showed that increasing severity of sleep apnea corresponds with an increased prevalence of AF and ventricular ectopy.53 This graded dose-response relationship suggests a causal relationship between sleep apnea and AF and ventricular ectopy. There also appears to be an immediate influence of apneic events and hypopneic events as it relates to arrhythmia. A case-crossover study showed an associated 18-fold increased risk of nocturnal arrhythmia within 90 seconds of an apneic or hypopneic event.54 This association was found with paroxysms of AF and with episodes of nonsustained ventricular tachycardia.

Data from a clinic-based cohort study show an association between AF and OSA.55 Specifically, increased severity of sleep apnea was associated with an increased prevalence of AF. Increasing degree of hypoxia or oxygen-lowering was also associated with increased incidence of AF or newly identified AF identified over time.

Longitudinal examination of 2 epidemiologic studies, the Sleep Heart Health Study and Outcomes of Sleep Disorders Study in Older Men, found CSA to be predictive of AF with a two- to threefold higher odds of developing incident AF as it related to baseline CSA.56 According to these data, CSA may pose a greater risk for development of AF than OSA.

With respect to AF after cardiac surgery, patients with sleep apnea and obesity appear to be at higher risk for developing AF as measured by the apnea–hypopnea index and oxygen desaturation index.57

Treatment of sleep apnea may improve arrhythmic burden. Case-based studies have shown reduced burden and resolution of baseline arrhythmia with CPAP treatment for OSA as therapeutic pressure was achieved.58 Another case-based study involved an individual with snoring and OSA and AF at baseline.59 Several retrospective studies have shown that treatment of OSA after ablation and after cardio­version results in reduced recurrence of AF; however, large definitive clinical trials are lacking.

Stroke

Sleep apnea is a risk factor for stroke due to intermittent hypoxia-mediated elevation of oxidative stress and systemic inflammation, hypercoaguability, and impairment of cerebral autoregulation.60 However, the relationship may be bidirectional in that stroke may be a risk factor for sleep apnea in the post-stroke period. The prevalence of sleep apnea post-stroke has been reported to be up to 70%. CSA can occur in up to 26% during the post-stroke phase.61 Data are inconsistent in terms of the location and size of stroke and the risk of sleep apnea, though cerebrovascular neuronal damage to the brainstem and cortical areas are evident.62 In one study, the incidence of stroke appeared to increase with the severity of sleep apnea.63 These findings were more pronounced in men than in women; however, this study may not have captured the increased cardiovascular risk in postmenopausal women. The Outcomes of Sleep Disorders in Older Men study found that severe hypoxia increased the incidence of stroke, and that hypoxia may be a predictor of newly diagnosed stroke in older men.64 Although definitive clinical trials are underway, post-hoc propensity-score matched analysis from the Sleep Apnea Cardiovascular Endpoints (SAVE) study showed a lower stroke risk in those adherent to CPAP compared with the control group (HR=0.56, 95% CI: 0.30-0.90).65

 

 

SLEEP APNEA, CORONARY ARTERY DISEASE, AND CARIOVASCULAR MORTALITY

The association between sleep apnea and coronary artery disease and cardiovascular mortality was considered in a Spanish study of 1,500 patients followed for 10 years, which reported that CPAP therapy reduced cardiac events in patients with OSA.66 Patients with sleep apnea had an increased risk of fatal myocardial infarction or stroke. Survival of patients treated for sleep apnea approached that of patients without OSA.

In a study of a racially diverse cohort, an association of physician diagnosed sleep apnea with cardiovascular events and survival was identified.67 Diagnosed sleep apnea was estimated to confer a two- to threefold increase in various cardiovascular outcomes and all-cause mortality.

mehra_sleepapneaandtheheart_f3.jpg
%3Cp%3EFigure%203.%20Survival%20of%20patients%20with%20obstructive%20sleep%20apnea%20by%20apnea%E2%80%93hypopnea%20index.%3C%2Fp%3E
All-cause mortality data from the Sleep Heart Health Study of more than 6,000 participants showed that progressive worsening of OSA as defined by the apnea–hypopnea index resulted in poorer survival even after accounting for confounding factors (Figure 3).68 Decreased survival appeared to mostly affect men or patients under age 70.

mehra_sleepapneaandtheheart_t1.jpg
The diurnal pattern of cardiovascular physiology as it relates to sleep is thought to be cardioprotective because of reductions in blood pressure and heart rate. However, in the case of OSA, there appears to be a nocturnal vulnerability or predilection for sudden cardiac death. Patients with OSA were shown to have a higher risk of sudden nocturnal cardiac death occurring from midnight to 6 am compared with individuals without OSA and the general population (Table 1).69

The effect of treatment for sleep apnea on cardiovascular outcomes was the focus of a recent randomized controlled trial of nearly 3,000 participants with a mean follow-up of 4 years.65 Use of CPAP compared with usual care found no difference in cardiovascular outcomes. However, secondary analysis revealed a possible benefit of a lower risk of stroke with use of CPAP therapy. Several factors should be considered in interpreting these findings: ie, low adherence with CPAP therapy (3 hours), whether the study was sufficiently powered to detect a change in cardiovascular outcomes, and if the duration of follow-up was adequate. In terms of patient demographics and study generalizability, the study did not include patients with severe sleep apnea and hypoxia, and most participants were men, of Asian descent, with a mean body mass index of 28 kg/m2, and low levels of sleepiness at baseline.

SLEEP AND CARDIOVASCULAR PHYSIOLOGY

Wakefullness and sleep, the latter comprised of non-rapid eye movement (NREM) sleep and rapid eye movement (REM) sleep, comprise our primary states of being. Sleep states oscillate between NREM and REM sleep. The first and shortest period of REM sleep typically occurs 90 to 120 minutes into the sleep cycle. Most REM sleep, including the longest period of REM sleep, occurs during the latter part of the sleep cycle.

With these sleep state changes, physiologic changes also occur, such as reduced heart rate and blood pressure because of enhanced parasympathetic tone. During REM sleep, there are also intermittent sympathetic nervous system surges. Other physiologic changes include a regular respiratory rate during NREM sleep and an irregular respiratory rate during REM sleep. Body temperature is normal during NREM sleep and poikilothermic (ie, tends to flucuate) during REM sleep. Blood pressure is reduced 10% to 15% during sleep1 and then rises, so that the highest blood pressure occurs in the morning. Data from 10 million users of activity-monitoring devices show that the heart rate changes during sleep.2 The heart rate is decreased in those who get less than 7 hours of sleep, then increases with longer sleep duration in a U-shaped distribution.

Cardiovascular events are more likely to occur at certain times of day. Myocardial infarction is more likely in the morning, with a threefold increased risk within the first 3 hours of awakening that peaks around 9 AM.3,4 Similar diurnal patterns have been observed with other cardiovascular conditions such as sudden cardiac death and ischemic episodes, with the highest risk during morning hours (6 to 9 AM).4

The reason for this morning predisposition for cardio­vascular events is unclear, but it is thought that perhaps the autonomic fluctuations that occur during REM sleep and the predominance of REM sleep in early morning may be a factor. Diurnal changes in blood pressure and cortisol levels may also contribute, as well as levels of systemic inflammatory and thrombotic markers such as plasminogen activator inhibitor 1.

Arrhythmias are also more likely to occur in a diurnal pattern. Atrial fibrillation (AF), particularly paroxysmal AF, is believed to be vagally mediated in 10% to 25% of patients.5 Therefore, for those who are predisposed, sleep may represent a period of increased risk for AF. In a study of individuals 60 years and older, the maximum duration and peak frequency of AF occurred from midnight to 2 AM.5

Recent studies have found that REM-related obstructive sleep apnea (OSA) is associated with increased cardiovascular risk. Experimental models show that REM sleep may increase the risk for compromised coronary blood flow.6 Increased heart rate corresponds to reduced coronary blood flow and thus, to decreased coronary perfusion time and less time for relaxation of the heart, increasing the risk for coronary artery disease, thrombosis, and ischemia.

SLEEP APNEA PATHOPHYSIOLOGY

The normal physiology of the sleep-heart inter­action is disrupted by sleep apnea. OSA is defined as episodes of complete or partial airway obstruction that occur during sleep with thoraco­abdominal effort. Central sleep apnea (CSA) is the cessation of breathing with no thoracoabdominal effort. The pathophysiology of the sleep-heart interaction varies for OSA and CSA.

Obstructive sleep apnea

OSA is a nocturnal physiologic stressor that is highly prevalent and underrecognized. It affects approximately 17% of the adult population, and the prevalence is increasing with the obesity epidemic. Nearly 1 in 15 individuals is estimated to be affected by at least moderate OSA.7,8 OSA is underdiagnosed particularly in minority populations.9 Data from the 2015 Multi-Ethnic Study of Atherosclerosis (MESA) showed undiagnosed moderate to severe sleep apnea in 84% to 93% of individuals,9 similar to an estimated 85% of undiagnosed cases in 2002.10

OSA is highly prevalent in individuals with underlying coronary disease11–13 and in those with cardiovascular risk factors such as diabetes, hypertension, and heart failure. The prevalence of OSA in patients with cardiovascular disease ranges from 30% (hypertension) to 60% (stroke or transient ischemic attack, arrhythmia, end-stage renal disease).14

 

 

Pathophysiology of OSA

mehra_sleepapneaandtheheart_f1.jpg
%3Cp%3EFigure%201.%20A%20polysomnogram%20showing%20autonomic%20nervous%20system%20disturbances%2C%20intermittent%20hypoxia%2C%20and%20intrathoracic%20pressure%20alterations.%3C%2Fp%3E
The pathophysiology of OSA can be observed during polysomnography, characterized by autonomic nervous system disturbances, intermittent hypoxia, and intrathoracic pressure alterations, (Figure 1). Intermittent bouts of hypoxia or oxygen-lowering occur because airflow is obstructed despite persistent thoracic and abdominal effort. Systemic inflammation and oxidative stress occur due to these intrathoracic pressure alterations, increased CO2 and reduced oxygen levels, and autonomic nervous system disturbances.

The alterations in sympathetic activation that occur during sleep in patients with OSA persist during wakefulness. Microneurographic recording of sympathetic nerve activity in the peroneal nerve reveal that the rate of sympathetic bursts doubles and the amplitude is greater in individuals with OSA compared with a control group.15

Sympathetic nerve activity, blood pressure, and heart rate were shown to increase during REM sleep in individuals with OSA on continuous positive airway pressure (CPAP) during an induced apneic event (pressure reduction from 8 cm to 6 cm of water).15

During OSA episodes, there is an increased cardiac load. Impaired diastolic function and atrial and aortic enlargement, and in particular, the thin-walled atria are very susceptible to the intra­thoracic pressure swings caused by OSA. Physiologic changes with OSA from pressure changes in the chest result in shift of the intraventricular septum, causing a reduction in cardiac output.16 With the lowering of oxygen during episodes of apnea, constriction of the pulmonary vasculature leads to elevation of pressure in the pulmonary vasculature reflected by the increase in mean pulmonary arterial pressures.17

Other studies have shown that OSA increases upregulation of markers of systemic inflammation and prothrombotic markers, the very markers that can increase cardiovascular or atherogenic risk.18–22 One example is the soluble interleukin 6 receptor, shown to be elevated in the morning relative to sleep apnea compared with the evening.20 Other biomarkers observed to be associated with sleep apnea include markers of prothrombotic potentials such as plasminogen activator inhibitor 1.19 Oxidative stress occurs because intermittent bouts of lower oxygen can lead to oxidation of serum proteins and lipids. Endothelial dysfunction has been observed as well as insulin resistance and dyslipidemia.23 Taken together, these are pathways that lead to atherogenesis and increased cardiovascular risk.

Central sleep apnea

CSA episodes are the cessation of breathing without thoracoabdominal effort, in contrast to the persistence of thoracoabdominal effort in OSA. CSA is characterized by breathing instability with highly sensitive chemoresponses and prolonged circulation time.24 This can be physiologic in some cases, as when it occurs after a very large breath or sigh and then a central apnea event occurs after the sigh. The alterations in oxygen and CO2 and the stretch of the receptors in the alveoli of the lungs initiate the Hering-Breuer inhalation reflex.

Pathophysiology of CSA

Complex pathways of medullary and aortic receptor chemosensitivity are at the root of the pathophysiology of CSA.24 With CSA there is often a relative state of hypocapnia at baseline. During sleep, there is reduction in drive, thus chemo­sensitivity can be activated so that central apnea episodes can ensue as a result of alterations in CO2 (ie, hypocapnia). Another factor that can contribute to the pathophysiology of CSA is arousal from sleep that can reduce CO2 levels and therefore perpetuate central events.

The concept of loop gain is used to understand the pathophysiology of CSA. Loop gain is a measure of the relative stability of a ventilation system and indicates the likelihood of an individual to have periodic breathing. It is calculated by the response to a disturbance divided by the disturbance itself.25 With a high loop gain, there is a more pronounced or exuberant response to the disturbance, indicating more instability in the system and increasing the tendency for irregular breathing and CSA episodes.

Hunter-Cheyne-Stokes respiration occurs with CSA and is characterized by cyclical crescendo-decrescendo respiratory effort that occurs during wakefulness and sleep without upper-airway obstruction.26,27 Unlike OSA, which is worse during REM sleep, Hunter-Cheyne-Stokes breathing in CSA is typically worse in NREM sleep, during N1 and N2 in particular.

 

 

SLEEP APNEA AND HEART FAILURE

Both OSA and CSA are prevalent in patients with heart failure and may be associated with the progression of heart failure. CSA often occurs in patients with heart failure. The pathophysiology is multi­factorial, including pulmonary congestion that results in stretch of the J receptors in the alveoli, prolonged circulation time, and increased chemosensitivity.

Complex pathways in the neuroaxis or somnogenic biomarkers of inflammation or both may be implicated in the paradoxical lack of subjective sleepiness in the presence of increased objective measures of sleepiness in systolic heart failure. One study found a relationship with one biomarker of inflammation and oxidative stress as it relates to objective symptoms of sleepiness but not subjective symptoms of sleepiness.28

Another contributing factor in the relationship between OSA and CSA in heart failure has also been described related to rostral shifts in fluid to the neck and to the pulmonary receptors in the alveoli of the lungs.29 These rostral shifts in fluids may contribute to sleep apnea with parapharyngeal edema leading to OSA and pulmonary congestion leading to CSA.

Sleep apnea is associated with increased post-discharge mortality and hospitalization readmissions in the setting of acute heart failure.30 Mortality analysis of 1,096 patients admitted for decompensated heart failure found CSA and OSA were independently associated with mortality in patients compared with patients with no or minimal sleep-disordered breathing.30

CSA has also been shown to be a predictor of readmission in patients admitted for heart failure exacerbations.31 Targeting underlying CSA may reduce readmissions in those admitted with acute decompensated heart failure. While men were identified to be at increased risk of death relative to sleep-disordered breathing based on the initial results of the Sleep Heart Health Study, a subsequent epidemiologic substudy reflective of an older age group showed that OSA was more strongly associated with left ventricular mass index, risk of heart failure, or death in women compared with men.32

Treatment

Standard therapy for treatment of OSA is CPAP. Adaptive servo-ventilation (ASV) and transvenous phrenic nerve stimulation are also available as treatment options in certain cases of CSA.

One of the first randomized controlled trials designed to assess the impact of CSA treatment on survival in patients with heart failure initially favored the control group then later the CPAP group and was terminated early based on stopping rules.33,34 While adherence to therapy was suboptimal at an average of 3.6 hours, post hoc analysis showed that patients with CSA using CPAP with effective suppression of CSA had improved survival compared with patients who did not have effective suppression using CPAP.34

ASV is mainly used for treatment of CSA. In ASV, positive airway pressure for ventilation support is provided and adjusts as apneic episodes are detected during sleep. The support provided adapts to the physiology of the patient and can deliver breaths and utilize anticyclic modes of ventilation to address crescendo-decrescendo breathing patterns observed in Hunter-Cheyne-Stokes respiration.

In the Treatment of Sleep-Disordered Breathing With Predominant Central Sleep Apnea by Adaptive Servo Ventilation in Patients With Heart Failure (SERVE-HF) trial, 1,300 patients with systolic heart failure and predominantly CSA were randomized to receive ASV vs solely standard medical management.35 The primary composite end point included all-cause mortality or unplanned admission or hospitalization for heart failure. No difference was found in the primary end point between the ASV and the control group; however, there was an unanticipated negative impact of ASV on cardiovascular outcomes in some secondary end points. Based on the secondary outcome of cardiovascular-specific mortality, clinicians were advised that ASV was contraindicated for the treatment of CSA in patients with symptomatic heart failure with a left ventricular ejection fraction less than 45%. The interpretation of this study was complicated by several methodologic limitations.36

The Cardiovascular Improvements With Minute Ventilation-Targeted Adaptive Servo-Ventilation Therapy in Heart Failure (CAT-HF) randomized controlled trial also evaluated ASV compared with standard medical management in 126 patients with heart failure.37 This trial was terminated early because of the results of the SERVE-HF trial. Compliance with therapy was suboptimal at an average of 2.7 hours per day. The composite end point did not differ between the 2 groups; however, this was likely because the study was underpowered and was terminated early. Subgroup analysis revealed that patients with heart failure with preserved ejection fraction may benefit from ASV; however, additional studies are needed to confirm these findings.

Therefore, although ASV is not indicated when there is predominantly CSA in patients with systolic heart failure, preliminary results support potential benefit in patients with OSA and preserved ejection fraction.

Another novel treatment for CSA is transvenous phrenic nerve stimulation. A device is implanted that stimulates the phrenic nerve to initiate breaths. The initial study of trans­venous phrenic nerve stimulation reported a significant reduction in the number of episodes of central apnea per hour of sleep.38,39 The apnea–hypopnea index improved overall and some types of obstructive apneic events were reduced with transvenous phrenic nerve stimulation.

A multicenter randomized control trial of trans­venous phrenic nerve stimulation found improvement in several sleep apnea indices, including central apnea, hypoxia, reduced arousals from sleep, and patient reported well-being.40 Transvenous phrenic nerve stimulation holds promise as a novel therapy for central predominant sleep apnea not only in terms of improving the degree of central apnea and sleep-disordered breathing, but also in improving functional outcomes. Longitudinal and intereventional trial data are needed to clarify the impact of transvenous phrenic nerve stimulation on long-term cardiac outcomes.

SLEEP APNEA AND ATRIAL FIBRILLATION AND STROKE

Atrial fibrillation

AF is the most common sustained cardiac arrhythmia. The number of Americans with AF is projected to increase from 2.3 million to more than 10 million by the year 2050.41 The increasing incidence and prevalence of AF is not fully explained by the aging population and established risk factors.42 Unrecognized sleep apnea, estimated to exist in 85% or more of the population, may partially account for the increasing incidence of AF.43

There are 3 types of AF, which are thought to follow a continuum: paroxysmal AF is characterized by episodes that occur intermittently; persistent AF is characterized by episodes that last longer than 7 days; chronic or permanent AF is typically characterized by AF that is ongoing over many years.44 As with sleep apnea, AF is often asymptomatic and is likely underdiagnosed.

Sleep apnea and AF share several risk factors. Obesity is a risk factor for both OSA and AF; however, a meta-analysis supported a stronger association of OSA and AF vs obesity and AF.45 Increasing age is a risk factor for both OSA and AF.46,47 Although white populations are at higher risk for AF, OSA is associated with a 58% increased risk of AF in African Americans.48 Nocturnal hypoxia has been associated with increased risk of AF in Asians.49

mehra_sleepapneaandtheheart_f2.jpg
%3Cp%3EFigure%202.%20Pathophysiologic%20pathways%20of%20obstructive%20sleep%20apnea%20and%20cardiac%20arrhythmia.%3C%2Fp%3E
In terms of pathophysiology of sleep apnea and cardiac arrhythmia, OSA increases inflammation, intrathoracic pressures, and CO2 levels. The increase in inflammation and oxidative stress is thought to alter the cardiac electrophysiology of the heart and contribute to structural remodeling of the heart that increases the risk of cardiac arrhythmia (Figure 2).50

Experimental data continue to accrue providing biologic plausibility of the relationship between sleep apnea and AF. OSA contributes to structural and electrical remodeling of the heart with evidence supporting increased fibrosis and electrical remodeling in patients with OSA compared with a control group.51 Markers of structural remodeling, such as atrial size, electrical silence, and atrial voltage conduction velocity, are altered in OSA.50

Data from the Sleep Heart Health Study show very strong associations between atrial and ventricular cardiac arrhythmias and sleep apnea with two- to fivefold higher odds of arrhythmias in patients with severe OSA compared with controls even after accounting for confounding factors such as obesity.52

A multicenter, epidemiological study of older men showed that increasing severity of sleep apnea corresponds with an increased prevalence of AF and ventricular ectopy.53 This graded dose-response relationship suggests a causal relationship between sleep apnea and AF and ventricular ectopy. There also appears to be an immediate influence of apneic events and hypopneic events as it relates to arrhythmia. A case-crossover study showed an associated 18-fold increased risk of nocturnal arrhythmia within 90 seconds of an apneic or hypopneic event.54 This association was found with paroxysms of AF and with episodes of nonsustained ventricular tachycardia.

Data from a clinic-based cohort study show an association between AF and OSA.55 Specifically, increased severity of sleep apnea was associated with an increased prevalence of AF. Increasing degree of hypoxia or oxygen-lowering was also associated with increased incidence of AF or newly identified AF identified over time.

Longitudinal examination of 2 epidemiologic studies, the Sleep Heart Health Study and Outcomes of Sleep Disorders Study in Older Men, found CSA to be predictive of AF with a two- to threefold higher odds of developing incident AF as it related to baseline CSA.56 According to these data, CSA may pose a greater risk for development of AF than OSA.

With respect to AF after cardiac surgery, patients with sleep apnea and obesity appear to be at higher risk for developing AF as measured by the apnea–hypopnea index and oxygen desaturation index.57

Treatment of sleep apnea may improve arrhythmic burden. Case-based studies have shown reduced burden and resolution of baseline arrhythmia with CPAP treatment for OSA as therapeutic pressure was achieved.58 Another case-based study involved an individual with snoring and OSA and AF at baseline.59 Several retrospective studies have shown that treatment of OSA after ablation and after cardio­version results in reduced recurrence of AF; however, large definitive clinical trials are lacking.

Stroke

Sleep apnea is a risk factor for stroke due to intermittent hypoxia-mediated elevation of oxidative stress and systemic inflammation, hypercoaguability, and impairment of cerebral autoregulation.60 However, the relationship may be bidirectional in that stroke may be a risk factor for sleep apnea in the post-stroke period. The prevalence of sleep apnea post-stroke has been reported to be up to 70%. CSA can occur in up to 26% during the post-stroke phase.61 Data are inconsistent in terms of the location and size of stroke and the risk of sleep apnea, though cerebrovascular neuronal damage to the brainstem and cortical areas are evident.62 In one study, the incidence of stroke appeared to increase with the severity of sleep apnea.63 These findings were more pronounced in men than in women; however, this study may not have captured the increased cardiovascular risk in postmenopausal women. The Outcomes of Sleep Disorders in Older Men study found that severe hypoxia increased the incidence of stroke, and that hypoxia may be a predictor of newly diagnosed stroke in older men.64 Although definitive clinical trials are underway, post-hoc propensity-score matched analysis from the Sleep Apnea Cardiovascular Endpoints (SAVE) study showed a lower stroke risk in those adherent to CPAP compared with the control group (HR=0.56, 95% CI: 0.30-0.90).65

 

 

SLEEP APNEA, CORONARY ARTERY DISEASE, AND CARIOVASCULAR MORTALITY

The association between sleep apnea and coronary artery disease and cardiovascular mortality was considered in a Spanish study of 1,500 patients followed for 10 years, which reported that CPAP therapy reduced cardiac events in patients with OSA.66 Patients with sleep apnea had an increased risk of fatal myocardial infarction or stroke. Survival of patients treated for sleep apnea approached that of patients without OSA.

In a study of a racially diverse cohort, an association of physician diagnosed sleep apnea with cardiovascular events and survival was identified.67 Diagnosed sleep apnea was estimated to confer a two- to threefold increase in various cardiovascular outcomes and all-cause mortality.

mehra_sleepapneaandtheheart_f3.jpg
%3Cp%3EFigure%203.%20Survival%20of%20patients%20with%20obstructive%20sleep%20apnea%20by%20apnea%E2%80%93hypopnea%20index.%3C%2Fp%3E
All-cause mortality data from the Sleep Heart Health Study of more than 6,000 participants showed that progressive worsening of OSA as defined by the apnea–hypopnea index resulted in poorer survival even after accounting for confounding factors (Figure 3).68 Decreased survival appeared to mostly affect men or patients under age 70.

mehra_sleepapneaandtheheart_t1.jpg
The diurnal pattern of cardiovascular physiology as it relates to sleep is thought to be cardioprotective because of reductions in blood pressure and heart rate. However, in the case of OSA, there appears to be a nocturnal vulnerability or predilection for sudden cardiac death. Patients with OSA were shown to have a higher risk of sudden nocturnal cardiac death occurring from midnight to 6 am compared with individuals without OSA and the general population (Table 1).69

The effect of treatment for sleep apnea on cardiovascular outcomes was the focus of a recent randomized controlled trial of nearly 3,000 participants with a mean follow-up of 4 years.65 Use of CPAP compared with usual care found no difference in cardiovascular outcomes. However, secondary analysis revealed a possible benefit of a lower risk of stroke with use of CPAP therapy. Several factors should be considered in interpreting these findings: ie, low adherence with CPAP therapy (3 hours), whether the study was sufficiently powered to detect a change in cardiovascular outcomes, and if the duration of follow-up was adequate. In terms of patient demographics and study generalizability, the study did not include patients with severe sleep apnea and hypoxia, and most participants were men, of Asian descent, with a mean body mass index of 28 kg/m2, and low levels of sleepiness at baseline.

References
  1. Kario K. Morning surge in blood pressure and cardiovascular risk: evidence and perspectives. Hypertension 2010; 56(5):765–773.
  2. FitBit: 150 billion data hrs shows sleep hours sweet spot, optimal health strategy. True Strange Library website. https://truestrange.com/2018/08/29/fitbit-150-billion-data-hrs-shows-sleep-hours-sweet-spot-optimal-health-strategy. Accessed August 19, 2019.
  3. Muller JE, Stone PH, Turi ZG, et al; MILIS Study Group. Circadian variation in the frequency of onset of acute myocardial infarction. N Engl J Med 1985; 313(21):1315–1322.
  4. Marler JR, Price TR, Clark GL, et al. Morning increase in onset of ischemic stroke. Stroke 1989; 20(4):473–476.
  5. Yamashita T, Murakawa Y, Hayami N, et al. Relation between aging and circadian variation of paroxysmal atrial fibrillation. Am J Cardiol 1998; 82(11):1364–1367.
  6. Kirby DA, Verrier RL. Differential effects of sleep stage on coronary hemodynamic function. Am J Physiol 1989; 256(5 Pt 2):H1378–H1383.
  7. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered breathing among middle-aged adults. N Engl J Med 1993; 328(17):1230–1235.
  8. 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.
  9. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  10. Kapur V, Strohl KP, Redline S, Iber C, O’Connor G, Nieto J. Under­diagnosis of sleep apnea syndrome in U.S. communities. Sleep Breath 2002; 6(2):49–54.
  11. Mooe T, Rabben T, Wiklund U, Franklin KA, Eriksson P. Sleep-disordered breathing in men with coronary artery disease. Chest 1996; 109(3):659–663.
  12. Schäfer H, Koehler U, Ewig S, Hasper E, Tasci S, Lüderitz B. Obstructive sleep apnea as a risk marker in coronary artery disease. Cardiology 1999; 92(2):79–84.
  13. Leung RST, Bradley TD. Sleep apnea and cardiovascular disease. Am J Respir Crit Care Med 2001; 164(12):2147–2165.
  14. Cepeda-Valery B, Acharjee S, Romero-Corral A, Pressman GS, Gami AS. Obstructive sleep apnea and acute coronary syndromes: etiology, risk, and management. Curr Cardiol Rep 2014; 16(10):535.
  15. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive sleep apnea. J Clin Invest 1995; 96(4):1897–1904.
  16. Kasai T, Bradley TD. Obstructive sleep apnea and heart failure: pathophysiologic and therapeutic implications. J Am Coll Cardiol 2011; 57(2):119–127.
  17. Sajkov D, McEvoy RD. Obstructive sleep apnea and pulmonary hypertension. Prog Cardiovasc Dis 2009; 51(5):363–370.
  18. Nadeem R, Molnar J, Madbouly EM, et al. Serum inflammatory markers in obstructive sleep apnea: a meta-analysis. J Clin Sleep Med 2013; 9(10):1003–1012.
  19. Mehra R, Xu F, Babineau DC, et al. Sleep-disordered breathing and prothrombotic biomarkers: cross-sectional results of the Cleveland Family Study. Am J Respir Crit Care Med 2010; 182(6):826–833.
  20. Mehra R, Storfer-Isser A, Kirchner HL, et al. Soluble interleukin 6 receptor: a novel marker of moderate to severe sleep-related breathing disorder. Arch Intern Med 2006; 166(16):1725–1731.
  21. Paz y Mar HL, Hazen SL, Tracy RP, et al. Effect of continuous positive airway pressure on cardiovascular biomarkers: the sleep apnea stress randomized controlled trial. Chest 2016; 150(1):80–90.
  22. Xie X, Pan L, Ren D, Du C, Guo Y. Effects of continuous positive airway pressure therapy on systemic inflammation in obstructive sleep apnea: a meta-analysis. Sleep Med 2013; 14(11):1139–1150.
  23. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005; 352(16):1685–1695.
  24. Eckert DJ, Jordan AS, Merchia P, Malhotra A. Central sleep apnea: pathophysiology and treatment. Chest 2007; 131(2):595–607.
  25. White DP. Pathogenesis of obstructive and central sleep apnea. Am J Respir Crit Care Med 2005; 172(11):1363–1370.
  26. Javaheri S. Heart failure. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th ed. Philadelphia, PA: Elsevier; 2017:1271–1285.
  27. Olson LJ, Somers VK. Treating central sleep apnea in heart failure: outcomes revisited. Circulation 2007; 115(25):3140–3142.
  28. Mehra R, Wang L, Andrews N, et al. Dissociation of objective and subjective daytime sleepiness and biomarkers of systemic inflammation in sleep-disordered breathing and systolic heart failure. J Clin Sleep Med 2017; 13(12):1411–1422.
  29. Kasai T, Floras JS, Bradley TD. Sleep apnea and cardiovascular disease: a bidirectional relationship. Circulation 2012; 126(12):1495–1510.
  30. Khayat R, Jarjoura D, Porter K, et al. Sleep disordered breathing and post-discharge mortality in patients with acute heart failure. Eur Heart J 2015; 36(23):1463–1469.
  31. Khayat R, Abraham W, Patt B, et al. Central sleep apnea is a predictor of cardiac readmission in hospitalized patients with systolic heart failure. J Card Fail 2012; 18(7):534–540.
  32. Roca GQ, Redline S, Claggett B, et al. Sex-specific association of sleep apnea severity with subclinical myocardial injury, ventricular hypertrophy, and heart failure risk in a community-dwelling cohort: the Atherosclerosis Risk in Communities–Sleep Heart Health Study. Circulation 2015; 132(14):1329–1337.
  33. Bradley TD, Logan AG, Kimoff RJ, et al; CANPAP Investigators. Continuous positive airway pressure for central sleep apnea and heart failure. N Engl J Med 2005; 353(19):2025–2033.
  34. Arzt M, Floras JS, Logan AG, et al; CANPAP Investigators. Suppression of central sleep apnea by continuous positive airway pressure and transplant-free survival in heart failure: a post hoc analysis of the Canadian Continuous Positive Airway Pressure for Patients with Central Sleep Apnea and Heart Failure Trial (CANPAP). Circulation 2007; 115(25):3173–3180.
  35. Cowie MR, Woehrle H, Wegscheider K, et al. Adaptive servo-ventilation for central sleep apnea in systolic heart failure. N Engl J Med 2015; 373(12):1095–1105.
  36. Mehra R, Gottlieb DJ. A paradigm shift in the treatment of central sleep apnea in heart failure. Chest 2015; 148(4):848–851.
  37. O’Connor CM, Whellan DJ, Fiuzat M, et al. Cardiovascular outcomes with minute ventilation-targeted adaptive servo-ventilation therapy in heart failure: the CAT-HF trial. J Am Coll Cardiol 2017; 69(12):1577–1587.
  38. Abraham WT, Jagielski D, Oldenburg O, et al; remede Pilot Study Investigators. Phrenic nerve stimulation for the treatment of central sleep apnea. JACC Heart Fail 2015; 3(5):360–369.
  39. Ponikowski P, Javaheri S, Michalkiewicz D, et al. Transvenous phrenic nerve stimulation for the treatment of central sleep apnoea in heart failure. Eur Heart J 2012; 33(7):889–894.
  40. Costanzo MR, Ponikowski P, Javaheri S, et al; remede System Pivotal Trial Study Group. Transvenous neurostimulation for central sleep apnoea: a randomised controlled trial. Lancet 2016; 388(10048):974–982.
  41. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370-2375.
  42. Wolf PA, Benjamin EJ, Belanger AJ, Kannel WB, Levy D, D’Agostino RB. Secular trends in the prevalence of atrial fibrillation: the Framingham Study. Am Heart J 1996; 131(4):790–795.
  43. Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125.
  44. Camm AJ, Kirchhof P, Lip GYH, et al; European Heart Rhythm Association; European Association for Cardio-Thoracic Surgery. Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart J 2010; 31(19):2369–2429.
  45. Trulock KM, Narayan SM, Piccini JP. Rhythm control in heart failure patients with atrial fibrillation: contemporary challenges including the role of ablation. J Am Coll Cardiol 2014; 64(7):710–721.
  46. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  47. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors In Atrial Fibrillation (ATRIA) study. JAMA 2001; 258(18):2370–2375.
  48. Kwon Y, Mehra R. Obstructive sleep apnea and atrial fibrillation: honing in on race-specific susceptibilities. J Clin Sleep Med 2018; 14(9):1459–1461.
  49. Mehra R. Sleep apnea and nocturnal cardiac arrhythmia: understanding differences across ethnicity. J Clin Sleep Med 2017; 13(11):1229–1231.
  50. May AM, Van Wagoner DR, Mehra R. OSA and cardiac arrhymogenesis: mechanistic insights. Chest 2017; 151(1):225–241.
  51. Dimitri H, Ng M, Brooks AG, et al. Atrial remodeling in obstructive sleep apnea: implications for atrial fibrillation. Heart Rhythm 2012; 9(3):321–327.
  52. Mehra R, Benjamin EJ, Shahar E, et al. Association of nocturnal arrhythmias with sleep-disordered breathing: the Sleep Heart Health Study. Am J Respir Crit Care Med 2006; 173(8):910–916.
  53. Mehra R, Stone KL, Varosy PD, et al. Nocturnal arrhythmias across a spectrum of obstructive and central sleep-disordered breathing in older men: outcomes of sleep disorders in older men (MrOS sleep) study. Arch Intern Med 2009; 169(12):1147–1155.
  54. Monahan K, Storfer-Isser A, Mehra R, et al. Triggering of nocturnal arrhythmias by sleep-disordered breathing events. J Am Coll Cardiol 2009; 54(19):1797–1804.
  55. Gami AS, Hodge DO, Herges RM, et al. Obstructive sleep apnea, obesity, and the risk of incident atrial fibrillation. J Am Coll Cardiol 2007; 49(5):565–571.
  56. May AM, Blackwell T, Stone PH, et al; MrOS Sleep (Outcomes of Sleep Disorders in Older Men) Study Group. Am J Respir Crit Care Med 2016; 193(7):783–791.
  57. Kaw R, El Zarif S, Wang L, Bena J, Blackstone EH, Mehra R. Obesity as an effect modifier in sleep-disordered breathing and postcardiac surgery atrial fibrillation. Chest 2017; 151(6):1279–1287.
  58. Walia H, Strohl KP, Mehra R. Effect of continuous positive airway pressure on an atrial arrhythmia in a patient with mild obstructive sleep apnea. J Clin Sleep Med 2011; 7(4):397–398.
  59. Walia HK, Chung MK, Ibrahim S, Mehra R. Positive airway pressure-induced conversion of atrial fibrillation to normal sinus rhythm in severe obstructive sleep apnea. J Clin Sleep Med 2016; 12(9):1301–1303.
  60. Veasey SC, Davis CW, Fenik P, et al. Long-term intermittent hypoxia in mice: protracted hypersomnolence with oxidative injury to sleep-wake brain regions. Sleep 2004; 27(2):194–201.
  61. Parra O, Arboix A, Bechich S, et al. Time course of sleep-related breathing disorders in first-ever stroke or transient ischemic attack. Am J Respir Crit Care Med 2000; 161(2I):375–380.
  62. Song TJ, Park JH, Choi K, et al. Moderate-to-severe obstructive sleep apnea is associated with cerebral small vessel disease. Sleep Med 2017; 30:36–42.
  63. 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.
  64. Stone KL, Blackwell TL, Ancoli-Israel S, et al; Osteoporotic Fractures in Men (MrOS) Study Research Group. Sleep disordered breathing and risk of stroke in older community-dwelling men. Sleep 2016; 39(3):531–540.
  65. McEvoy RD, Antic NA, Heeley E, et al; SAVE Investigators and Coordinators. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med 2016; 375(10):919–931.
  66. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardiovascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  67. Yeboah J, Redline S, Johnson C, et al. Association between sleep apnea, snoring, incident cardiovascular events and all-cause mortality in an adult population: MESA. Atherosclerosis 2011; 219(2):963–968.
  68. Punjabi NM, Caffo BS, Goodwin JL, et al. Sleep-disordered breathing and mortality: a prospective cohort study. PLoS Med 2009; 6(8):e1000132.
  69. Gami AS, Howard DE, Olson EJ, Somers VK. Day–night pattern of sudden death in obstructive sleep apnea. N Engl J Med 2005; 352(12):1206–1214.
References
  1. Kario K. Morning surge in blood pressure and cardiovascular risk: evidence and perspectives. Hypertension 2010; 56(5):765–773.
  2. FitBit: 150 billion data hrs shows sleep hours sweet spot, optimal health strategy. True Strange Library website. https://truestrange.com/2018/08/29/fitbit-150-billion-data-hrs-shows-sleep-hours-sweet-spot-optimal-health-strategy. Accessed August 19, 2019.
  3. Muller JE, Stone PH, Turi ZG, et al; MILIS Study Group. Circadian variation in the frequency of onset of acute myocardial infarction. N Engl J Med 1985; 313(21):1315–1322.
  4. Marler JR, Price TR, Clark GL, et al. Morning increase in onset of ischemic stroke. Stroke 1989; 20(4):473–476.
  5. Yamashita T, Murakawa Y, Hayami N, et al. Relation between aging and circadian variation of paroxysmal atrial fibrillation. Am J Cardiol 1998; 82(11):1364–1367.
  6. Kirby DA, Verrier RL. Differential effects of sleep stage on coronary hemodynamic function. Am J Physiol 1989; 256(5 Pt 2):H1378–H1383.
  7. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered breathing among middle-aged adults. N Engl J Med 1993; 328(17):1230–1235.
  8. 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.
  9. Chen X, Wang R, Zee P, et al. Racial/ethnic differences in sleep disturbances: the Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 2015; 38(6):877–888.
  10. Kapur V, Strohl KP, Redline S, Iber C, O’Connor G, Nieto J. Under­diagnosis of sleep apnea syndrome in U.S. communities. Sleep Breath 2002; 6(2):49–54.
  11. Mooe T, Rabben T, Wiklund U, Franklin KA, Eriksson P. Sleep-disordered breathing in men with coronary artery disease. Chest 1996; 109(3):659–663.
  12. Schäfer H, Koehler U, Ewig S, Hasper E, Tasci S, Lüderitz B. Obstructive sleep apnea as a risk marker in coronary artery disease. Cardiology 1999; 92(2):79–84.
  13. Leung RST, Bradley TD. Sleep apnea and cardiovascular disease. Am J Respir Crit Care Med 2001; 164(12):2147–2165.
  14. Cepeda-Valery B, Acharjee S, Romero-Corral A, Pressman GS, Gami AS. Obstructive sleep apnea and acute coronary syndromes: etiology, risk, and management. Curr Cardiol Rep 2014; 16(10):535.
  15. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive sleep apnea. J Clin Invest 1995; 96(4):1897–1904.
  16. Kasai T, Bradley TD. Obstructive sleep apnea and heart failure: pathophysiologic and therapeutic implications. J Am Coll Cardiol 2011; 57(2):119–127.
  17. Sajkov D, McEvoy RD. Obstructive sleep apnea and pulmonary hypertension. Prog Cardiovasc Dis 2009; 51(5):363–370.
  18. Nadeem R, Molnar J, Madbouly EM, et al. Serum inflammatory markers in obstructive sleep apnea: a meta-analysis. J Clin Sleep Med 2013; 9(10):1003–1012.
  19. Mehra R, Xu F, Babineau DC, et al. Sleep-disordered breathing and prothrombotic biomarkers: cross-sectional results of the Cleveland Family Study. Am J Respir Crit Care Med 2010; 182(6):826–833.
  20. Mehra R, Storfer-Isser A, Kirchner HL, et al. Soluble interleukin 6 receptor: a novel marker of moderate to severe sleep-related breathing disorder. Arch Intern Med 2006; 166(16):1725–1731.
  21. Paz y Mar HL, Hazen SL, Tracy RP, et al. Effect of continuous positive airway pressure on cardiovascular biomarkers: the sleep apnea stress randomized controlled trial. Chest 2016; 150(1):80–90.
  22. Xie X, Pan L, Ren D, Du C, Guo Y. Effects of continuous positive airway pressure therapy on systemic inflammation in obstructive sleep apnea: a meta-analysis. Sleep Med 2013; 14(11):1139–1150.
  23. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005; 352(16):1685–1695.
  24. Eckert DJ, Jordan AS, Merchia P, Malhotra A. Central sleep apnea: pathophysiology and treatment. Chest 2007; 131(2):595–607.
  25. White DP. Pathogenesis of obstructive and central sleep apnea. Am J Respir Crit Care Med 2005; 172(11):1363–1370.
  26. Javaheri S. Heart failure. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th ed. Philadelphia, PA: Elsevier; 2017:1271–1285.
  27. Olson LJ, Somers VK. Treating central sleep apnea in heart failure: outcomes revisited. Circulation 2007; 115(25):3140–3142.
  28. Mehra R, Wang L, Andrews N, et al. Dissociation of objective and subjective daytime sleepiness and biomarkers of systemic inflammation in sleep-disordered breathing and systolic heart failure. J Clin Sleep Med 2017; 13(12):1411–1422.
  29. Kasai T, Floras JS, Bradley TD. Sleep apnea and cardiovascular disease: a bidirectional relationship. Circulation 2012; 126(12):1495–1510.
  30. Khayat R, Jarjoura D, Porter K, et al. Sleep disordered breathing and post-discharge mortality in patients with acute heart failure. Eur Heart J 2015; 36(23):1463–1469.
  31. Khayat R, Abraham W, Patt B, et al. Central sleep apnea is a predictor of cardiac readmission in hospitalized patients with systolic heart failure. J Card Fail 2012; 18(7):534–540.
  32. Roca GQ, Redline S, Claggett B, et al. Sex-specific association of sleep apnea severity with subclinical myocardial injury, ventricular hypertrophy, and heart failure risk in a community-dwelling cohort: the Atherosclerosis Risk in Communities–Sleep Heart Health Study. Circulation 2015; 132(14):1329–1337.
  33. Bradley TD, Logan AG, Kimoff RJ, et al; CANPAP Investigators. Continuous positive airway pressure for central sleep apnea and heart failure. N Engl J Med 2005; 353(19):2025–2033.
  34. Arzt M, Floras JS, Logan AG, et al; CANPAP Investigators. Suppression of central sleep apnea by continuous positive airway pressure and transplant-free survival in heart failure: a post hoc analysis of the Canadian Continuous Positive Airway Pressure for Patients with Central Sleep Apnea and Heart Failure Trial (CANPAP). Circulation 2007; 115(25):3173–3180.
  35. Cowie MR, Woehrle H, Wegscheider K, et al. Adaptive servo-ventilation for central sleep apnea in systolic heart failure. N Engl J Med 2015; 373(12):1095–1105.
  36. Mehra R, Gottlieb DJ. A paradigm shift in the treatment of central sleep apnea in heart failure. Chest 2015; 148(4):848–851.
  37. O’Connor CM, Whellan DJ, Fiuzat M, et al. Cardiovascular outcomes with minute ventilation-targeted adaptive servo-ventilation therapy in heart failure: the CAT-HF trial. J Am Coll Cardiol 2017; 69(12):1577–1587.
  38. Abraham WT, Jagielski D, Oldenburg O, et al; remede Pilot Study Investigators. Phrenic nerve stimulation for the treatment of central sleep apnea. JACC Heart Fail 2015; 3(5):360–369.
  39. Ponikowski P, Javaheri S, Michalkiewicz D, et al. Transvenous phrenic nerve stimulation for the treatment of central sleep apnoea in heart failure. Eur Heart J 2012; 33(7):889–894.
  40. Costanzo MR, Ponikowski P, Javaheri S, et al; remede System Pivotal Trial Study Group. Transvenous neurostimulation for central sleep apnoea: a randomised controlled trial. Lancet 2016; 388(10048):974–982.
  41. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors in Atrial Fibrillation (ATRIA) Study. JAMA 2001; 285(18):2370-2375.
  42. Wolf PA, Benjamin EJ, Belanger AJ, Kannel WB, Levy D, D’Agostino RB. Secular trends in the prevalence of atrial fibrillation: the Framingham Study. Am Heart J 1996; 131(4):790–795.
  43. Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006; 114(2):119–125.
  44. Camm AJ, Kirchhof P, Lip GYH, et al; European Heart Rhythm Association; European Association for Cardio-Thoracic Surgery. Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart J 2010; 31(19):2369–2429.
  45. Trulock KM, Narayan SM, Piccini JP. Rhythm control in heart failure patients with atrial fibrillation: contemporary challenges including the role of ablation. J Am Coll Cardiol 2014; 64(7):710–721.
  46. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  47. Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors In Atrial Fibrillation (ATRIA) study. JAMA 2001; 258(18):2370–2375.
  48. Kwon Y, Mehra R. Obstructive sleep apnea and atrial fibrillation: honing in on race-specific susceptibilities. J Clin Sleep Med 2018; 14(9):1459–1461.
  49. Mehra R. Sleep apnea and nocturnal cardiac arrhythmia: understanding differences across ethnicity. J Clin Sleep Med 2017; 13(11):1229–1231.
  50. May AM, Van Wagoner DR, Mehra R. OSA and cardiac arrhymogenesis: mechanistic insights. Chest 2017; 151(1):225–241.
  51. Dimitri H, Ng M, Brooks AG, et al. Atrial remodeling in obstructive sleep apnea: implications for atrial fibrillation. Heart Rhythm 2012; 9(3):321–327.
  52. Mehra R, Benjamin EJ, Shahar E, et al. Association of nocturnal arrhythmias with sleep-disordered breathing: the Sleep Heart Health Study. Am J Respir Crit Care Med 2006; 173(8):910–916.
  53. Mehra R, Stone KL, Varosy PD, et al. Nocturnal arrhythmias across a spectrum of obstructive and central sleep-disordered breathing in older men: outcomes of sleep disorders in older men (MrOS sleep) study. Arch Intern Med 2009; 169(12):1147–1155.
  54. Monahan K, Storfer-Isser A, Mehra R, et al. Triggering of nocturnal arrhythmias by sleep-disordered breathing events. J Am Coll Cardiol 2009; 54(19):1797–1804.
  55. Gami AS, Hodge DO, Herges RM, et al. Obstructive sleep apnea, obesity, and the risk of incident atrial fibrillation. J Am Coll Cardiol 2007; 49(5):565–571.
  56. May AM, Blackwell T, Stone PH, et al; MrOS Sleep (Outcomes of Sleep Disorders in Older Men) Study Group. Am J Respir Crit Care Med 2016; 193(7):783–791.
  57. Kaw R, El Zarif S, Wang L, Bena J, Blackstone EH, Mehra R. Obesity as an effect modifier in sleep-disordered breathing and postcardiac surgery atrial fibrillation. Chest 2017; 151(6):1279–1287.
  58. Walia H, Strohl KP, Mehra R. Effect of continuous positive airway pressure on an atrial arrhythmia in a patient with mild obstructive sleep apnea. J Clin Sleep Med 2011; 7(4):397–398.
  59. Walia HK, Chung MK, Ibrahim S, Mehra R. Positive airway pressure-induced conversion of atrial fibrillation to normal sinus rhythm in severe obstructive sleep apnea. J Clin Sleep Med 2016; 12(9):1301–1303.
  60. Veasey SC, Davis CW, Fenik P, et al. Long-term intermittent hypoxia in mice: protracted hypersomnolence with oxidative injury to sleep-wake brain regions. Sleep 2004; 27(2):194–201.
  61. Parra O, Arboix A, Bechich S, et al. Time course of sleep-related breathing disorders in first-ever stroke or transient ischemic attack. Am J Respir Crit Care Med 2000; 161(2I):375–380.
  62. Song TJ, Park JH, Choi K, et al. Moderate-to-severe obstructive sleep apnea is associated with cerebral small vessel disease. Sleep Med 2017; 30:36–42.
  63. 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.
  64. Stone KL, Blackwell TL, Ancoli-Israel S, et al; Osteoporotic Fractures in Men (MrOS) Study Research Group. Sleep disordered breathing and risk of stroke in older community-dwelling men. Sleep 2016; 39(3):531–540.
  65. McEvoy RD, Antic NA, Heeley E, et al; SAVE Investigators and Coordinators. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med 2016; 375(10):919–931.
  66. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardiovascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  67. Yeboah J, Redline S, Johnson C, et al. Association between sleep apnea, snoring, incident cardiovascular events and all-cause mortality in an adult population: MESA. Atherosclerosis 2011; 219(2):963–968.
  68. Punjabi NM, Caffo BS, Goodwin JL, et al. Sleep-disordered breathing and mortality: a prospective cohort study. PLoS Med 2009; 6(8):e1000132.
  69. Gami AS, Howard DE, Olson EJ, Somers VK. Day–night pattern of sudden death in obstructive sleep apnea. N Engl J Med 2005; 352(12):1206–1214.
Page Number
10-18
Page Number
10-18
Publications
Publications
Topics
Article Type
Display Headline
Sleep apnea and the heart
Display Headline
Sleep apnea and the heart
Legacy Keywords
continuous positive airway pressure, CPAP, adaptive servo ventilation; phrenic nerve stimulation; sleep-heart interaction, reena mehra, nancy foldvary-schaefer
Legacy Keywords
continuous positive airway pressure, CPAP, adaptive servo ventilation; phrenic nerve stimulation; sleep-heart interaction, reena mehra, nancy foldvary-schaefer
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2019 September;86(9 suppl 1):10-18
Inside the Article

KEY POINTS

  • Diurnal variations in blood pressure, heart rate, and cardiac events occur during normal sleep.
  • While normal sleep may be cardioprotective, sleep apnea disrupts the normal sleep-heart interaction.
  • Untreated severe sleep apnea increases the risk for cardiovascular events.
  • Treatment with continuous positive airway pressure (CPAP) may reduce the risk of cardiac events based on some data, though randomized studies suggest no improvement in cardiovascular mortality.
  • Poor patient adherence to CPAP makes it difficult to evaluate the efficacy of CPAP treatment in clinical trials.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 07:30
Un-Gate On Date
Wed, 08/28/2019 - 07:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 07:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Beyond heart health: Consequences of obstructive sleep apnea

Article Type
Changed
Tue, 09/03/2019 - 12:15
Display Headline
Beyond heart health: Consequences of obstructive sleep apnea

Obstructive sleep apnea (OSA) is a serious condition that impacts quality of life and causes drowsy driving and depression. Research also reveals an interrelationship between OSA and metabolic disease and an association between OSA and cognitive impairment.

The diagnostic criteria of OSA are based on the number of apneic or hypopneic episodes per hour of sleep, called the apnea–hypopnea index (AHI) as recorded during sleep testing. Diagnosis of OSA is warranted if the AHI is 15 or more per hour or if the AHI is 5 or more per hour with 1 or more of the following features: sleepiness; nonrestorative sleep; fatigue or insomnia; waking up with breath-holding spells, gasping, or choking; snoring or breathing interruptions; or a coexisting diagnosis of hypertension, mood disorder, cognitive dysfunction, coronary artery disease, stroke, congestive heart failure, atrial fibrillation, or type II diabetes.1

Many patients with an AHI less than 15 may also have OSA, given the number of coexisting medical conditions included in the OSA diagnostic criteria. Heart conditions such as coronary artery disease, atrial fibrillation, and congestive heart failure encompassed in the OSA diagnostic criteria have increased awareness of the link between OSA and heart health. Less well-known, and the subject of this review, are the negative consequences of OSA, particularly poor quality of life, drowsy driving, depression, metabolic disease, and cognitive impairment.

QUALITY OF LIFE

Reduced quality of life is the most fundamental patient-reported outcome of OSA. OSA is associated with excessive daytime sleepiness, inattention, and fatigue, which increase the risk of accidents and medical disability. These quality-of-life impairments are often the main reason patients seek medical care for sleep disorders.2 Improved quality of life is a central goal of OSA treatment and is the best indicator of the effectiveness of treatment.3 Sleep health and its effect on quality of life is an area of focus of Healthy People 2020 (healthypeople.gov).

The American Academy of Sleep Medicine identified quality of life, along with detection of disease and cardiovascular consequences, as an outcome measure for assessing the quality of care for adults with OSA.2 The assessment of quality of life for patients with OSA is a 4-part process: use evidence-based therapy, monitor the therapy, assess symptoms with a validated tool such as Epworth Sleepiness Scale, and assess the incidence of motor vehicle accidents. Information from these 4 processes can inform changes in a patient’s quality of life.

Treatment for OSA has been shown to improve quality of life. A study of 2,027 patients with OSA evaluated therapy adherence relative to mean Functional Outcomes of Sleep Questionnaire and European Quality of Life-5D scores.4 In patients with the most impaired quality of life, those adherent to positive airway pressure (PAP) therapy had improved quality of life as measured by these scores.

With respect to sleepiness, a systematic review of continuous PAP (CPAP) in patients with OSA found a 2.7-point reduction (mean difference; 95% confidence interval 3.45–1.96) in the Epworth Sleepiness Scale in patients using CPAP compared with the control group.5 Treatment of OSA improves patient quality of life and symptoms such as sleepiness.

DROWSY DRIVING

Drowsy driving by people with OSA can lead to motor vehicle accidents, which result in economic and health burdens.6,7 National Center for Statistics and Analysis data reveal that of 6 million motor vehicle accidents (5-year average, 2005–2009), 1.4% involve drowsy driving and 2.5% of fatal crashes involve drowsy driving.8 Among noncommercial drivers, untreated OSA increases the risk of motor vehicle accidents 3- to 13-fold.9 The odds ratio of traffic accidents in drivers with untreated OSA is 6 times greater than in the general population.7

In a study of men and women in the general population (N = 913), individuals with moderate to severe OSA (AHI > 15) were more likely to have multiple motor vehicle accidents in the course of 5 years (odds ratio = 7.3) compared with those with no sleep-disordered breathing.10 The association between OSA and motor vehicle accidents is independent of sleepiness, and drivers with OSA may not perceive performance impairment.

There are 2 main reasons OSA increases the risk and incidence of motor vehicle accidents. OSA causes changes in attention and vigilance resulting from sleep deprivation and fragmentation. OSA also affects global cognition function, which may be due to intermittent hypoxia attributable to OSA.2

Treatment for OSA is effective in reducing the incidence of motor vehicle accidents. One study found the risk of motor vehicle accidents was eliminated with the use of CPAP treatment in patients with OSA.11 A recent study of nearly 2,000 patients with OSA found a reduction in self-reported near-accidents from 14% before PAP therapy to 5.3% after starting PAP therapy.12

 

 

DEPRESSION

walia_osaconsequences_t1.jpg
Depression can occur as a consequence of OSA. Depression and OSA have several symptoms in common (Table 1).13

Estimates of the prevalence of depression in patients with OSA range from 5% to 63%.13,14 One year after patients were diagnosed with OSA, the incidence of depression per 1,000 person-years was 18% compared with 8% in a group without OSA.14 Women with OSA reportedly have a higher risk of depression (adjusted hazard ratio [HR] 2.7) than men (HR 1.81) at 1-year follow-up.14 In the same study, with respect to age, there was no significant relationship noted between OSA and patients over age 64.

A one-level increase in the severity of OSA (ie, from minimal to mild) is associated with a nearly twofold increase in the adjusted odds for depression.15 On the other hand, studies have also found that patients on antidepressants may have an increased risk of OSA.16

Several potential mechanisms have been proposed to explain the link between depression and OSA.13 Poor-quality sleep, frequent arousal, and fragmentation of sleep in OSA may lead to frontal lobe emotional modulation changes. Intermittent hypoxia in OSA may result in neuronal injury and disruption of noradrenergic and dopaminergic pathways. Pro-inflammatory substances such as interleukin 6 and interleukin 1 are increased in OSA and depression and are mediators between both conditions. Neurotransmitters may be affected by a disrupted sleep-wake cycle. And serotonin, which may be impeded in depression, could influence the upper-airway dilator motor neurons.

Treatment of OSA improves symptoms of depression as measured by the Patient Health Questionnaire (PHQ-9). After 3 months of compliance with CPAP therapy, mean PHQ-9 scores decreased from 11.3 to 2.7 in a study of 228 patients with OSA.17 A study of 1,981 patients with sleep-disordered breathing found improved PHQ-9 scores in patients compliant with CPAP therapy and a greater improvement in patients with a baseline PHQ-9 higher than 10 (moderate severity).18

METABOLIC SYNDROME

OSA is associated with metabolic disorders, including metabolic syndrome, though the causality between these 2 conditions is yet to be illuminated. Metabolic syndrome is a term used when an individual has 3 or more of the following features or conditions:

  • Waist circumference greater than 40 inches (men), greater than 35 inches (women)
  • Triglycerides 150 mg/dL or greater or treatment for hypertriglyceridemia
  • High-density lipoprotein cholesterol less than 40 mg/dL (men), less than 50 mg/dL (women), or treatment for cholesterol
  • Blood pressure 130/85 mm Hg or greater, or treatment for hypertension
  • Fasting blood glucose 100 mg/dL or greater, or treatment for hyperglycemia.19

Metabolic syndrome increases an individual’s risk of diabetes and cardiovascular disease and overall mortality. Like OSA, the prevalence of metabolic syndrome increases with age in both men and women.20,21 The risk of metabolic syndrome is greater with more severe OSA. The Wisconsin Sleep Cohort (N = 546) reported an odds ratio for having metabolic syndrome of 2.5 for patients with mild OSA and 2.2 for patients with moderate or severe OSA.22 A meta-analysis also found a 2.4 times higher odds of metabolic syndrome in patients with mild OSA, but a 3.5 times higher odds of metabolic syndrome in patients with moderate to severe OSA compared with the control group.23

Patients with both OSA and metabolic syndrome are said to have syndrome Z24 and are at increased risk of cardiovascular morbidity and mortality.25 Syndrome Z imparts a higher risk of atherogenic burden and prevalence of atheroma compared with patients with either condition alone.26 In comparing patients with metabolic syndrome with and without OSA, those with OSA had increased atherosclerotic burden as measured by intima-media thickness and carotid femoral pulse-wave velocity.27 Syndrome Z is also linked to intracoronary stenosis related to changes in cardiac morphology28 and is associated with left ventricular hypertrophy and diastolic dysfunction.29

OSA and hypertension

Hypertension is one of the conditions encompassed in metabolic syndrome. Several studies report increased risk and incidence of hypertension in patients with OSA. In a community-based study of 6,123 individuals age 40 and older, sleep-disordered breathing was associated with hypertension, and the odds ratio of hypertension was greater in individuals with more severe sleep apnea.30 Similarly, a landmark prospective, population-based study of 709 individuals over 4 years reported a dose-response relationship between patients with OSA and newly diagnosed hypertension independent of confounding factors.31 Patients with moderate to severe OSA had an odds ratio of 2.89 of developing hypertension after adjusting for confounding variables.

A study of 1,889 individuals followed for 12 years found a dose-response relationship based on OSA severity for developing hypertension.32 This study also assessed the incidence of hypertension based on CPAP use. Patients with poor adherence to CPAP use had an 80% increased incidence of hypertension, whereas patients adhering to CPAP use had a 30% decrease in the incidence of hypertension.

Resistant hypertension (ie, uncontrolled hypertension despite use of 3 or more antihypertensive and diuretic medications) has been shown to be highly prevalent (85%) in patients with severe OSA.33 An analysis of patients at increased risk of cardiovascular disease and untreated severe OSA was associated with a 4 times higher risk of elevated blood pressure despite intensive medical therapy.34

 

 

Mechanisms of altered metabolic regulation in OSA

Mechanisms implicated in altering metabolic regulation in OSA include intermittent hypoxia, sleep fragmentation and glucose homeostasis, and obesity. Intermittent hypoxia from OSA results in sympathetic nervous system activation that affects the pancreas, skeletal muscle, liver, and fat cells resulting in altered insulin secretion, lipid-bile synthesis, glucose metabolism, and lipoprotein metabolism.35

Sleep fragmentation is a cardinal feature of OSA and the resulting suppression of sleep may alter insulin sensitivity. Studies have implicated disruptions to slow-wave sleep specifically, as well as disruption of any stage of sleep in reduced insulin sensitivity.35,36 In addition to decreased insulin sensitivity, sleep fragmentation also increases morning cortisol levels and increases sympathetic nervous system activation.37

Obesity and OSA share a pathway imparting increased cardiometabolic risk.38 Fat tissue causes higher systemic inflammation and inflammatory markers. A recent report describes a bidirectional relationship between metabolic syndrome and OSA.39 While OSA increases the risk for metabolic syndrome, metabolic syndrome by virtue of body mass index with changes in mechanical load and narrow airway and physiology can predispose for OSA.

Effect of treatment for OSA on metabolic syndrome

Several studies have evaluated the effect of CPAP treatment for OSA on metabolic syndrome overall, as well as the specific conditions that comprise metabolic syndrome. In evaluating CPAP use and metabolic syndrome overall, studies have found a reduced prevalence of metabolic syndrome,40,41 CPAP benefit only in complying patients,42 and a reduction in oxidative stress with a single-night use of CPAP.43

With respect to insulin sensitivity, a study of 40 men with moderate OSA using CPAP therapy (mean use 5 hours) reported an increase in the insulin sensitivity index after 2 days, and a further increase after 3 months.44 Another study found no improvement in insulin resistance in severe OSA.45 A meta-analysis reported improved insulin resistance with CPAP,46 although a recent meta-analysis assessing hemoglobin A1c level, fasting insulin level, and fasting glucose did not show improvement in these parameters. Large-scale clinical trials with longer treatment duration and better CPAP compliance are warranted.47

walia_osaconsequences_t2.jpg
CPAP use in patients with OSA has been found to affect hypertension in a number of studies (Table 2).48–55 In a comparison of therapeutic CPAP with suboptimal CPAP for 9 weeks, ambulatory blood pressure was reduced in the therapeutic group, and no change was seen in the subtherapeutic group, illustrating the importance of optimal pressure settings in treating OSA.48

walia_osaconsequences_f1.jpg
%3Cp%3EFigure%201.%20Blood%20pressure%20trends%20in%20patients%20before%20and%20after%20continuous%20positive%20airway%20pressure%20(CPAP)%20use.%3C%2Fp%3E
A randomized controlled trial of nearly 300 individuals found improvement in 6 blood pressure parameters in a group using CPAP compared with a group using sham CPAP after 12 weeks.50 A large clinic-based cohort of 894 individuals with hypertension and resistant hypertension (15%) found that after 1 year, CPAP use was associated with 2 to 3 mm Hg of reduction in blood pressure (Figure 1).56 Meta-analysis of randomized controlled trials on the effectiveness of CPAP on hypertension found reductions of 2 mm Hg to 3 mm Hg in blood pressure.57 Another meta-analyses showed a reduction of 2.6 mm Hg in 24-hour mean blood pressure with CPAP therapy (Table 2).48–55 This reduction may appear modest in nature; however, any reduction in blood pressure can result in decreased cardiovascular morbidity and mortality. A meta-analysis of randomized controlled trials indicated reductions in mean systolic blood pressure of 5.4 mm Hg and diastolic blood pressure of 3.86 mm Hg after CPAP in those with resistant hypertension and OSA.58

Weight loss has been shown to reduce the AHI and other parameters related to sleep apnea such as oxygen desaturation index in patients with obesity and diabetes.59 Weight loss combined with CPAP compared with CPAP or weight loss alone showed an incremental benefit in improving glucose parameters, triglycerides, and possibly systolic blood pressure and triglycerides.60

 

 

COGNITIVE IMPAIRMENT

Data suggest that OSA is linked with cognitive impairment, may advance cognitive decline, and is a bidirectional relationship. Women with OSA were reportedly more likely to develop mild cognitive impairment compared with women without OSA.61 An elevated oxygen desaturation index and a high percentage of time spent with hypoxia was associated with increased risk of developing mild cognitive impairment and dementia.

OSA was found to be an independent risk factor for cerebral white matter changes in middle-age and older individuals. Moderate to severe OSA imparted a 2 times higher risk of cerebral white matter changes compared with individuals without OSA.62 Another study of 20 patients with severe OSA compared with 40 healthy volunteers found diffusion imaging consistent with impaired fibrin integrity in those with OSA, indicative of white matter microstructure damage, and the impairment was associated with increased disease severity and higher systemic inflammation.63

Individuals with hypoxia for a high percentage of time during sleep had a 4 times higher odds of cerebral microinfarcts.64 Cognitive scores decreased less in men. Men typically have more time in slow-wave sleep, suggesting that slow-wave sleep may be protective against cognitive decline. Mild cognitive impairment and Alzheimer disease were found more likely to develop and occur at an earlier age in individuals with sleep-disordered breathing compared with individuals without sleep-disordered breathing.65

OSA was also associated with increased serum amyloid beta levels in a study of 45 cognitively normal patients with OSA compared with 49 age- and sex-matched control patients. Increased amyloid beta levels correlated with increasing severity of sleep apnea as measured by the AHI.66

Mechanism linking OSA and cognition

One possible mechanism linking sleep quality and cognitive impairment or Alzheimer disease is the role of unfragmented sleep in attenuating the apolipo­protein E e4 allele on the incidence of Alzheimer disease.67 Beta amyloid is released during synaptic activity. Neuronal and synaptic activity decreases during sleep, and disrupted sleep could increase beta amyloid release.68 Sleep has been found to enhance the clearance of beta amyloid peptide from the brain interstitial fluid in a mice model.69

Recent data point toward the bidirectional relationship between the sleep and Alzheimer disease in that excessive and prolonged neuronal activity in the absence of appropriately structured sleep may be the reason for both Alzheimer disease and OSA.70,71

Effect of treatment for OSA on cognition

White matter integrity in 15 patients with OSA before and after treatment with CPAP was compared with 15 matched controls. Over 12 months, there was a nearly complete reversal of white matter abnormalities in patients on CPAP therapy.72 Improvement in memory, attention, and executive function paralleled the changes in white matter after the treatment.

CONCLUSION

OSA is a serious condition with far-reaching consequences associated with impaired quality of life, depressive symptoms, drowsy driving, metabolic disease, and cognitive decline. Treatment of OSA improves many of these health consequences, emphasizing the need for OSA screening. Large randomized studies are needed to assess the efficacy of CPAP on metabolic outcomes, including insulin sensitivity and glucose tolerance, in reducing disease burden. Study of the endophenotypes of patients with OSA is needed to define and target the mechanisms of OSA-induced adverse health outcomes and perhaps lead to personalized care for patients with OSA.

References
  1. Sateia MJ. International Classification of Sleep Disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  2. Aurora RN, Collop NA, Jacobowitz O, Thomas SM, Quan SF, Aronsky AJ. Quality measures for the care of adult patients with obstructive sleep apnea. J Clin Sleep Med 2015; 11(3):357–383.
  3. Flemons WW. Obstructive sleep apnea. N Engl J Med 2002; 347(7): 498–504.
  4. Walia HK, Thompson NR, Katzan I, Foldvary-Schaefer N, Moul DE, Mehra R. Impact of sleep-disordered breathing treatment on quality of life measures in a large clinic-based cohort. J Clin Sleep Med; 2017;13(11):1255–1263. doi:10.5664/jcsm.6792
  5. McDaid C, Dureé KH, Griffin SC, et al. A systematic review of continuous positive airway pressure for obstructive sleep apnoea–hypopnoea syndrome. Sleep Med Rev 2009; 13(6):427–436.
  6. Arita A, Sasanabe R, Hasegawa R, et al. Risk factors for auto­mobile accidents caused by falling asleep while driving in obstructive sleep apnea syndrome. Sleep Breath 2015; 19(4):1229–1234.
  7. Terán-Santos J, Jiménez-Gómez A, Cordero-Guevara J; the Cooperative Group Burgos–Santander. The association between sleep apnea and the risk of traffic accidents. N Engl J Med 1999; 340(11):847–851.
  8. NHTSA. Drowsy driving. Washington, DC: National Highway Traffic Safety Administration. https://crashstats.nhtsa.dot.gov/Api/Public/ViewPublication/811449. Published March 2011. Accessed August 19, 2019.
  9. Vakulin A, D’Rozario A, Kim J-W, et al. Quantitative sleep EEG and polysomnographic predictors of driving simulator performance in obstructive sleep apnea. Clin Neurophysiol 2016; 127(2):1428–1435.
  10. Young T, Blustein J, Finn L, Palta M. Sleep-disordered breathing and motor vehicle accidents in a population-based sample of employed adults. Sleep 1997; 20(8):608–613.
  11. George CFP. Reduction in motor vehicle collisions following treatment of sleep apnoea with nasal CPAP. Thorax 2001; 56(7):508–512.
  12. Walia HK, Thompson N, Pascoe M, et al. Impact of positive airway pressure therapy on drowsy driving in a large clinic-based obstructive sleep apnea cohort. J Clin Sleep Med (in press).
  13. Ejaz SM, Khawaja IS, Bhatia S, Hurwitz TD. Obstructive sleep apnea and depression: a review. Innov Clin Neurosci 2011; 8(8):17–25.
  14. Chen Y-H, Keller JK, Kang J-H, Hsieh H-J, Lin H-C. Obstructive sleep apnea and the subsequent risk of depressive disorder: a population-based follow-up study. J Clin Sleep Med 2013; 9(5):417–423.
  15. Peppard PE, Szklo-Coxe M, Hla KM, Young T. Longitudinal association of sleep-related breathing disorder and depression. Arch Intern Med 2006; 166(16):1709–1715.
  16. Farney RJ, Lugo A, Jensen RL, Walker JM, Cloward TV. Simultaneous use of antidepressant and antihypertensive medications increases likelihood of diagnosis of obstructive sleep apnea syndrome. Chest; 2004;125(4):1279–1285.
  17. Edwards C, Mukherjee S, Simpson L, Palmer LJ, Almeida OP, Hillman DR. Depressive symptoms before and after treatment of obstructive sleep apnea in men and women. J Clin Sleep Med 2015; 11(9):1029–1038.
  18. Relia S, Thompson NR, Mehra R, et al. Depression score changes in response to sleep disordered breathing treatment with positive airway pressure in a large clinic-based cohort. Sleep Breath 2018; 22(1):195–203.
  19. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002; 106(25):3143–3421.
  20. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the Third National Health and Nutrition Examination Survey. JAMA 2002; 287(3):356–359.
  21. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  22. Nieto FJ, Peppard PE, Young TB. Sleep disordered breathing and metabolic syndrome. WMJ 2009; 108(5):263–265.
  23. Xu S, Wan Y, Xu M, et al. The association between obstructive sleep apnea and metabolic syndrome: a systematic review and meta-analysis. BMC Pulm Med 2015; 15:105.
  24. Nock NL, Li L, Larkin EK, Patel SR, Redline S. Empirical evidence for “syndrome Z”: a hierarchical 5-factor model of the metabolic syndrome incorporating sleep disturbance measures. Sleep 2009; 32(5):615–622.
  25. Sadasivam K, Chinnasami B, Ayyavo S, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diagn Res 2015; 9(4):CC07–CC10.
  26. Chang TI, Tanner JM, Harada ND, Garrett NR, Friedlander AH. Prevalence of calcified carotid artery atheromas on the panoramic images of patients with syndrome Z, coexisting obstructive sleep apnea, and metabolic syndrome. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113(1):134–141.
  27. Drager LF, Bortolotto LA, Maki-Nunes C, et al. The incremental role of obstructive sleep apnoea on markers of atherosclerosis in patients with metabolic syndrome. Atherosclerosis 2010; 208(2):490–495.
  28. Nakanishi-Minami T, Kishida K, Nakagawa Y, et al. Metabolic syndrome correlates intracoronary stenosis detected by multislice computed tomography in male subjects with sleep-disordered breathing. Diabetol Metab Syndr 2012; 4:6.
  29. Usui Y, Takata Y, Inoue Y, et al. Coexistence of obstructive sleep apnoea and metabolic syndrome is independently associated with left ventricular hypertrophy and diastolic dysfunction. Sleep Breath 2012; 16(3):677–684.
  30. Nieto FJ, Young TB, Lind BK, et al; for the Sleep Heart Health Study. Association of sleep-disordered breathing, sleep apnea, and hypertension in a large community-based study. JAMA 2000; 283(14):1829–1836.
  31. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of the association between sleep-disordered breathing and hypertension. N Engl J Med 2000; 342(19):1378–1384.
  32. Marin JM, Agusti A, Villar I, et al. Association between treated and untreated obstructive sleep apnea and risk of hypertension. JAMA 2012; 307(20):2169–2176.
  33. Gonçalves SC, Martinez D, Gus M, et al. Obstructive sleep apnea and resistant hypertension: a case-control study. Chest 2007; 132(6):1858–1862.
  34. Walia HK, Li H, Rueschman M, et al. Association of severe obstructive sleep apnea and elevated blood pressure despite anti­hypertensive medication use. J Clin Sleep Med 2014; 10(8):835–843.
  35. Braincon-Marjollet A, Weiszenstein M, Henri M, Thomas A, Godin-Ribuot D, Polak J. The impact of sleep disorders on glucose metabolism: endocrine and molecular mechanisms. Diabetol Metab Syndr 2015; 7:25. doi:10.1186/s13098-015-0018-3
  36. Stamatakis KA, Punjabi NM. Effects of sleep fragmentation on glucose metabolism in normal subjects. Chest 2010; 137(1):95–101.
  37. Spiegel K. Knutson K, Leproult R, Tasali E, Van Cauter E. Sleep loss: a novel risk factor for insulin resistance and type 2 diabetes. J Appl Physiol (1985) 2005; 99(5):2008–2019.
  38. Pépin J-L, Tamisier R, Lévy P. Obstructive sleep apnoea and metabolic syndrome: put CPAP efficacy in a more realistic perspective. Thorax 2012; 67(12):1025–1027.
  39. Framnes SN, Arble DM. The bidirectional relationship between obstructive sleep apnea and metabolic disease. Front Endocrinol (Lausanne) 2018; 9:440.
  40. Oktay B, Akbal E, Firat H, Ardiç S, Kizilgun M. CPAP treatment in the coexistence of obstructive sleep apnea syndrome and metabolic syndrome, results of one year follow up. Acta Clin Belg 2009; 64(4):329–334.
  41. Mota PC, Drummond M, Winck JC, Santos AC, Almeida J, Marques JA. APAP impact on metabolic syndrome in obstructive sleep apnea patients. Sleep Breath 2011; 15(4):665–672.
  42. Dorkova Z, Petrasova D, Molcanyiova A, Popovnakova M, Tkacova R. Effects of continuous positive airway pressure on cardio­vascular risk profile in patients with severe obstructive sleep apnea and metabolic syndrome. Chest 2008; 134(4):686–692.
  43. Kanimozhi S, Balaji C, Saravanan A, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diag Research 2015; 9(4):CC07–CC10.
  44. Harsch IA, Schahin SP, Radespiel-Tröger M, et al. Continuous positive airway pressure treatment rapidly improves insulin sensitivity in patients with obstructive sleep apnea syndrome. Am J Respir Crit Care Med 2004; 169(2):156–162.
  45. Trenell MI, Ward JA, Yee BJ, et al. Influence of constant positive airway pressure therapy on lipid storage, muscle metabolism and insulin action in obese patients with severe obstructive sleep apnoea syndrome. Diabetes Obes Metab 2007; 9(5):679–687.
  46. Iftikhar IH, Hoyos CM, Phillips CL, Magalang UJ. Meta-analysis of the association of sleep apnea with insulin resistance, and the effects of CPAP on HOMA-IR, adiponectin, and visceral adipose fat. J Clin Sleep Med 2015; 11(4):475–485.
  47. Zhu B, Ma C, Chaiard J, Shi C. Effect of continuous positive airway pressure on glucose metabolism in adults with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Sleep Breath 2018; 22(2):287–295.
  48. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  49. Campos-Rodriguez F, Grilo-Reina A, Perez-Ronchel J, et al. Effect of continuous positive airway pressure on ambulatory BP in patients with sleep apnea and hypertension: a placebo-controlled trial. Chest 2006; 129(6):1459–1467.
  50. Durán-Cantolla J, Aizpuru F, Montserrat JM, et al; on behalf of the Spanish Sleep and Breathing Group. Continuous positive airway pressure as treatment for systemic hypertension in people with obstructive sleep apnoea: randomised controlled trial. BMJ 2010; 341:c5991.
  51. Gottlieb DJ, Punjabi NM, Mehra R, et al. CPAP versus oxygen in obstructive sleep apnea. N Engl J Med 2014; 370(24):2276–2285.
  52. Hui DS, To KW, Ko FW, et al. Nasal CPAP reduces systemic blood pressure in patients with obstructive sleep apnoea and mild sleepiness. Thorax 2006; 61(12):1083–1090.
  53. Martinez-Garcia MA, Capote F, Campos-Rodriguez F, et al. Effect of CPAP on blood pressure in patients with obstructive sleep apnea and resistant hypertension: the HIPARCO randomized clinical trial. JAMA 2013; 310(22):2407–2415.
  54. Pepperell JC, Ramdassingh-Dow S, Crosthwaite N, et al. Ambulatory blood pressure after therapeutic and subtherapeutic nasal continuous positive airway pressure for obstructive sleep apnoea: a randomised parallel trial. Lancet 2002; 359(9302):204–210.
  55. Robinson GV, Smith DM, Langford BA, Davies RJ, Stradling JR. Continuous positive airway pressure does not reduce blood pressure in nonsleepy hypertensive OSA patients. Eur Respir J 2006; 27(6):1229–1235.
  56. Walia HK, Griffith SD, Foldvary-Schaefer N, et al. Longitudinal effect of CPAP on BP in resistant and nonresistant hypertension in a large clinic-based cohort. Chest 2016; 149(3):747–755.
  57. Montesi SB, Edwards BA, Malhotra A, Bakker JP. The effect of continuous positive airway pressure treatment on blood pressure: a systematic review and meta-analysis of randomized controlled trials. J Clin Sleep Med 2012; 8(5):587–596.
  58. Lei Q, Lv Y, Li K, Ma L, Du G, Xiang Y, Li X. Effects of continuous positive airway pressure on blood pressure in patients with resistant hypertension and obstructive sleep apnea: a systematic review and meta-analysis of six randomized controlled trials. J Bras Pneumol 2017;43(5):373–379. doi:10.1590/S1806-37562016000000190. [Article in English, Portuguese]
  59. Foster GD, Borradaile KE, Sanders MH; for the Sleep AHEAD Research Group of Look AHEAD Research Group. A randomized study on the effect of weight loss on obstructive sleep apnea among obese patients with type 2 diabetes: the Sleep AHEAD study. Arch Intern Med 2009; 169(17):1619–1626.
  60. Chirinos JA, Gurubhagavatula I, Teff K, et al. CPAP, weight loss, or both for obstructive sleep apnea. N Engl J Med 2014; 370(24):2265–2275.
  61. Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306(6):613–619.
  62. Kim H, Yun C-H, Thomas RJ, et al. Obstructive sleep apnea as a risk factor for cerebral white matter change in a middle-aged and older general population. Sleep 2013; 36(5):709–715B.
  63. Chen H-L, Lu C-H, Lin H-C, et al. White matter damage and systemic inflammation in obstructive sleep apnea. Sleep 2015; 38(3):361–370.
  64. Gelber RP, Redline S, Ross GW, et al. Associations of brain lesions at autopsy with polysomnography features before death. Neurology 2015; 84(3):296–303.
  65. Osorio RS, Gumb T, Pirraglia E, et al; for the Alzheimer’s Disease Neuroimaging Initiative. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology 2015; 84(19):1964–1971.
  66. Bu X-L, Liu Y-H, Wang Q-H, et al. Serum amyloid-beta levels are increased in patients with obstructive sleep apnea syndrome. Sci Rep 2015; 5:13917.
  67. Lim ASP, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the relationship of the apolipoprotein e 4 allele to the risk of Alzheimer disease and neurofibrillary tangle density by sleep. JAMA Neurol 2013; 70(12):1544–1551.
  68. Lucey BP, Bateman RJ. Amyloid-beta diurnal pattern: possible role of sleep in Alzheimer’s disease pathogenesis. Neurobiol Aging 2014; 35(suppl 2):S29–S34.
  69. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science 2013; 342(6156):373–377.
  70. Polsek D, Gildeh N, Cash D, et al. Obstructive sleep apnoea and Alzheimer’s disease: in search of shared pathomechanisms. Neurosci Biobehav Rev 2018; 86:142–149.
  71. Ju Y-ES, Lucey BP, Holtzman DM. Sleep and Alzheimer disease pathology—a bidirectional relationship. Nat Rev Neurol 2014; 10(2):115–119.
  72. Castronovo V, Scifo P, Castellano A, et al. White matter integrity in obstructive sleep apnea before and after treatment. Sleep 2014; 37(9):1465–1475.
Article PDF
Author and Disclosure Information

Harneet K. Walia, MD, FAASM
Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Staff, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Harneet K. Walia, MD, FAASM, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; waliah@ccf.org

Dr. Walia is principal investigator of a ResMed funded grant.

This article is based on Dr. Walia’s “Beyond Heart Health: Consequences of Obstructive Sleep Apnea” webcast released March 29, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Walia.

Publications
Topics
Page Number
19-25
Legacy Keywords
sleep apnea, obstructive sleep apnea, OSA, quality of life, depression, drowsy driving, metabolic syndrome, syndrome Z, hypertension, Alzheimer disease, cognitive decline, harneet walia, nancy foldvary-schaefer
Sections
Author and Disclosure Information

Harneet K. Walia, MD, FAASM
Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Staff, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Harneet K. Walia, MD, FAASM, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; waliah@ccf.org

Dr. Walia is principal investigator of a ResMed funded grant.

This article is based on Dr. Walia’s “Beyond Heart Health: Consequences of Obstructive Sleep Apnea” webcast released March 29, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Walia.

Author and Disclosure Information

Harneet K. Walia, MD, FAASM
Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Staff, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Harneet K. Walia, MD, FAASM, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; waliah@ccf.org

Dr. Walia is principal investigator of a ResMed funded grant.

This article is based on Dr. Walia’s “Beyond Heart Health: Consequences of Obstructive Sleep Apnea” webcast released March 29, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Walia.

Article PDF
Article PDF
Related Articles

Obstructive sleep apnea (OSA) is a serious condition that impacts quality of life and causes drowsy driving and depression. Research also reveals an interrelationship between OSA and metabolic disease and an association between OSA and cognitive impairment.

The diagnostic criteria of OSA are based on the number of apneic or hypopneic episodes per hour of sleep, called the apnea–hypopnea index (AHI) as recorded during sleep testing. Diagnosis of OSA is warranted if the AHI is 15 or more per hour or if the AHI is 5 or more per hour with 1 or more of the following features: sleepiness; nonrestorative sleep; fatigue or insomnia; waking up with breath-holding spells, gasping, or choking; snoring or breathing interruptions; or a coexisting diagnosis of hypertension, mood disorder, cognitive dysfunction, coronary artery disease, stroke, congestive heart failure, atrial fibrillation, or type II diabetes.1

Many patients with an AHI less than 15 may also have OSA, given the number of coexisting medical conditions included in the OSA diagnostic criteria. Heart conditions such as coronary artery disease, atrial fibrillation, and congestive heart failure encompassed in the OSA diagnostic criteria have increased awareness of the link between OSA and heart health. Less well-known, and the subject of this review, are the negative consequences of OSA, particularly poor quality of life, drowsy driving, depression, metabolic disease, and cognitive impairment.

QUALITY OF LIFE

Reduced quality of life is the most fundamental patient-reported outcome of OSA. OSA is associated with excessive daytime sleepiness, inattention, and fatigue, which increase the risk of accidents and medical disability. These quality-of-life impairments are often the main reason patients seek medical care for sleep disorders.2 Improved quality of life is a central goal of OSA treatment and is the best indicator of the effectiveness of treatment.3 Sleep health and its effect on quality of life is an area of focus of Healthy People 2020 (healthypeople.gov).

The American Academy of Sleep Medicine identified quality of life, along with detection of disease and cardiovascular consequences, as an outcome measure for assessing the quality of care for adults with OSA.2 The assessment of quality of life for patients with OSA is a 4-part process: use evidence-based therapy, monitor the therapy, assess symptoms with a validated tool such as Epworth Sleepiness Scale, and assess the incidence of motor vehicle accidents. Information from these 4 processes can inform changes in a patient’s quality of life.

Treatment for OSA has been shown to improve quality of life. A study of 2,027 patients with OSA evaluated therapy adherence relative to mean Functional Outcomes of Sleep Questionnaire and European Quality of Life-5D scores.4 In patients with the most impaired quality of life, those adherent to positive airway pressure (PAP) therapy had improved quality of life as measured by these scores.

With respect to sleepiness, a systematic review of continuous PAP (CPAP) in patients with OSA found a 2.7-point reduction (mean difference; 95% confidence interval 3.45–1.96) in the Epworth Sleepiness Scale in patients using CPAP compared with the control group.5 Treatment of OSA improves patient quality of life and symptoms such as sleepiness.

DROWSY DRIVING

Drowsy driving by people with OSA can lead to motor vehicle accidents, which result in economic and health burdens.6,7 National Center for Statistics and Analysis data reveal that of 6 million motor vehicle accidents (5-year average, 2005–2009), 1.4% involve drowsy driving and 2.5% of fatal crashes involve drowsy driving.8 Among noncommercial drivers, untreated OSA increases the risk of motor vehicle accidents 3- to 13-fold.9 The odds ratio of traffic accidents in drivers with untreated OSA is 6 times greater than in the general population.7

In a study of men and women in the general population (N = 913), individuals with moderate to severe OSA (AHI > 15) were more likely to have multiple motor vehicle accidents in the course of 5 years (odds ratio = 7.3) compared with those with no sleep-disordered breathing.10 The association between OSA and motor vehicle accidents is independent of sleepiness, and drivers with OSA may not perceive performance impairment.

There are 2 main reasons OSA increases the risk and incidence of motor vehicle accidents. OSA causes changes in attention and vigilance resulting from sleep deprivation and fragmentation. OSA also affects global cognition function, which may be due to intermittent hypoxia attributable to OSA.2

Treatment for OSA is effective in reducing the incidence of motor vehicle accidents. One study found the risk of motor vehicle accidents was eliminated with the use of CPAP treatment in patients with OSA.11 A recent study of nearly 2,000 patients with OSA found a reduction in self-reported near-accidents from 14% before PAP therapy to 5.3% after starting PAP therapy.12

 

 

DEPRESSION

walia_osaconsequences_t1.jpg
Depression can occur as a consequence of OSA. Depression and OSA have several symptoms in common (Table 1).13

Estimates of the prevalence of depression in patients with OSA range from 5% to 63%.13,14 One year after patients were diagnosed with OSA, the incidence of depression per 1,000 person-years was 18% compared with 8% in a group without OSA.14 Women with OSA reportedly have a higher risk of depression (adjusted hazard ratio [HR] 2.7) than men (HR 1.81) at 1-year follow-up.14 In the same study, with respect to age, there was no significant relationship noted between OSA and patients over age 64.

A one-level increase in the severity of OSA (ie, from minimal to mild) is associated with a nearly twofold increase in the adjusted odds for depression.15 On the other hand, studies have also found that patients on antidepressants may have an increased risk of OSA.16

Several potential mechanisms have been proposed to explain the link between depression and OSA.13 Poor-quality sleep, frequent arousal, and fragmentation of sleep in OSA may lead to frontal lobe emotional modulation changes. Intermittent hypoxia in OSA may result in neuronal injury and disruption of noradrenergic and dopaminergic pathways. Pro-inflammatory substances such as interleukin 6 and interleukin 1 are increased in OSA and depression and are mediators between both conditions. Neurotransmitters may be affected by a disrupted sleep-wake cycle. And serotonin, which may be impeded in depression, could influence the upper-airway dilator motor neurons.

Treatment of OSA improves symptoms of depression as measured by the Patient Health Questionnaire (PHQ-9). After 3 months of compliance with CPAP therapy, mean PHQ-9 scores decreased from 11.3 to 2.7 in a study of 228 patients with OSA.17 A study of 1,981 patients with sleep-disordered breathing found improved PHQ-9 scores in patients compliant with CPAP therapy and a greater improvement in patients with a baseline PHQ-9 higher than 10 (moderate severity).18

METABOLIC SYNDROME

OSA is associated with metabolic disorders, including metabolic syndrome, though the causality between these 2 conditions is yet to be illuminated. Metabolic syndrome is a term used when an individual has 3 or more of the following features or conditions:

  • Waist circumference greater than 40 inches (men), greater than 35 inches (women)
  • Triglycerides 150 mg/dL or greater or treatment for hypertriglyceridemia
  • High-density lipoprotein cholesterol less than 40 mg/dL (men), less than 50 mg/dL (women), or treatment for cholesterol
  • Blood pressure 130/85 mm Hg or greater, or treatment for hypertension
  • Fasting blood glucose 100 mg/dL or greater, or treatment for hyperglycemia.19

Metabolic syndrome increases an individual’s risk of diabetes and cardiovascular disease and overall mortality. Like OSA, the prevalence of metabolic syndrome increases with age in both men and women.20,21 The risk of metabolic syndrome is greater with more severe OSA. The Wisconsin Sleep Cohort (N = 546) reported an odds ratio for having metabolic syndrome of 2.5 for patients with mild OSA and 2.2 for patients with moderate or severe OSA.22 A meta-analysis also found a 2.4 times higher odds of metabolic syndrome in patients with mild OSA, but a 3.5 times higher odds of metabolic syndrome in patients with moderate to severe OSA compared with the control group.23

Patients with both OSA and metabolic syndrome are said to have syndrome Z24 and are at increased risk of cardiovascular morbidity and mortality.25 Syndrome Z imparts a higher risk of atherogenic burden and prevalence of atheroma compared with patients with either condition alone.26 In comparing patients with metabolic syndrome with and without OSA, those with OSA had increased atherosclerotic burden as measured by intima-media thickness and carotid femoral pulse-wave velocity.27 Syndrome Z is also linked to intracoronary stenosis related to changes in cardiac morphology28 and is associated with left ventricular hypertrophy and diastolic dysfunction.29

OSA and hypertension

Hypertension is one of the conditions encompassed in metabolic syndrome. Several studies report increased risk and incidence of hypertension in patients with OSA. In a community-based study of 6,123 individuals age 40 and older, sleep-disordered breathing was associated with hypertension, and the odds ratio of hypertension was greater in individuals with more severe sleep apnea.30 Similarly, a landmark prospective, population-based study of 709 individuals over 4 years reported a dose-response relationship between patients with OSA and newly diagnosed hypertension independent of confounding factors.31 Patients with moderate to severe OSA had an odds ratio of 2.89 of developing hypertension after adjusting for confounding variables.

A study of 1,889 individuals followed for 12 years found a dose-response relationship based on OSA severity for developing hypertension.32 This study also assessed the incidence of hypertension based on CPAP use. Patients with poor adherence to CPAP use had an 80% increased incidence of hypertension, whereas patients adhering to CPAP use had a 30% decrease in the incidence of hypertension.

Resistant hypertension (ie, uncontrolled hypertension despite use of 3 or more antihypertensive and diuretic medications) has been shown to be highly prevalent (85%) in patients with severe OSA.33 An analysis of patients at increased risk of cardiovascular disease and untreated severe OSA was associated with a 4 times higher risk of elevated blood pressure despite intensive medical therapy.34

 

 

Mechanisms of altered metabolic regulation in OSA

Mechanisms implicated in altering metabolic regulation in OSA include intermittent hypoxia, sleep fragmentation and glucose homeostasis, and obesity. Intermittent hypoxia from OSA results in sympathetic nervous system activation that affects the pancreas, skeletal muscle, liver, and fat cells resulting in altered insulin secretion, lipid-bile synthesis, glucose metabolism, and lipoprotein metabolism.35

Sleep fragmentation is a cardinal feature of OSA and the resulting suppression of sleep may alter insulin sensitivity. Studies have implicated disruptions to slow-wave sleep specifically, as well as disruption of any stage of sleep in reduced insulin sensitivity.35,36 In addition to decreased insulin sensitivity, sleep fragmentation also increases morning cortisol levels and increases sympathetic nervous system activation.37

Obesity and OSA share a pathway imparting increased cardiometabolic risk.38 Fat tissue causes higher systemic inflammation and inflammatory markers. A recent report describes a bidirectional relationship between metabolic syndrome and OSA.39 While OSA increases the risk for metabolic syndrome, metabolic syndrome by virtue of body mass index with changes in mechanical load and narrow airway and physiology can predispose for OSA.

Effect of treatment for OSA on metabolic syndrome

Several studies have evaluated the effect of CPAP treatment for OSA on metabolic syndrome overall, as well as the specific conditions that comprise metabolic syndrome. In evaluating CPAP use and metabolic syndrome overall, studies have found a reduced prevalence of metabolic syndrome,40,41 CPAP benefit only in complying patients,42 and a reduction in oxidative stress with a single-night use of CPAP.43

With respect to insulin sensitivity, a study of 40 men with moderate OSA using CPAP therapy (mean use 5 hours) reported an increase in the insulin sensitivity index after 2 days, and a further increase after 3 months.44 Another study found no improvement in insulin resistance in severe OSA.45 A meta-analysis reported improved insulin resistance with CPAP,46 although a recent meta-analysis assessing hemoglobin A1c level, fasting insulin level, and fasting glucose did not show improvement in these parameters. Large-scale clinical trials with longer treatment duration and better CPAP compliance are warranted.47

walia_osaconsequences_t2.jpg
CPAP use in patients with OSA has been found to affect hypertension in a number of studies (Table 2).48–55 In a comparison of therapeutic CPAP with suboptimal CPAP for 9 weeks, ambulatory blood pressure was reduced in the therapeutic group, and no change was seen in the subtherapeutic group, illustrating the importance of optimal pressure settings in treating OSA.48

walia_osaconsequences_f1.jpg
%3Cp%3EFigure%201.%20Blood%20pressure%20trends%20in%20patients%20before%20and%20after%20continuous%20positive%20airway%20pressure%20(CPAP)%20use.%3C%2Fp%3E
A randomized controlled trial of nearly 300 individuals found improvement in 6 blood pressure parameters in a group using CPAP compared with a group using sham CPAP after 12 weeks.50 A large clinic-based cohort of 894 individuals with hypertension and resistant hypertension (15%) found that after 1 year, CPAP use was associated with 2 to 3 mm Hg of reduction in blood pressure (Figure 1).56 Meta-analysis of randomized controlled trials on the effectiveness of CPAP on hypertension found reductions of 2 mm Hg to 3 mm Hg in blood pressure.57 Another meta-analyses showed a reduction of 2.6 mm Hg in 24-hour mean blood pressure with CPAP therapy (Table 2).48–55 This reduction may appear modest in nature; however, any reduction in blood pressure can result in decreased cardiovascular morbidity and mortality. A meta-analysis of randomized controlled trials indicated reductions in mean systolic blood pressure of 5.4 mm Hg and diastolic blood pressure of 3.86 mm Hg after CPAP in those with resistant hypertension and OSA.58

Weight loss has been shown to reduce the AHI and other parameters related to sleep apnea such as oxygen desaturation index in patients with obesity and diabetes.59 Weight loss combined with CPAP compared with CPAP or weight loss alone showed an incremental benefit in improving glucose parameters, triglycerides, and possibly systolic blood pressure and triglycerides.60

 

 

COGNITIVE IMPAIRMENT

Data suggest that OSA is linked with cognitive impairment, may advance cognitive decline, and is a bidirectional relationship. Women with OSA were reportedly more likely to develop mild cognitive impairment compared with women without OSA.61 An elevated oxygen desaturation index and a high percentage of time spent with hypoxia was associated with increased risk of developing mild cognitive impairment and dementia.

OSA was found to be an independent risk factor for cerebral white matter changes in middle-age and older individuals. Moderate to severe OSA imparted a 2 times higher risk of cerebral white matter changes compared with individuals without OSA.62 Another study of 20 patients with severe OSA compared with 40 healthy volunteers found diffusion imaging consistent with impaired fibrin integrity in those with OSA, indicative of white matter microstructure damage, and the impairment was associated with increased disease severity and higher systemic inflammation.63

Individuals with hypoxia for a high percentage of time during sleep had a 4 times higher odds of cerebral microinfarcts.64 Cognitive scores decreased less in men. Men typically have more time in slow-wave sleep, suggesting that slow-wave sleep may be protective against cognitive decline. Mild cognitive impairment and Alzheimer disease were found more likely to develop and occur at an earlier age in individuals with sleep-disordered breathing compared with individuals without sleep-disordered breathing.65

OSA was also associated with increased serum amyloid beta levels in a study of 45 cognitively normal patients with OSA compared with 49 age- and sex-matched control patients. Increased amyloid beta levels correlated with increasing severity of sleep apnea as measured by the AHI.66

Mechanism linking OSA and cognition

One possible mechanism linking sleep quality and cognitive impairment or Alzheimer disease is the role of unfragmented sleep in attenuating the apolipo­protein E e4 allele on the incidence of Alzheimer disease.67 Beta amyloid is released during synaptic activity. Neuronal and synaptic activity decreases during sleep, and disrupted sleep could increase beta amyloid release.68 Sleep has been found to enhance the clearance of beta amyloid peptide from the brain interstitial fluid in a mice model.69

Recent data point toward the bidirectional relationship between the sleep and Alzheimer disease in that excessive and prolonged neuronal activity in the absence of appropriately structured sleep may be the reason for both Alzheimer disease and OSA.70,71

Effect of treatment for OSA on cognition

White matter integrity in 15 patients with OSA before and after treatment with CPAP was compared with 15 matched controls. Over 12 months, there was a nearly complete reversal of white matter abnormalities in patients on CPAP therapy.72 Improvement in memory, attention, and executive function paralleled the changes in white matter after the treatment.

CONCLUSION

OSA is a serious condition with far-reaching consequences associated with impaired quality of life, depressive symptoms, drowsy driving, metabolic disease, and cognitive decline. Treatment of OSA improves many of these health consequences, emphasizing the need for OSA screening. Large randomized studies are needed to assess the efficacy of CPAP on metabolic outcomes, including insulin sensitivity and glucose tolerance, in reducing disease burden. Study of the endophenotypes of patients with OSA is needed to define and target the mechanisms of OSA-induced adverse health outcomes and perhaps lead to personalized care for patients with OSA.

Obstructive sleep apnea (OSA) is a serious condition that impacts quality of life and causes drowsy driving and depression. Research also reveals an interrelationship between OSA and metabolic disease and an association between OSA and cognitive impairment.

The diagnostic criteria of OSA are based on the number of apneic or hypopneic episodes per hour of sleep, called the apnea–hypopnea index (AHI) as recorded during sleep testing. Diagnosis of OSA is warranted if the AHI is 15 or more per hour or if the AHI is 5 or more per hour with 1 or more of the following features: sleepiness; nonrestorative sleep; fatigue or insomnia; waking up with breath-holding spells, gasping, or choking; snoring or breathing interruptions; or a coexisting diagnosis of hypertension, mood disorder, cognitive dysfunction, coronary artery disease, stroke, congestive heart failure, atrial fibrillation, or type II diabetes.1

Many patients with an AHI less than 15 may also have OSA, given the number of coexisting medical conditions included in the OSA diagnostic criteria. Heart conditions such as coronary artery disease, atrial fibrillation, and congestive heart failure encompassed in the OSA diagnostic criteria have increased awareness of the link between OSA and heart health. Less well-known, and the subject of this review, are the negative consequences of OSA, particularly poor quality of life, drowsy driving, depression, metabolic disease, and cognitive impairment.

QUALITY OF LIFE

Reduced quality of life is the most fundamental patient-reported outcome of OSA. OSA is associated with excessive daytime sleepiness, inattention, and fatigue, which increase the risk of accidents and medical disability. These quality-of-life impairments are often the main reason patients seek medical care for sleep disorders.2 Improved quality of life is a central goal of OSA treatment and is the best indicator of the effectiveness of treatment.3 Sleep health and its effect on quality of life is an area of focus of Healthy People 2020 (healthypeople.gov).

The American Academy of Sleep Medicine identified quality of life, along with detection of disease and cardiovascular consequences, as an outcome measure for assessing the quality of care for adults with OSA.2 The assessment of quality of life for patients with OSA is a 4-part process: use evidence-based therapy, monitor the therapy, assess symptoms with a validated tool such as Epworth Sleepiness Scale, and assess the incidence of motor vehicle accidents. Information from these 4 processes can inform changes in a patient’s quality of life.

Treatment for OSA has been shown to improve quality of life. A study of 2,027 patients with OSA evaluated therapy adherence relative to mean Functional Outcomes of Sleep Questionnaire and European Quality of Life-5D scores.4 In patients with the most impaired quality of life, those adherent to positive airway pressure (PAP) therapy had improved quality of life as measured by these scores.

With respect to sleepiness, a systematic review of continuous PAP (CPAP) in patients with OSA found a 2.7-point reduction (mean difference; 95% confidence interval 3.45–1.96) in the Epworth Sleepiness Scale in patients using CPAP compared with the control group.5 Treatment of OSA improves patient quality of life and symptoms such as sleepiness.

DROWSY DRIVING

Drowsy driving by people with OSA can lead to motor vehicle accidents, which result in economic and health burdens.6,7 National Center for Statistics and Analysis data reveal that of 6 million motor vehicle accidents (5-year average, 2005–2009), 1.4% involve drowsy driving and 2.5% of fatal crashes involve drowsy driving.8 Among noncommercial drivers, untreated OSA increases the risk of motor vehicle accidents 3- to 13-fold.9 The odds ratio of traffic accidents in drivers with untreated OSA is 6 times greater than in the general population.7

In a study of men and women in the general population (N = 913), individuals with moderate to severe OSA (AHI > 15) were more likely to have multiple motor vehicle accidents in the course of 5 years (odds ratio = 7.3) compared with those with no sleep-disordered breathing.10 The association between OSA and motor vehicle accidents is independent of sleepiness, and drivers with OSA may not perceive performance impairment.

There are 2 main reasons OSA increases the risk and incidence of motor vehicle accidents. OSA causes changes in attention and vigilance resulting from sleep deprivation and fragmentation. OSA also affects global cognition function, which may be due to intermittent hypoxia attributable to OSA.2

Treatment for OSA is effective in reducing the incidence of motor vehicle accidents. One study found the risk of motor vehicle accidents was eliminated with the use of CPAP treatment in patients with OSA.11 A recent study of nearly 2,000 patients with OSA found a reduction in self-reported near-accidents from 14% before PAP therapy to 5.3% after starting PAP therapy.12

 

 

DEPRESSION

walia_osaconsequences_t1.jpg
Depression can occur as a consequence of OSA. Depression and OSA have several symptoms in common (Table 1).13

Estimates of the prevalence of depression in patients with OSA range from 5% to 63%.13,14 One year after patients were diagnosed with OSA, the incidence of depression per 1,000 person-years was 18% compared with 8% in a group without OSA.14 Women with OSA reportedly have a higher risk of depression (adjusted hazard ratio [HR] 2.7) than men (HR 1.81) at 1-year follow-up.14 In the same study, with respect to age, there was no significant relationship noted between OSA and patients over age 64.

A one-level increase in the severity of OSA (ie, from minimal to mild) is associated with a nearly twofold increase in the adjusted odds for depression.15 On the other hand, studies have also found that patients on antidepressants may have an increased risk of OSA.16

Several potential mechanisms have been proposed to explain the link between depression and OSA.13 Poor-quality sleep, frequent arousal, and fragmentation of sleep in OSA may lead to frontal lobe emotional modulation changes. Intermittent hypoxia in OSA may result in neuronal injury and disruption of noradrenergic and dopaminergic pathways. Pro-inflammatory substances such as interleukin 6 and interleukin 1 are increased in OSA and depression and are mediators between both conditions. Neurotransmitters may be affected by a disrupted sleep-wake cycle. And serotonin, which may be impeded in depression, could influence the upper-airway dilator motor neurons.

Treatment of OSA improves symptoms of depression as measured by the Patient Health Questionnaire (PHQ-9). After 3 months of compliance with CPAP therapy, mean PHQ-9 scores decreased from 11.3 to 2.7 in a study of 228 patients with OSA.17 A study of 1,981 patients with sleep-disordered breathing found improved PHQ-9 scores in patients compliant with CPAP therapy and a greater improvement in patients with a baseline PHQ-9 higher than 10 (moderate severity).18

METABOLIC SYNDROME

OSA is associated with metabolic disorders, including metabolic syndrome, though the causality between these 2 conditions is yet to be illuminated. Metabolic syndrome is a term used when an individual has 3 or more of the following features or conditions:

  • Waist circumference greater than 40 inches (men), greater than 35 inches (women)
  • Triglycerides 150 mg/dL or greater or treatment for hypertriglyceridemia
  • High-density lipoprotein cholesterol less than 40 mg/dL (men), less than 50 mg/dL (women), or treatment for cholesterol
  • Blood pressure 130/85 mm Hg or greater, or treatment for hypertension
  • Fasting blood glucose 100 mg/dL or greater, or treatment for hyperglycemia.19

Metabolic syndrome increases an individual’s risk of diabetes and cardiovascular disease and overall mortality. Like OSA, the prevalence of metabolic syndrome increases with age in both men and women.20,21 The risk of metabolic syndrome is greater with more severe OSA. The Wisconsin Sleep Cohort (N = 546) reported an odds ratio for having metabolic syndrome of 2.5 for patients with mild OSA and 2.2 for patients with moderate or severe OSA.22 A meta-analysis also found a 2.4 times higher odds of metabolic syndrome in patients with mild OSA, but a 3.5 times higher odds of metabolic syndrome in patients with moderate to severe OSA compared with the control group.23

Patients with both OSA and metabolic syndrome are said to have syndrome Z24 and are at increased risk of cardiovascular morbidity and mortality.25 Syndrome Z imparts a higher risk of atherogenic burden and prevalence of atheroma compared with patients with either condition alone.26 In comparing patients with metabolic syndrome with and without OSA, those with OSA had increased atherosclerotic burden as measured by intima-media thickness and carotid femoral pulse-wave velocity.27 Syndrome Z is also linked to intracoronary stenosis related to changes in cardiac morphology28 and is associated with left ventricular hypertrophy and diastolic dysfunction.29

OSA and hypertension

Hypertension is one of the conditions encompassed in metabolic syndrome. Several studies report increased risk and incidence of hypertension in patients with OSA. In a community-based study of 6,123 individuals age 40 and older, sleep-disordered breathing was associated with hypertension, and the odds ratio of hypertension was greater in individuals with more severe sleep apnea.30 Similarly, a landmark prospective, population-based study of 709 individuals over 4 years reported a dose-response relationship between patients with OSA and newly diagnosed hypertension independent of confounding factors.31 Patients with moderate to severe OSA had an odds ratio of 2.89 of developing hypertension after adjusting for confounding variables.

A study of 1,889 individuals followed for 12 years found a dose-response relationship based on OSA severity for developing hypertension.32 This study also assessed the incidence of hypertension based on CPAP use. Patients with poor adherence to CPAP use had an 80% increased incidence of hypertension, whereas patients adhering to CPAP use had a 30% decrease in the incidence of hypertension.

Resistant hypertension (ie, uncontrolled hypertension despite use of 3 or more antihypertensive and diuretic medications) has been shown to be highly prevalent (85%) in patients with severe OSA.33 An analysis of patients at increased risk of cardiovascular disease and untreated severe OSA was associated with a 4 times higher risk of elevated blood pressure despite intensive medical therapy.34

 

 

Mechanisms of altered metabolic regulation in OSA

Mechanisms implicated in altering metabolic regulation in OSA include intermittent hypoxia, sleep fragmentation and glucose homeostasis, and obesity. Intermittent hypoxia from OSA results in sympathetic nervous system activation that affects the pancreas, skeletal muscle, liver, and fat cells resulting in altered insulin secretion, lipid-bile synthesis, glucose metabolism, and lipoprotein metabolism.35

Sleep fragmentation is a cardinal feature of OSA and the resulting suppression of sleep may alter insulin sensitivity. Studies have implicated disruptions to slow-wave sleep specifically, as well as disruption of any stage of sleep in reduced insulin sensitivity.35,36 In addition to decreased insulin sensitivity, sleep fragmentation also increases morning cortisol levels and increases sympathetic nervous system activation.37

Obesity and OSA share a pathway imparting increased cardiometabolic risk.38 Fat tissue causes higher systemic inflammation and inflammatory markers. A recent report describes a bidirectional relationship between metabolic syndrome and OSA.39 While OSA increases the risk for metabolic syndrome, metabolic syndrome by virtue of body mass index with changes in mechanical load and narrow airway and physiology can predispose for OSA.

Effect of treatment for OSA on metabolic syndrome

Several studies have evaluated the effect of CPAP treatment for OSA on metabolic syndrome overall, as well as the specific conditions that comprise metabolic syndrome. In evaluating CPAP use and metabolic syndrome overall, studies have found a reduced prevalence of metabolic syndrome,40,41 CPAP benefit only in complying patients,42 and a reduction in oxidative stress with a single-night use of CPAP.43

With respect to insulin sensitivity, a study of 40 men with moderate OSA using CPAP therapy (mean use 5 hours) reported an increase in the insulin sensitivity index after 2 days, and a further increase after 3 months.44 Another study found no improvement in insulin resistance in severe OSA.45 A meta-analysis reported improved insulin resistance with CPAP,46 although a recent meta-analysis assessing hemoglobin A1c level, fasting insulin level, and fasting glucose did not show improvement in these parameters. Large-scale clinical trials with longer treatment duration and better CPAP compliance are warranted.47

walia_osaconsequences_t2.jpg
CPAP use in patients with OSA has been found to affect hypertension in a number of studies (Table 2).48–55 In a comparison of therapeutic CPAP with suboptimal CPAP for 9 weeks, ambulatory blood pressure was reduced in the therapeutic group, and no change was seen in the subtherapeutic group, illustrating the importance of optimal pressure settings in treating OSA.48

walia_osaconsequences_f1.jpg
%3Cp%3EFigure%201.%20Blood%20pressure%20trends%20in%20patients%20before%20and%20after%20continuous%20positive%20airway%20pressure%20(CPAP)%20use.%3C%2Fp%3E
A randomized controlled trial of nearly 300 individuals found improvement in 6 blood pressure parameters in a group using CPAP compared with a group using sham CPAP after 12 weeks.50 A large clinic-based cohort of 894 individuals with hypertension and resistant hypertension (15%) found that after 1 year, CPAP use was associated with 2 to 3 mm Hg of reduction in blood pressure (Figure 1).56 Meta-analysis of randomized controlled trials on the effectiveness of CPAP on hypertension found reductions of 2 mm Hg to 3 mm Hg in blood pressure.57 Another meta-analyses showed a reduction of 2.6 mm Hg in 24-hour mean blood pressure with CPAP therapy (Table 2).48–55 This reduction may appear modest in nature; however, any reduction in blood pressure can result in decreased cardiovascular morbidity and mortality. A meta-analysis of randomized controlled trials indicated reductions in mean systolic blood pressure of 5.4 mm Hg and diastolic blood pressure of 3.86 mm Hg after CPAP in those with resistant hypertension and OSA.58

Weight loss has been shown to reduce the AHI and other parameters related to sleep apnea such as oxygen desaturation index in patients with obesity and diabetes.59 Weight loss combined with CPAP compared with CPAP or weight loss alone showed an incremental benefit in improving glucose parameters, triglycerides, and possibly systolic blood pressure and triglycerides.60

 

 

COGNITIVE IMPAIRMENT

Data suggest that OSA is linked with cognitive impairment, may advance cognitive decline, and is a bidirectional relationship. Women with OSA were reportedly more likely to develop mild cognitive impairment compared with women without OSA.61 An elevated oxygen desaturation index and a high percentage of time spent with hypoxia was associated with increased risk of developing mild cognitive impairment and dementia.

OSA was found to be an independent risk factor for cerebral white matter changes in middle-age and older individuals. Moderate to severe OSA imparted a 2 times higher risk of cerebral white matter changes compared with individuals without OSA.62 Another study of 20 patients with severe OSA compared with 40 healthy volunteers found diffusion imaging consistent with impaired fibrin integrity in those with OSA, indicative of white matter microstructure damage, and the impairment was associated with increased disease severity and higher systemic inflammation.63

Individuals with hypoxia for a high percentage of time during sleep had a 4 times higher odds of cerebral microinfarcts.64 Cognitive scores decreased less in men. Men typically have more time in slow-wave sleep, suggesting that slow-wave sleep may be protective against cognitive decline. Mild cognitive impairment and Alzheimer disease were found more likely to develop and occur at an earlier age in individuals with sleep-disordered breathing compared with individuals without sleep-disordered breathing.65

OSA was also associated with increased serum amyloid beta levels in a study of 45 cognitively normal patients with OSA compared with 49 age- and sex-matched control patients. Increased amyloid beta levels correlated with increasing severity of sleep apnea as measured by the AHI.66

Mechanism linking OSA and cognition

One possible mechanism linking sleep quality and cognitive impairment or Alzheimer disease is the role of unfragmented sleep in attenuating the apolipo­protein E e4 allele on the incidence of Alzheimer disease.67 Beta amyloid is released during synaptic activity. Neuronal and synaptic activity decreases during sleep, and disrupted sleep could increase beta amyloid release.68 Sleep has been found to enhance the clearance of beta amyloid peptide from the brain interstitial fluid in a mice model.69

Recent data point toward the bidirectional relationship between the sleep and Alzheimer disease in that excessive and prolonged neuronal activity in the absence of appropriately structured sleep may be the reason for both Alzheimer disease and OSA.70,71

Effect of treatment for OSA on cognition

White matter integrity in 15 patients with OSA before and after treatment with CPAP was compared with 15 matched controls. Over 12 months, there was a nearly complete reversal of white matter abnormalities in patients on CPAP therapy.72 Improvement in memory, attention, and executive function paralleled the changes in white matter after the treatment.

CONCLUSION

OSA is a serious condition with far-reaching consequences associated with impaired quality of life, depressive symptoms, drowsy driving, metabolic disease, and cognitive decline. Treatment of OSA improves many of these health consequences, emphasizing the need for OSA screening. Large randomized studies are needed to assess the efficacy of CPAP on metabolic outcomes, including insulin sensitivity and glucose tolerance, in reducing disease burden. Study of the endophenotypes of patients with OSA is needed to define and target the mechanisms of OSA-induced adverse health outcomes and perhaps lead to personalized care for patients with OSA.

References
  1. Sateia MJ. International Classification of Sleep Disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  2. Aurora RN, Collop NA, Jacobowitz O, Thomas SM, Quan SF, Aronsky AJ. Quality measures for the care of adult patients with obstructive sleep apnea. J Clin Sleep Med 2015; 11(3):357–383.
  3. Flemons WW. Obstructive sleep apnea. N Engl J Med 2002; 347(7): 498–504.
  4. Walia HK, Thompson NR, Katzan I, Foldvary-Schaefer N, Moul DE, Mehra R. Impact of sleep-disordered breathing treatment on quality of life measures in a large clinic-based cohort. J Clin Sleep Med; 2017;13(11):1255–1263. doi:10.5664/jcsm.6792
  5. McDaid C, Dureé KH, Griffin SC, et al. A systematic review of continuous positive airway pressure for obstructive sleep apnoea–hypopnoea syndrome. Sleep Med Rev 2009; 13(6):427–436.
  6. Arita A, Sasanabe R, Hasegawa R, et al. Risk factors for auto­mobile accidents caused by falling asleep while driving in obstructive sleep apnea syndrome. Sleep Breath 2015; 19(4):1229–1234.
  7. Terán-Santos J, Jiménez-Gómez A, Cordero-Guevara J; the Cooperative Group Burgos–Santander. The association between sleep apnea and the risk of traffic accidents. N Engl J Med 1999; 340(11):847–851.
  8. NHTSA. Drowsy driving. Washington, DC: National Highway Traffic Safety Administration. https://crashstats.nhtsa.dot.gov/Api/Public/ViewPublication/811449. Published March 2011. Accessed August 19, 2019.
  9. Vakulin A, D’Rozario A, Kim J-W, et al. Quantitative sleep EEG and polysomnographic predictors of driving simulator performance in obstructive sleep apnea. Clin Neurophysiol 2016; 127(2):1428–1435.
  10. Young T, Blustein J, Finn L, Palta M. Sleep-disordered breathing and motor vehicle accidents in a population-based sample of employed adults. Sleep 1997; 20(8):608–613.
  11. George CFP. Reduction in motor vehicle collisions following treatment of sleep apnoea with nasal CPAP. Thorax 2001; 56(7):508–512.
  12. Walia HK, Thompson N, Pascoe M, et al. Impact of positive airway pressure therapy on drowsy driving in a large clinic-based obstructive sleep apnea cohort. J Clin Sleep Med (in press).
  13. Ejaz SM, Khawaja IS, Bhatia S, Hurwitz TD. Obstructive sleep apnea and depression: a review. Innov Clin Neurosci 2011; 8(8):17–25.
  14. Chen Y-H, Keller JK, Kang J-H, Hsieh H-J, Lin H-C. Obstructive sleep apnea and the subsequent risk of depressive disorder: a population-based follow-up study. J Clin Sleep Med 2013; 9(5):417–423.
  15. Peppard PE, Szklo-Coxe M, Hla KM, Young T. Longitudinal association of sleep-related breathing disorder and depression. Arch Intern Med 2006; 166(16):1709–1715.
  16. Farney RJ, Lugo A, Jensen RL, Walker JM, Cloward TV. Simultaneous use of antidepressant and antihypertensive medications increases likelihood of diagnosis of obstructive sleep apnea syndrome. Chest; 2004;125(4):1279–1285.
  17. Edwards C, Mukherjee S, Simpson L, Palmer LJ, Almeida OP, Hillman DR. Depressive symptoms before and after treatment of obstructive sleep apnea in men and women. J Clin Sleep Med 2015; 11(9):1029–1038.
  18. Relia S, Thompson NR, Mehra R, et al. Depression score changes in response to sleep disordered breathing treatment with positive airway pressure in a large clinic-based cohort. Sleep Breath 2018; 22(1):195–203.
  19. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002; 106(25):3143–3421.
  20. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the Third National Health and Nutrition Examination Survey. JAMA 2002; 287(3):356–359.
  21. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  22. Nieto FJ, Peppard PE, Young TB. Sleep disordered breathing and metabolic syndrome. WMJ 2009; 108(5):263–265.
  23. Xu S, Wan Y, Xu M, et al. The association between obstructive sleep apnea and metabolic syndrome: a systematic review and meta-analysis. BMC Pulm Med 2015; 15:105.
  24. Nock NL, Li L, Larkin EK, Patel SR, Redline S. Empirical evidence for “syndrome Z”: a hierarchical 5-factor model of the metabolic syndrome incorporating sleep disturbance measures. Sleep 2009; 32(5):615–622.
  25. Sadasivam K, Chinnasami B, Ayyavo S, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diagn Res 2015; 9(4):CC07–CC10.
  26. Chang TI, Tanner JM, Harada ND, Garrett NR, Friedlander AH. Prevalence of calcified carotid artery atheromas on the panoramic images of patients with syndrome Z, coexisting obstructive sleep apnea, and metabolic syndrome. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113(1):134–141.
  27. Drager LF, Bortolotto LA, Maki-Nunes C, et al. The incremental role of obstructive sleep apnoea on markers of atherosclerosis in patients with metabolic syndrome. Atherosclerosis 2010; 208(2):490–495.
  28. Nakanishi-Minami T, Kishida K, Nakagawa Y, et al. Metabolic syndrome correlates intracoronary stenosis detected by multislice computed tomography in male subjects with sleep-disordered breathing. Diabetol Metab Syndr 2012; 4:6.
  29. Usui Y, Takata Y, Inoue Y, et al. Coexistence of obstructive sleep apnoea and metabolic syndrome is independently associated with left ventricular hypertrophy and diastolic dysfunction. Sleep Breath 2012; 16(3):677–684.
  30. Nieto FJ, Young TB, Lind BK, et al; for the Sleep Heart Health Study. Association of sleep-disordered breathing, sleep apnea, and hypertension in a large community-based study. JAMA 2000; 283(14):1829–1836.
  31. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of the association between sleep-disordered breathing and hypertension. N Engl J Med 2000; 342(19):1378–1384.
  32. Marin JM, Agusti A, Villar I, et al. Association between treated and untreated obstructive sleep apnea and risk of hypertension. JAMA 2012; 307(20):2169–2176.
  33. Gonçalves SC, Martinez D, Gus M, et al. Obstructive sleep apnea and resistant hypertension: a case-control study. Chest 2007; 132(6):1858–1862.
  34. Walia HK, Li H, Rueschman M, et al. Association of severe obstructive sleep apnea and elevated blood pressure despite anti­hypertensive medication use. J Clin Sleep Med 2014; 10(8):835–843.
  35. Braincon-Marjollet A, Weiszenstein M, Henri M, Thomas A, Godin-Ribuot D, Polak J. The impact of sleep disorders on glucose metabolism: endocrine and molecular mechanisms. Diabetol Metab Syndr 2015; 7:25. doi:10.1186/s13098-015-0018-3
  36. Stamatakis KA, Punjabi NM. Effects of sleep fragmentation on glucose metabolism in normal subjects. Chest 2010; 137(1):95–101.
  37. Spiegel K. Knutson K, Leproult R, Tasali E, Van Cauter E. Sleep loss: a novel risk factor for insulin resistance and type 2 diabetes. J Appl Physiol (1985) 2005; 99(5):2008–2019.
  38. Pépin J-L, Tamisier R, Lévy P. Obstructive sleep apnoea and metabolic syndrome: put CPAP efficacy in a more realistic perspective. Thorax 2012; 67(12):1025–1027.
  39. Framnes SN, Arble DM. The bidirectional relationship between obstructive sleep apnea and metabolic disease. Front Endocrinol (Lausanne) 2018; 9:440.
  40. Oktay B, Akbal E, Firat H, Ardiç S, Kizilgun M. CPAP treatment in the coexistence of obstructive sleep apnea syndrome and metabolic syndrome, results of one year follow up. Acta Clin Belg 2009; 64(4):329–334.
  41. Mota PC, Drummond M, Winck JC, Santos AC, Almeida J, Marques JA. APAP impact on metabolic syndrome in obstructive sleep apnea patients. Sleep Breath 2011; 15(4):665–672.
  42. Dorkova Z, Petrasova D, Molcanyiova A, Popovnakova M, Tkacova R. Effects of continuous positive airway pressure on cardio­vascular risk profile in patients with severe obstructive sleep apnea and metabolic syndrome. Chest 2008; 134(4):686–692.
  43. Kanimozhi S, Balaji C, Saravanan A, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diag Research 2015; 9(4):CC07–CC10.
  44. Harsch IA, Schahin SP, Radespiel-Tröger M, et al. Continuous positive airway pressure treatment rapidly improves insulin sensitivity in patients with obstructive sleep apnea syndrome. Am J Respir Crit Care Med 2004; 169(2):156–162.
  45. Trenell MI, Ward JA, Yee BJ, et al. Influence of constant positive airway pressure therapy on lipid storage, muscle metabolism and insulin action in obese patients with severe obstructive sleep apnoea syndrome. Diabetes Obes Metab 2007; 9(5):679–687.
  46. Iftikhar IH, Hoyos CM, Phillips CL, Magalang UJ. Meta-analysis of the association of sleep apnea with insulin resistance, and the effects of CPAP on HOMA-IR, adiponectin, and visceral adipose fat. J Clin Sleep Med 2015; 11(4):475–485.
  47. Zhu B, Ma C, Chaiard J, Shi C. Effect of continuous positive airway pressure on glucose metabolism in adults with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Sleep Breath 2018; 22(2):287–295.
  48. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  49. Campos-Rodriguez F, Grilo-Reina A, Perez-Ronchel J, et al. Effect of continuous positive airway pressure on ambulatory BP in patients with sleep apnea and hypertension: a placebo-controlled trial. Chest 2006; 129(6):1459–1467.
  50. Durán-Cantolla J, Aizpuru F, Montserrat JM, et al; on behalf of the Spanish Sleep and Breathing Group. Continuous positive airway pressure as treatment for systemic hypertension in people with obstructive sleep apnoea: randomised controlled trial. BMJ 2010; 341:c5991.
  51. Gottlieb DJ, Punjabi NM, Mehra R, et al. CPAP versus oxygen in obstructive sleep apnea. N Engl J Med 2014; 370(24):2276–2285.
  52. Hui DS, To KW, Ko FW, et al. Nasal CPAP reduces systemic blood pressure in patients with obstructive sleep apnoea and mild sleepiness. Thorax 2006; 61(12):1083–1090.
  53. Martinez-Garcia MA, Capote F, Campos-Rodriguez F, et al. Effect of CPAP on blood pressure in patients with obstructive sleep apnea and resistant hypertension: the HIPARCO randomized clinical trial. JAMA 2013; 310(22):2407–2415.
  54. Pepperell JC, Ramdassingh-Dow S, Crosthwaite N, et al. Ambulatory blood pressure after therapeutic and subtherapeutic nasal continuous positive airway pressure for obstructive sleep apnoea: a randomised parallel trial. Lancet 2002; 359(9302):204–210.
  55. Robinson GV, Smith DM, Langford BA, Davies RJ, Stradling JR. Continuous positive airway pressure does not reduce blood pressure in nonsleepy hypertensive OSA patients. Eur Respir J 2006; 27(6):1229–1235.
  56. Walia HK, Griffith SD, Foldvary-Schaefer N, et al. Longitudinal effect of CPAP on BP in resistant and nonresistant hypertension in a large clinic-based cohort. Chest 2016; 149(3):747–755.
  57. Montesi SB, Edwards BA, Malhotra A, Bakker JP. The effect of continuous positive airway pressure treatment on blood pressure: a systematic review and meta-analysis of randomized controlled trials. J Clin Sleep Med 2012; 8(5):587–596.
  58. Lei Q, Lv Y, Li K, Ma L, Du G, Xiang Y, Li X. Effects of continuous positive airway pressure on blood pressure in patients with resistant hypertension and obstructive sleep apnea: a systematic review and meta-analysis of six randomized controlled trials. J Bras Pneumol 2017;43(5):373–379. doi:10.1590/S1806-37562016000000190. [Article in English, Portuguese]
  59. Foster GD, Borradaile KE, Sanders MH; for the Sleep AHEAD Research Group of Look AHEAD Research Group. A randomized study on the effect of weight loss on obstructive sleep apnea among obese patients with type 2 diabetes: the Sleep AHEAD study. Arch Intern Med 2009; 169(17):1619–1626.
  60. Chirinos JA, Gurubhagavatula I, Teff K, et al. CPAP, weight loss, or both for obstructive sleep apnea. N Engl J Med 2014; 370(24):2265–2275.
  61. Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306(6):613–619.
  62. Kim H, Yun C-H, Thomas RJ, et al. Obstructive sleep apnea as a risk factor for cerebral white matter change in a middle-aged and older general population. Sleep 2013; 36(5):709–715B.
  63. Chen H-L, Lu C-H, Lin H-C, et al. White matter damage and systemic inflammation in obstructive sleep apnea. Sleep 2015; 38(3):361–370.
  64. Gelber RP, Redline S, Ross GW, et al. Associations of brain lesions at autopsy with polysomnography features before death. Neurology 2015; 84(3):296–303.
  65. Osorio RS, Gumb T, Pirraglia E, et al; for the Alzheimer’s Disease Neuroimaging Initiative. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology 2015; 84(19):1964–1971.
  66. Bu X-L, Liu Y-H, Wang Q-H, et al. Serum amyloid-beta levels are increased in patients with obstructive sleep apnea syndrome. Sci Rep 2015; 5:13917.
  67. Lim ASP, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the relationship of the apolipoprotein e 4 allele to the risk of Alzheimer disease and neurofibrillary tangle density by sleep. JAMA Neurol 2013; 70(12):1544–1551.
  68. Lucey BP, Bateman RJ. Amyloid-beta diurnal pattern: possible role of sleep in Alzheimer’s disease pathogenesis. Neurobiol Aging 2014; 35(suppl 2):S29–S34.
  69. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science 2013; 342(6156):373–377.
  70. Polsek D, Gildeh N, Cash D, et al. Obstructive sleep apnoea and Alzheimer’s disease: in search of shared pathomechanisms. Neurosci Biobehav Rev 2018; 86:142–149.
  71. Ju Y-ES, Lucey BP, Holtzman DM. Sleep and Alzheimer disease pathology—a bidirectional relationship. Nat Rev Neurol 2014; 10(2):115–119.
  72. Castronovo V, Scifo P, Castellano A, et al. White matter integrity in obstructive sleep apnea before and after treatment. Sleep 2014; 37(9):1465–1475.
References
  1. Sateia MJ. International Classification of Sleep Disorders—3rd ed: highlights and modifications. Chest 2014; 146(5):1387–1394.
  2. Aurora RN, Collop NA, Jacobowitz O, Thomas SM, Quan SF, Aronsky AJ. Quality measures for the care of adult patients with obstructive sleep apnea. J Clin Sleep Med 2015; 11(3):357–383.
  3. Flemons WW. Obstructive sleep apnea. N Engl J Med 2002; 347(7): 498–504.
  4. Walia HK, Thompson NR, Katzan I, Foldvary-Schaefer N, Moul DE, Mehra R. Impact of sleep-disordered breathing treatment on quality of life measures in a large clinic-based cohort. J Clin Sleep Med; 2017;13(11):1255–1263. doi:10.5664/jcsm.6792
  5. McDaid C, Dureé KH, Griffin SC, et al. A systematic review of continuous positive airway pressure for obstructive sleep apnoea–hypopnoea syndrome. Sleep Med Rev 2009; 13(6):427–436.
  6. Arita A, Sasanabe R, Hasegawa R, et al. Risk factors for auto­mobile accidents caused by falling asleep while driving in obstructive sleep apnea syndrome. Sleep Breath 2015; 19(4):1229–1234.
  7. Terán-Santos J, Jiménez-Gómez A, Cordero-Guevara J; the Cooperative Group Burgos–Santander. The association between sleep apnea and the risk of traffic accidents. N Engl J Med 1999; 340(11):847–851.
  8. NHTSA. Drowsy driving. Washington, DC: National Highway Traffic Safety Administration. https://crashstats.nhtsa.dot.gov/Api/Public/ViewPublication/811449. Published March 2011. Accessed August 19, 2019.
  9. Vakulin A, D’Rozario A, Kim J-W, et al. Quantitative sleep EEG and polysomnographic predictors of driving simulator performance in obstructive sleep apnea. Clin Neurophysiol 2016; 127(2):1428–1435.
  10. Young T, Blustein J, Finn L, Palta M. Sleep-disordered breathing and motor vehicle accidents in a population-based sample of employed adults. Sleep 1997; 20(8):608–613.
  11. George CFP. Reduction in motor vehicle collisions following treatment of sleep apnoea with nasal CPAP. Thorax 2001; 56(7):508–512.
  12. Walia HK, Thompson N, Pascoe M, et al. Impact of positive airway pressure therapy on drowsy driving in a large clinic-based obstructive sleep apnea cohort. J Clin Sleep Med (in press).
  13. Ejaz SM, Khawaja IS, Bhatia S, Hurwitz TD. Obstructive sleep apnea and depression: a review. Innov Clin Neurosci 2011; 8(8):17–25.
  14. Chen Y-H, Keller JK, Kang J-H, Hsieh H-J, Lin H-C. Obstructive sleep apnea and the subsequent risk of depressive disorder: a population-based follow-up study. J Clin Sleep Med 2013; 9(5):417–423.
  15. Peppard PE, Szklo-Coxe M, Hla KM, Young T. Longitudinal association of sleep-related breathing disorder and depression. Arch Intern Med 2006; 166(16):1709–1715.
  16. Farney RJ, Lugo A, Jensen RL, Walker JM, Cloward TV. Simultaneous use of antidepressant and antihypertensive medications increases likelihood of diagnosis of obstructive sleep apnea syndrome. Chest; 2004;125(4):1279–1285.
  17. Edwards C, Mukherjee S, Simpson L, Palmer LJ, Almeida OP, Hillman DR. Depressive symptoms before and after treatment of obstructive sleep apnea in men and women. J Clin Sleep Med 2015; 11(9):1029–1038.
  18. Relia S, Thompson NR, Mehra R, et al. Depression score changes in response to sleep disordered breathing treatment with positive airway pressure in a large clinic-based cohort. Sleep Breath 2018; 22(1):195–203.
  19. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002; 106(25):3143–3421.
  20. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the Third National Health and Nutrition Examination Survey. JAMA 2002; 287(3):356–359.
  21. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health perspective. Am J Respir Crit Care Med 2002; 165(9):1217–1239.
  22. Nieto FJ, Peppard PE, Young TB. Sleep disordered breathing and metabolic syndrome. WMJ 2009; 108(5):263–265.
  23. Xu S, Wan Y, Xu M, et al. The association between obstructive sleep apnea and metabolic syndrome: a systematic review and meta-analysis. BMC Pulm Med 2015; 15:105.
  24. Nock NL, Li L, Larkin EK, Patel SR, Redline S. Empirical evidence for “syndrome Z”: a hierarchical 5-factor model of the metabolic syndrome incorporating sleep disturbance measures. Sleep 2009; 32(5):615–622.
  25. Sadasivam K, Chinnasami B, Ayyavo S, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diagn Res 2015; 9(4):CC07–CC10.
  26. Chang TI, Tanner JM, Harada ND, Garrett NR, Friedlander AH. Prevalence of calcified carotid artery atheromas on the panoramic images of patients with syndrome Z, coexisting obstructive sleep apnea, and metabolic syndrome. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113(1):134–141.
  27. Drager LF, Bortolotto LA, Maki-Nunes C, et al. The incremental role of obstructive sleep apnoea on markers of atherosclerosis in patients with metabolic syndrome. Atherosclerosis 2010; 208(2):490–495.
  28. Nakanishi-Minami T, Kishida K, Nakagawa Y, et al. Metabolic syndrome correlates intracoronary stenosis detected by multislice computed tomography in male subjects with sleep-disordered breathing. Diabetol Metab Syndr 2012; 4:6.
  29. Usui Y, Takata Y, Inoue Y, et al. Coexistence of obstructive sleep apnoea and metabolic syndrome is independently associated with left ventricular hypertrophy and diastolic dysfunction. Sleep Breath 2012; 16(3):677–684.
  30. Nieto FJ, Young TB, Lind BK, et al; for the Sleep Heart Health Study. Association of sleep-disordered breathing, sleep apnea, and hypertension in a large community-based study. JAMA 2000; 283(14):1829–1836.
  31. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of the association between sleep-disordered breathing and hypertension. N Engl J Med 2000; 342(19):1378–1384.
  32. Marin JM, Agusti A, Villar I, et al. Association between treated and untreated obstructive sleep apnea and risk of hypertension. JAMA 2012; 307(20):2169–2176.
  33. Gonçalves SC, Martinez D, Gus M, et al. Obstructive sleep apnea and resistant hypertension: a case-control study. Chest 2007; 132(6):1858–1862.
  34. Walia HK, Li H, Rueschman M, et al. Association of severe obstructive sleep apnea and elevated blood pressure despite anti­hypertensive medication use. J Clin Sleep Med 2014; 10(8):835–843.
  35. Braincon-Marjollet A, Weiszenstein M, Henri M, Thomas A, Godin-Ribuot D, Polak J. The impact of sleep disorders on glucose metabolism: endocrine and molecular mechanisms. Diabetol Metab Syndr 2015; 7:25. doi:10.1186/s13098-015-0018-3
  36. Stamatakis KA, Punjabi NM. Effects of sleep fragmentation on glucose metabolism in normal subjects. Chest 2010; 137(1):95–101.
  37. Spiegel K. Knutson K, Leproult R, Tasali E, Van Cauter E. Sleep loss: a novel risk factor for insulin resistance and type 2 diabetes. J Appl Physiol (1985) 2005; 99(5):2008–2019.
  38. Pépin J-L, Tamisier R, Lévy P. Obstructive sleep apnoea and metabolic syndrome: put CPAP efficacy in a more realistic perspective. Thorax 2012; 67(12):1025–1027.
  39. Framnes SN, Arble DM. The bidirectional relationship between obstructive sleep apnea and metabolic disease. Front Endocrinol (Lausanne) 2018; 9:440.
  40. Oktay B, Akbal E, Firat H, Ardiç S, Kizilgun M. CPAP treatment in the coexistence of obstructive sleep apnea syndrome and metabolic syndrome, results of one year follow up. Acta Clin Belg 2009; 64(4):329–334.
  41. Mota PC, Drummond M, Winck JC, Santos AC, Almeida J, Marques JA. APAP impact on metabolic syndrome in obstructive sleep apnea patients. Sleep Breath 2011; 15(4):665–672.
  42. Dorkova Z, Petrasova D, Molcanyiova A, Popovnakova M, Tkacova R. Effects of continuous positive airway pressure on cardio­vascular risk profile in patients with severe obstructive sleep apnea and metabolic syndrome. Chest 2008; 134(4):686–692.
  43. Kanimozhi S, Balaji C, Saravanan A, Ravi K. Effect of short term CPAP therapy in obstructive sleep apnea patients with metabolic syndrome. J Clin Diag Research 2015; 9(4):CC07–CC10.
  44. Harsch IA, Schahin SP, Radespiel-Tröger M, et al. Continuous positive airway pressure treatment rapidly improves insulin sensitivity in patients with obstructive sleep apnea syndrome. Am J Respir Crit Care Med 2004; 169(2):156–162.
  45. Trenell MI, Ward JA, Yee BJ, et al. Influence of constant positive airway pressure therapy on lipid storage, muscle metabolism and insulin action in obese patients with severe obstructive sleep apnoea syndrome. Diabetes Obes Metab 2007; 9(5):679–687.
  46. Iftikhar IH, Hoyos CM, Phillips CL, Magalang UJ. Meta-analysis of the association of sleep apnea with insulin resistance, and the effects of CPAP on HOMA-IR, adiponectin, and visceral adipose fat. J Clin Sleep Med 2015; 11(4):475–485.
  47. Zhu B, Ma C, Chaiard J, Shi C. Effect of continuous positive airway pressure on glucose metabolism in adults with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Sleep Breath 2018; 22(2):287–295.
  48. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  49. Campos-Rodriguez F, Grilo-Reina A, Perez-Ronchel J, et al. Effect of continuous positive airway pressure on ambulatory BP in patients with sleep apnea and hypertension: a placebo-controlled trial. Chest 2006; 129(6):1459–1467.
  50. Durán-Cantolla J, Aizpuru F, Montserrat JM, et al; on behalf of the Spanish Sleep and Breathing Group. Continuous positive airway pressure as treatment for systemic hypertension in people with obstructive sleep apnoea: randomised controlled trial. BMJ 2010; 341:c5991.
  51. Gottlieb DJ, Punjabi NM, Mehra R, et al. CPAP versus oxygen in obstructive sleep apnea. N Engl J Med 2014; 370(24):2276–2285.
  52. Hui DS, To KW, Ko FW, et al. Nasal CPAP reduces systemic blood pressure in patients with obstructive sleep apnoea and mild sleepiness. Thorax 2006; 61(12):1083–1090.
  53. Martinez-Garcia MA, Capote F, Campos-Rodriguez F, et al. Effect of CPAP on blood pressure in patients with obstructive sleep apnea and resistant hypertension: the HIPARCO randomized clinical trial. JAMA 2013; 310(22):2407–2415.
  54. Pepperell JC, Ramdassingh-Dow S, Crosthwaite N, et al. Ambulatory blood pressure after therapeutic and subtherapeutic nasal continuous positive airway pressure for obstructive sleep apnoea: a randomised parallel trial. Lancet 2002; 359(9302):204–210.
  55. Robinson GV, Smith DM, Langford BA, Davies RJ, Stradling JR. Continuous positive airway pressure does not reduce blood pressure in nonsleepy hypertensive OSA patients. Eur Respir J 2006; 27(6):1229–1235.
  56. Walia HK, Griffith SD, Foldvary-Schaefer N, et al. Longitudinal effect of CPAP on BP in resistant and nonresistant hypertension in a large clinic-based cohort. Chest 2016; 149(3):747–755.
  57. Montesi SB, Edwards BA, Malhotra A, Bakker JP. The effect of continuous positive airway pressure treatment on blood pressure: a systematic review and meta-analysis of randomized controlled trials. J Clin Sleep Med 2012; 8(5):587–596.
  58. Lei Q, Lv Y, Li K, Ma L, Du G, Xiang Y, Li X. Effects of continuous positive airway pressure on blood pressure in patients with resistant hypertension and obstructive sleep apnea: a systematic review and meta-analysis of six randomized controlled trials. J Bras Pneumol 2017;43(5):373–379. doi:10.1590/S1806-37562016000000190. [Article in English, Portuguese]
  59. Foster GD, Borradaile KE, Sanders MH; for the Sleep AHEAD Research Group of Look AHEAD Research Group. A randomized study on the effect of weight loss on obstructive sleep apnea among obese patients with type 2 diabetes: the Sleep AHEAD study. Arch Intern Med 2009; 169(17):1619–1626.
  60. Chirinos JA, Gurubhagavatula I, Teff K, et al. CPAP, weight loss, or both for obstructive sleep apnea. N Engl J Med 2014; 370(24):2265–2275.
  61. Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306(6):613–619.
  62. Kim H, Yun C-H, Thomas RJ, et al. Obstructive sleep apnea as a risk factor for cerebral white matter change in a middle-aged and older general population. Sleep 2013; 36(5):709–715B.
  63. Chen H-L, Lu C-H, Lin H-C, et al. White matter damage and systemic inflammation in obstructive sleep apnea. Sleep 2015; 38(3):361–370.
  64. Gelber RP, Redline S, Ross GW, et al. Associations of brain lesions at autopsy with polysomnography features before death. Neurology 2015; 84(3):296–303.
  65. Osorio RS, Gumb T, Pirraglia E, et al; for the Alzheimer’s Disease Neuroimaging Initiative. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology 2015; 84(19):1964–1971.
  66. Bu X-L, Liu Y-H, Wang Q-H, et al. Serum amyloid-beta levels are increased in patients with obstructive sleep apnea syndrome. Sci Rep 2015; 5:13917.
  67. Lim ASP, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the relationship of the apolipoprotein e 4 allele to the risk of Alzheimer disease and neurofibrillary tangle density by sleep. JAMA Neurol 2013; 70(12):1544–1551.
  68. Lucey BP, Bateman RJ. Amyloid-beta diurnal pattern: possible role of sleep in Alzheimer’s disease pathogenesis. Neurobiol Aging 2014; 35(suppl 2):S29–S34.
  69. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science 2013; 342(6156):373–377.
  70. Polsek D, Gildeh N, Cash D, et al. Obstructive sleep apnoea and Alzheimer’s disease: in search of shared pathomechanisms. Neurosci Biobehav Rev 2018; 86:142–149.
  71. Ju Y-ES, Lucey BP, Holtzman DM. Sleep and Alzheimer disease pathology—a bidirectional relationship. Nat Rev Neurol 2014; 10(2):115–119.
  72. Castronovo V, Scifo P, Castellano A, et al. White matter integrity in obstructive sleep apnea before and after treatment. Sleep 2014; 37(9):1465–1475.
Page Number
19-25
Page Number
19-25
Publications
Publications
Topics
Article Type
Display Headline
Beyond heart health: Consequences of obstructive sleep apnea
Display Headline
Beyond heart health: Consequences of obstructive sleep apnea
Legacy Keywords
sleep apnea, obstructive sleep apnea, OSA, quality of life, depression, drowsy driving, metabolic syndrome, syndrome Z, hypertension, Alzheimer disease, cognitive decline, harneet walia, nancy foldvary-schaefer
Legacy Keywords
sleep apnea, obstructive sleep apnea, OSA, quality of life, depression, drowsy driving, metabolic syndrome, syndrome Z, hypertension, Alzheimer disease, cognitive decline, harneet walia, nancy foldvary-schaefer
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2019 September;86(9 suppl 1):19-25
Inside the Article

KEY POINTS

  • OSA is associated with negative health consequences such as depression, drowsy driving, metabolic disease, and cognitive decline.
  • Several possible mechanisms to explain the health consequences of OSA have been explored.
  • Treatment of patients with OSA may improve outcomes for many of the health consequences associated with OSA.
  • Screening for OSA is important to identify and treat patients to reduce the associated health risks.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 07:30
Un-Gate On Date
Wed, 08/28/2019 - 07:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 07:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Positive airway pressure: Making an impact on sleep apnea

Article Type
Changed
Tue, 09/03/2019 - 12:13
Display Headline
Positive airway pressure: Making an impact on sleep apnea

lance_papimpactonapnea_f1.jpg
%3Cp%3EFigure%201.%20Obstructed%20airway%20(left)%20is%20opened%20with%20a%20column%20of%20air%20delivered%20using%20positive%20airway%20pressure%20therapy%20(right).%3C%2Fp%3E
Positive airway pressure (PAP) therapy is used to open an obstructed upper airway (Figure 1). PAP therapy consists of a small bedside unit that creates a pressurized column of air that is delivered through tubing to a facial interface, which can be nasal, oral, or both. Collin Sullivan, MD, created the nasal continuous PAP (CPAP) in 1982 using parts of a vacuum cleaner to create positive pressure that successfully resolved hypoxemia in a patient.1 Today, the various forms of PAP therapy include CPAP, the most common, auto-PAP (APAP), and bilevel PAP (BiPAP).

EFFICACY OF PAP THERAPY

The American Academy of Sleep Medicine practice guidelines for PAP are based on 342 articles, most rated as evidence levels I and II, concluding that CPAP is superior to conservative treatment to:

  • Eliminate respiratory disturbances
  • Reduce the apnea–hypopnea index
  • Decrease the arousal index on electroencephalogram
  • Increase in the total amount of slow-wave or N3 sleep
  • Reduce daytime sleepiness.2

These practice parameters are based on evidence of improved daytime sleepiness and reduced incidence of cardiovascular events in patients with moderate to severe obstructive sleep apnea (OSA) treated with PAP. The evidence is less clear for neurocognitive markers and cardiovascular events in the treatment of patients with mild sleep apnea.

Sleepiness

A study evaluated sleepiness outcomes in 149 patients with severe sleep apnea with an average apnea–hypopnea index of 69 relative to the duration of nightly CPAP use. Sleepiness was measured using the Functional Outcomes of Sleep Questionnaire, Epworth Sleepiness Scale, and Multiple Sleep Latency Test. Results suggest that a greater percentage of patients had improved daytime sleepiness as the total hours of sleep using CPAP increased.3

The Apnea Positive Pressure Long-term Efficacy Study (APPLES) was a 6-month, multicenter, randomized study of neurocognitive function in patients with OSA (N = 1,098).4 Subjective sleepiness as measured by the Epworth Sleepiness Scale showed statistically significant improvement at 2 and 6 months for patients with moderate to severe OSA using CPAP. Objective sleepiness as measured by the Maintenance of Wakefulness Test showed statistically significant improvement (ie, improved daytime alertness) at 2 and 6 months for patients with severe OSA using CPAP.

Neurocognitive function

APPLES also tested for attention and psycho­motor function as well as verbal learning and memory, though no statistically significant improvements were found in these parameters.4 Executive function and frontal lobe function showed transient improvement at 2 months in patients with severe sleep apnea using CPAP, but the improvement was not statistically significant at 6 months.

Cardiovascular outcomes

Hypertension and cardiovascular disease. Use of CPAP therapy reduces blood pressure in individuals with hypertension. A study of 32 patients who had a baseline polysomnography with 19 hours of continuous mean arterial blood pressure monitoring were treated with therapeutic CPAP (n = 16) or subtherapeutic CPAP (n = 16).5 Therapeutic treatment with CPAP for patients with moderate to severe OSA resulted in statistically significant reductions in mean arterial pressure for both systolic and diastolic pressures. The blood pressure reductions achieved are estimated to reduce coronary artery diseases by 37% and stroke by 56%.5

The risk of cardiovascular events in men with severe sleep apnea is high but mitigated by the use of CPAP. In a cohort of 1,651 men, untreated severe sleep apnea resulted in a threefold increase in the rate of cardiovascular events per 1,000 patient-years compared with 4 other groups: a control group, men who snore, men with untreated mild to moderate sleep apnea, and men with OSA using CPAP.6 However, when men with severe sleep apnea use CPAP, the risk of cardiovascular events is reduced to the rate in men who snore.

Atrial fibrillation. In patients with atrial fibrillation treated with direct-current cardioversion-
defibrillation, the recurrence of atrial fibrillation at 12 months was greater in patients with untreated OSA (82%) compared with a control group (53%) and patients treated for OSA (42%).7

Heart failure. In a study of 24 patients with heart failure, an ejection fraction less than 45%, and OSA, patients were randomized to a control group for medical treatment or medical treatment and nasal CPAP for 1 month.8 In the CPAP group, mean systolic blood pressure and heart rate were reduced, resulting in an improved ejection fraction compared with baseline, as well as compared with patients in the control group.

In patients with heart failure (N = 66) with and without Cheyne-Stokes respirations in central sleep apnea, patients treated with CPAP were found to have a 60% relative risk reduction in mortality-cardiac transplant rate compared with the control group not using CPAP.9 Further stratification in this study showed that patients with significant Cheyne-Stokes respirations and central sleep apnea had an improved ejection fraction at 3 months and an 81% reduced mortality-cardiac transplant rate.9

 

 

ADHERENCE

Adherence to PAP therapy is a problem in terms of both frequency of use and duration of use per night. A review of randomized control trials of CPAP compliance between 2011 and 2015 found adherence varied widely from 35% to 87%.10 The average hours of PAP use per night was found to be 5 hours in APPLES.4 Patients adherent to PAP therapy at 1 month remained adherent at 1 year, suggesting patients using CPAP for 1 month were more likely to continue use at 1 year.10 Impediments to PAP use typically involve the facial interface discomfort, lack of humidity, and pressure intolerance.

FEATURES OF PAP DEVICES

Today’s PAP devices have features designed to make them easier to use and more comfortable to improve adherence to therapy. Facial interface options, heated humidifiers, tubing accessories, cleaning devices, reporting of compliance data via telecommunication, and pressure adjustment features of PAP devices may improve patient adherence and comfort, as highlighted in the case scenarios presented below.

Interfaces

Case scenario #1

A 32-year-old woman with moderate sleep apnea complains that her PAP nasal mask is making very loud noises and is disturbing her bed partner. She is a side sleeper and also reports that she wakes with an extremely dry mouth.

Management of the leak could include which of the following?

  1. Chin strap
  2. Avoidance of facial creams before bedtime
  3. CPAP pillow
  4. Clean the mask daily
  5. All of the above

Answer: All of the above.

lance_papimpactonapnea_f2.jpg
%3Cp%3EFigure%202.%20Download%20of%20positive%20airway%20pressure%20use%20data%20for%20a%20month%20(A)%20and%20leak%20data%20for%20a%20night%20(B).%3C%2Fp%3E
Figure 2 shows an overview of data from the patient’s machine for the past month and 1 night of leak data. Both the month-use data and single-night leak data show mask leakage.

There are many types of PAP interfaces such as nasal masks, nasal pillows, nasal cushions, full-face masks, and less frequently used oral and total face masks. The mask interface is a common impediment to use of PAP therapy often due to poor mask fit or leakage.

Nasal masks cover only the nose and require that the mouth remains closed, which can be achieved with the addition of a chin strap. Nasal masks are available in a variety of materials including cloth. Nasal pillows actually go into the nostrils whereas the nasal mask is positioned under the nose. A nasal cushion mask sits under the nose but does not go into the nostrils.

A study by Lanza and colleagues11 evaluated patient comfort with PAP therapy based on the type of nasal interface mask. Patients using nasal pillows had improvement with respect to swollen eyes, discomfort, skin breakdown, and marks on the face compared with patients using nasal masks; however, nasal pillows can cause nostril pain.

Several types of full-face masks are available, some that fit over the bridge of the nose and some that fit just under the nose. A variety of head straps are available to secure full-face masks. One benefit of full-face masks is that air pressure is delivered to both the nose and the mouth, so the mouth can be open or closed. However, the larger surface area of the full-face mask increases the potential for leaks. A study of adherence in 20 patients using CPAP with nasal masks or full-face masks evaluated hours per use, adherence at 12 months, and comfort.12 Patients using full-face masks had more hours per use, better adherence at 12 months, and more comfort than patients using nasal masks.

Interface skin irritation and leak management. To help combat skin irritation, particularly for patients with rosacea, cloth products are available for use beneath the mask and headgear. Silicone pads for masks that cause pressure on the bridge of the nose can help protect against skin breakdown. Sleeping positions other than the supine position can contribute to mask leak. CPAP pillows are designed to allow patients to sleep in their desired position while maintaining an adequate mask seal. The pillows are shaped or have cutouts that prevent the mask from pushing on the pillow and creating a leak.

Humidification

Case scenario #2

A 54-year-old man with severe sleep apnea recently initiated CPAP therapy. He quickly discontinued use due to nasal congestion.

Which of the following is NOT recommended?

  1. Assure adequate heated humidification
  2. Assure that the apnea is adequately treated
  3. Use of a full-face mask
  4. Use of short-acting nasal decongestants
  5. Use of a topical nasal steroid

Answer: Use of short-acting nasal decongestants.

Nasal congestion is a common reason for nonadherence to CPAP therapy.13 Pressurized air is very drying and can be very uncomfortable. Residual apneic events can even precipitate further congestion. The use of humidification with CPAP can improve patient comfort and compliance. The vast majority of patients use CPAP devices with heated humidifiers. Heated humidification has been found to increase CPAP use and improve daytime sleepiness and feelings of satisfaction and being refreshed compared with cold humidity or no humidity.14 Cold humidification improved daytime sleepiness and satisfaction, but not to the degree found with heated humidification.

Heated humidifiers are incorporated in the CPAP machine or attach to it. Heated in-line tubing helps with “rain out,” which refers to water condensation inside the tubing and mask associated with CPAP humidification.

Topical decongestants can actually worsen congestion and cause a reflex vasodilation. Topical nasal steroids can be used for nasal congestion and may be beneficial.

 

 

Tubing

The tubing from the PAP device to the facial interface can be a source of irritation to patients due to rubbing against the skin or entanglement. Products to cover the tubing to reduce irritation and avoid entanglement are available. Extra-long tubing is also available.

Cleaning

Some people find cleaning CPAP equipment daunting. Cleaning devices are available and recommended to patients looking for reassurance about how to keep their CPAP equipment clean. There are also CPAP wipes to clean the mask of oils and creams from the skin to improve the mask seal and reduce leaks.

Pressure control

Advanced modalities are available to adjust how pressure is delivered by PAP devices, including ramp, APAP, pressure relief, and BiPAP. Ramp is a feature that delivers a lower pressure at the beginning of the sleep cycle and slowly increases pressure to therapeutic levels. The lower pressure makes it easier for the user to fall asleep and builds to therapeutic pressure once asleep. APAP adjusts the pressure automatically when needed and reduces the pressure when not needed. Pressure relief is a feature that allows the PAP pressure to decrease at the point of expiration. BiPAP gives a distinct pressure on inspiration and a distinctively different and lower pressure at the point of expiration.

Auto-PAP

Case scenario #3

A 52-year-old woman with hypertension and mild sleep apnea has a polysomnogram with an apnea–hypopnea index of 7 events per hour that increase to 32 events per hour in rapid eye movement (REM) sleep. She is on CPAP at 5 cm of water, but complains of waking every 2 hours with a sense of panic and hot flashes.

Which of the following is the most likely cause of her symptoms?

  1. An underlying anxiety disorder
  2. An underlying heart condition
  3. Perimenopausal symptoms
  4. Undertreated REM-related apnea
  5. None of the above

Answer: While all of these choices can occur, the most likely cause is undertreated REM-related apnea.

lance_papimpactonapnea_f3.jpg
%3Cp%3EFigure%203.%20Sleep%20study%20overview%20showing%20rapid%20eye%20movement%20sleep%20(arrow%2Fblack%20bar)%20associated%20with%20increased%20arousals%20and%20apneic%20events%20and%20decreased%20oxygen%20levels.%3C%2Fp%3E
The sleep study overview for this patient is shown in Figure 3. During REM sleep, arousals and apneas are clustered and associated with a severe drop in oxygen levels. While doing well on CPAP at 5 cm of water, when the patient dreams, the apnea may become worse and more pressure may be needed.

What would be the best next step in treatment for this patient?

  1. Hormonal replacement therapy
  2. Positional therapy in addition to CPAP
  3. APAP
  4. Anxiolytic medication
  5. All of the above

Answer: APAP.

APAP incorporates an algorithm that detects and adjusts to airflow, pressure fluctuations, and airway resistance. The consensus from the American Academy of Sleep Medicine is that APAP is useful in the case of:

  • Pressure intolerance
  • REM apnea or positional apnea
  • Inadequate in lab PAP titration
  • Planned weight loss (bariatric surgery)
  • Recurrent symptoms after long-term CPAP use.15

Pressure relief

Case scenario #4

A 45-year-old man with severe sleep apnea uses CPAP at 10 cm of water. He complains of the inability to exhale against the pressure from the device.

What would be the best next step?

  1. Set the pressure relief to a maximum of 3
  2. Lower the pressure of CPAP and check a download use at a lower pressure
  3. BiPAP titration study in the laboratory
  4. Switch to BiPAP if insurance allows
  5. Change to a different mask

Answer: Set the pressure relief to a maximum of 3.

The CPAP device delivers pressure in conjunction with the patient’s inspiration and expiration. At the point of expiration, there is a decrease in the pressure delivered by the device to make it easier for the user to exhale. Three selectable settings provide flow-based pressure relief with a setting of 1 for the least degree of pressure reduction and a setting of 3 for the greatest degree of pressure reduction.16

In a study of the effect of PAP with pressure relief, 93 patients were assigned to use APAP without pressure relief, CPAP with pressure relief (C-Flex), or APAP with pressure relief (A-Flex).16 At 3 and 6 months, patients using A-Flex had the best adherence to therapy.

Quality of life was also examined in this same study.16 For patients using APAP alone, there was no statistically significant difference in the Epworth Sleepiness Scale measuring daytime sleepiness or the Pittsburgh Sleep Quality Index. However, in patients using A-Flex, daytime sleepiness improved, as did sleep quality, with statistically significant improvement at 3 months.

Bilevel PAP

Case scenario #5

A 62-year-old man with severe sleep apnea uses CPAP set at 17 cm of water and pressure relief set at 3. He stopped using CPAP due to abdominal pain, extreme belching, and pressure intolerance.

What would be the appropriate next step?

  1. Use of simethicone
  2. Elevate the head while using PAP therapy
  3. BiPAP titration study in the laboratory
  4. Switching directly to BiPAP if insurance allows
  5. All of the above

Answer: All of the above.

BiPAP devices provide 2 distinct pressures, one for inhalation and one for exhalation. BiPAP also has the ability to deliver a higher overall pressure. A CPAP device typically has a maximum pressure of 20 cm of water, but BiPAP has a maximum pressure of 25 cm of water on inspiration. BiPAP may be helpful in patients with air aphasia and extreme belching. If a patient cannot tolerate CPAP because of the pressure, and if C-Flex has not alleviated the problem, BiPAP would be the next step.

The effectiveness and level of comfort of BiPAP compared with CPAP for the treatment of OSA was evaluated by the American Academy of Sleep Medicine.2 The analysis of 7 randomized control trials reporting level I and II evidence found that BiPAP was as effective as CPAP in the treatment of OSA in patients with no comorbidities. For patients with OSA and comorbidities, a level III evidence study reported an increased level of comfort in patients using BiPAP.

 

 

PATIENT-CENTERED STRATEGIES TO IMPROVE ADHERENCE

Innovative strategies and approaches focusing on patient factors affecting PAP adherence include motivational interviewing, motivational enhancement, telemedicine, and desensitization techniques.

Motivational interviews and enhancement

Motivational interviewing and motivational enhancement were first used to help with alcohol abuse.17 Motivational interviewing is goal-oriented, patient-centered counseling to elicit a particular behavioral change. The goal is to explore and resolve ambivalence, increase engagement, and evoke a positive response and perspective that builds momentum and results in action.

Motivational enhancement and motivational engagement in the use of PAP therapy were evaluated in the Patient Engagement Study.18 Patients were assigned to usual care (n = 85,358) or active patient engagement (APE) (n = 42,679). Usual care involved diagnosis of apnea, initiation of CPAP, and follow-up, whereas APE included daily feedback (ie, daily scores of apnea–hypopnea index, mask leaks, hours used), positive praise messages, and personal coaching assistance. Overall adherence for patients assigned to APE was 87% compared with 70% in the usual-care group. The hours of use per night also increased for patients in the APE group.

The Best Apnea Interventions for Research trial randomized patients with or at risk of cardiovascular disease (N = 169) to CPAP alone or CPAP with motivational enhancements for 6 months.19 Motivational enhancements and interventions included brief in-person and phone interventions. An overall average difference of 99 minutes per night improvement in CPAP use was reported in the motivational enhancement group compared with CPAP alone.

The Motivational Interviewing Nurse Therapy study trained nurses in motivational interviewing and randomized 106 patients with newly diagnosed OSA to CPAP alone or CPAP plus motivational interviews.20 Motivational interviews involved 3 sessions: 1 to build motivation prior to the CPAP titration, 1 to strengthen the commitment to achieve the prescribed time, and a booster session 1 month after CPAP setup. Adherence was found to improve at 1, 2, and 3 months in the motivational interview group; however, no difference between the 2 groups in adherence was noted at 12 months.

Telemedicine

The role of telemedicine in improving adherence with CPAP therapy was evaluated in 75 patients with moderate to severe apnea randomized to APAP alone or with phone call support from a research coordinator.21 Phone calls occurred 2 days after device setup, and daily monitoring of several factors was done via modem. Patients were contacted if the mask was leaking more than 30% of the night, use was less than 4 hours per night on 2 consecutive nights, the apnea–hypopnea index was greater than 10, or the average pressure needed was higher than 16 cm of water. Statistically significant improvement was found in the telemedicine group in mean adherence, minutes used per day, and mean amount of time spent with the patients.

Desensitization to PAP therapy

Case scenario #6

A 33-year-old woman with a history of anxiety and depression and a remote history of abuse as a child was diagnosed with severe apnea. When she tries to use her CPAP, she has a sense of panic and cannot proceed.

Which of the following has NOT been shown to be beneficial in this situation?

  1. Psychologist for behavioral therapy
  2. Desensitization protocol
  3. PAP “NAP”
  4. Short-acting hypnotics
  5. None of the above

Answer: Short-acting hypnotics.

The short-acting hypnotic zaleplon (Sonata) was evaluated in a 1-month study of 88 patients compared with placebo control, and no difference was found between the 2 groups in measures of adherence to therapy or symptoms.22

lance_papimpactonapnea_t1.jpg
A protocol for desensitization to CPAP use is helpful to assist patients in acclimating to therapy. An example of the steps in a desensitization protocol that patients can do at home is provided in Table 1.

PAP NAP. For some people, at-home desensitization is not enough, and a sleep lab session may be needed. A PAP NAP is a daytime study conducted in the sleep lab. Patients do not necessarily sleep, but work with a technologist with a minimal hookup to polysomnography equipment on mask desensitization, as well as biofeedback techniques. PAP NAPs are indicated for patients with claustrophobia, anxiety surrounding PAP therapy, or pressure intolerance.

A study of 99 patients with moderate to severe apnea and insomnia and concomitant psychiatric disorders resistant to CPAP evaluated adherence in a group receiving a PAP NAP (n = 39) compared with a control group (n = 60).23 The PAP NAP group had marked improvement in completion of CPAP titration in the lab, filling the CPAP prescription, and using CPAP more than 5 days a week and more than 4 hours a night.

A new innovative concept called the Sleep Apnea Patient-Centered Outcomes Network is a collaborative group that includes patients, researchers, and clinicians.24 The group addresses issues such as cost, outcomes, and value in the diagnosis and treatment of sleep apnea. A patient-centered website provides forums, education, and data collection capability for researchers (myapnea.org).

SUMMARY

PAP therapy is the gold standard for treatment of patients with moderate to severe OSA, though poor adherence to PAP therapy is a persistent problem. Advanced features in PAP devices such as APAP and other innovative strategies like motivational enhancement and desensitization protocols and PAP NAP are being used to address poor adherence. More randomized controlled trials are needed to evaluate PAP for sleep apnea.

References
  1. Sullivan CE, Issa FG, Berthon-Jones M, Eves L. Reversal of obstructive sleep apnoea by continuous positive airway pressure applied through the nares. Lancet 1981; 1(8225):862–865.
  2. Gay P, Weaver T, Loube D, Iber C. Evaluation of positive airway pressure treatment for sleep related breathing disorders in adults: a review by the Positive Airway Pressure Task Force of the Standards of Practice Committee of the American Academy of Sleep Medicine. Sleep 2006; 29(3):381–401.
  3. Weaver TE, Maislin G, Dinges DF, et al. Relationship between hours of CPAP use and achieving normal levels of sleepiness and daily functioning. Sleep 2007; 30(6):711–719.
  4. Kushida CA, Nichols DA, Holmes TH, et al. Effects of continuous positive airway pressure on neurocognitive function in obstructive sleep apnea patients: the Apnea Positive Pressure Long-term Efficacy Study (APPLES). Sleep 2012; 35(12):1593–1602.
  5. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  6. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardio­vascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  7. Kanagala R, Murali NS, Friedman PA, et al. Obstructive sleep apnea and the recurrence of atrial fibrillation. Circulation 2003; 107(20):2589–2594.
  8. Kaneko Y, Floras JS, Usui K, et al. Cardiovascular effects of continuous positive airway pressure in patients with heart failure and obstructive sleep apnea. N Engl J Med 2003; 348(13):1233–1241.
  9. Sin DD, Logan AG, Fitzgerald FS, Liu PP, Bradley TD. Effects of continuous positive airway pressure on cardiovascular outcomes in heart failure patients with and without Cheyne-Stokes respiration. Circulation 2000; 102(1):61–66.
  10. Tan B, Tan A, Huak CY, Yingjuan M, Siang WH, Poh HP. Adherence to continuous positive airway pressure therapy in Singaporean patients with obstructive sleep apnea. Am J Otolaryngol 2018; 39(5):501–506.
  11. Lanza A, Mariani S, Sommariva M, et al. Continuous positive airway pressure treatment with nasal pillows in obstructive sleep apnea: long-term effectiveness and adherence. Sleep Med 2018; 41:94–99.
  12. Mortimore IL, Whittle AT, Douglas NJ. Comparison of nose and face mask CPAP therapy for sleep apnoea. Thorax 1998; 53(4):290–292.
  13. Morgenthaler TI, Kapen S, Lee-Chiong T, et al. Practice parameters for the medical therapy of obstructive sleep apnea. Sleep 2006; 29(8):1031–1035.
  14. Massie CA, Hart RW, Peralez K, Richards GN. Effects of humidification on nasal symptoms and compliance in sleep apnea patients using continuous positive airway pressure. Chest 1999; 116(2):403–408.
  15. Morgenthaler TI, Aurora RN, Brown T, et al; Standards of Practice Committee of the AASM. Practice parameters for the use of auto­titrating continuous positive airway pressure devices for titrating pressures and treating adult patients with obstructive sleep apnea syndrome: an update for 2007. Sleep 2008; 31(1):141–147.
  16. Chihara Y, Tsuboi T, Hitomi T, et al. Flexible positive airway pressure improves treatment adherence compared with auto-adjusting PAP. Sleep 2013; 36(2):229–236.
  17. Miller WR, Rollnick S. Motivational interviewing: Preparing people to change addictive behavior. New York: Guilford Press; 1991.
  18. Malhotra A, Crocker ME, Willes L, Kelly C, Lynch S, Benjafield AV. Patient engagement using new technology to improve adherence to positive airway pressure therapy: a retrospective analysis. Chest 2018; 153(4):843–850.
  19. Bakker JP, Wang R, Weng J, et al. Motivational enhancement for increasing adherence to CPAP: a randomized controlled trial. Chest 2016; 150(2):337–345.
  20. Olsen S, Smith SS, Oei TP, Douglas J. Motivational interviewing (MINT) improves continuous positive airway pressure (CPAP) acceptance and adherence: a randomized controlled trial. J Consult Clin Psychol 2012; 80(1):151–163.
  21. Fox N, Hirsch-Allen AJ, Goodfellow E, et al. The impact of a telemedicine monitoring system on positive airway pressure adherence in patients with obstructive sleep apnea: a randomized controlled trial. Sleep 2012; 35(4):477–481.
  22. Park JG, Olson EJ, Morgenthaler TI. Impact of zaleplon on continuous positive airway pressure therapy compliance. J Clin Sleep Med 2013; 9(5):439–444.
  23. Krakow B, Ulibarri V, Melendrez D, Kikta S, Togami L, Haynes P. A daytime, abbreviated cardio-respiratory sleep study (CPT 95807-52) to acclimate insomnia patients with sleep disordered breathing to positive airway pressure (PAP-NAP). J Clin Sleep Med 2008; 4(3):212–222.
  24. Redline S, Baker-Goodwin S, Bakker JP, et al; for the Sleep Apnea Patient-Centered Outcomes Network. Patient partnerships transforming sleep medicine research and clinical care: perspectives from the sleep apnea patient-centered outcomes network. J Clin Sleep Med 2016; 12(7):1053–1058.
Article PDF
Author and Disclosure Information

Colleen G. Lance, MD
Medical Director, Sleep Laboratory, Hillcrest Hospital, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Colleen G. Lance, MD, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; lancec@ccf.org

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

This article is based on Dr. Lance’s “Positive Airway Pressure: Making an Impact on Sleep Apnea” webcast released April 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Lance.

Publications
Topics
Page Number
26-33
Legacy Keywords
Positive airway pressure, PAP, continuous positive airway pressure, CPAP, auto-positive airway pressure, APAP, bilevel positive airway pressure, BiPAP, colleen lance, nancy foldvary-schaefer
Sections
Author and Disclosure Information

Colleen G. Lance, MD
Medical Director, Sleep Laboratory, Hillcrest Hospital, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Colleen G. Lance, MD, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; lancec@ccf.org

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

This article is based on Dr. Lance’s “Positive Airway Pressure: Making an Impact on Sleep Apnea” webcast released April 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Lance.

Author and Disclosure Information

Colleen G. Lance, MD
Medical Director, Sleep Laboratory, Hillcrest Hospital, Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Colleen G. Lance, MD, Sleep Disorders Center, Neurological Institute, S73, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; lancec@ccf.org

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

This article is based on Dr. Lance’s “Positive Airway Pressure: Making an Impact on Sleep Apnea” webcast released April 30, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Lance.

Article PDF
Article PDF
Related Articles

lance_papimpactonapnea_f1.jpg
%3Cp%3EFigure%201.%20Obstructed%20airway%20(left)%20is%20opened%20with%20a%20column%20of%20air%20delivered%20using%20positive%20airway%20pressure%20therapy%20(right).%3C%2Fp%3E
Positive airway pressure (PAP) therapy is used to open an obstructed upper airway (Figure 1). PAP therapy consists of a small bedside unit that creates a pressurized column of air that is delivered through tubing to a facial interface, which can be nasal, oral, or both. Collin Sullivan, MD, created the nasal continuous PAP (CPAP) in 1982 using parts of a vacuum cleaner to create positive pressure that successfully resolved hypoxemia in a patient.1 Today, the various forms of PAP therapy include CPAP, the most common, auto-PAP (APAP), and bilevel PAP (BiPAP).

EFFICACY OF PAP THERAPY

The American Academy of Sleep Medicine practice guidelines for PAP are based on 342 articles, most rated as evidence levels I and II, concluding that CPAP is superior to conservative treatment to:

  • Eliminate respiratory disturbances
  • Reduce the apnea–hypopnea index
  • Decrease the arousal index on electroencephalogram
  • Increase in the total amount of slow-wave or N3 sleep
  • Reduce daytime sleepiness.2

These practice parameters are based on evidence of improved daytime sleepiness and reduced incidence of cardiovascular events in patients with moderate to severe obstructive sleep apnea (OSA) treated with PAP. The evidence is less clear for neurocognitive markers and cardiovascular events in the treatment of patients with mild sleep apnea.

Sleepiness

A study evaluated sleepiness outcomes in 149 patients with severe sleep apnea with an average apnea–hypopnea index of 69 relative to the duration of nightly CPAP use. Sleepiness was measured using the Functional Outcomes of Sleep Questionnaire, Epworth Sleepiness Scale, and Multiple Sleep Latency Test. Results suggest that a greater percentage of patients had improved daytime sleepiness as the total hours of sleep using CPAP increased.3

The Apnea Positive Pressure Long-term Efficacy Study (APPLES) was a 6-month, multicenter, randomized study of neurocognitive function in patients with OSA (N = 1,098).4 Subjective sleepiness as measured by the Epworth Sleepiness Scale showed statistically significant improvement at 2 and 6 months for patients with moderate to severe OSA using CPAP. Objective sleepiness as measured by the Maintenance of Wakefulness Test showed statistically significant improvement (ie, improved daytime alertness) at 2 and 6 months for patients with severe OSA using CPAP.

Neurocognitive function

APPLES also tested for attention and psycho­motor function as well as verbal learning and memory, though no statistically significant improvements were found in these parameters.4 Executive function and frontal lobe function showed transient improvement at 2 months in patients with severe sleep apnea using CPAP, but the improvement was not statistically significant at 6 months.

Cardiovascular outcomes

Hypertension and cardiovascular disease. Use of CPAP therapy reduces blood pressure in individuals with hypertension. A study of 32 patients who had a baseline polysomnography with 19 hours of continuous mean arterial blood pressure monitoring were treated with therapeutic CPAP (n = 16) or subtherapeutic CPAP (n = 16).5 Therapeutic treatment with CPAP for patients with moderate to severe OSA resulted in statistically significant reductions in mean arterial pressure for both systolic and diastolic pressures. The blood pressure reductions achieved are estimated to reduce coronary artery diseases by 37% and stroke by 56%.5

The risk of cardiovascular events in men with severe sleep apnea is high but mitigated by the use of CPAP. In a cohort of 1,651 men, untreated severe sleep apnea resulted in a threefold increase in the rate of cardiovascular events per 1,000 patient-years compared with 4 other groups: a control group, men who snore, men with untreated mild to moderate sleep apnea, and men with OSA using CPAP.6 However, when men with severe sleep apnea use CPAP, the risk of cardiovascular events is reduced to the rate in men who snore.

Atrial fibrillation. In patients with atrial fibrillation treated with direct-current cardioversion-
defibrillation, the recurrence of atrial fibrillation at 12 months was greater in patients with untreated OSA (82%) compared with a control group (53%) and patients treated for OSA (42%).7

Heart failure. In a study of 24 patients with heart failure, an ejection fraction less than 45%, and OSA, patients were randomized to a control group for medical treatment or medical treatment and nasal CPAP for 1 month.8 In the CPAP group, mean systolic blood pressure and heart rate were reduced, resulting in an improved ejection fraction compared with baseline, as well as compared with patients in the control group.

In patients with heart failure (N = 66) with and without Cheyne-Stokes respirations in central sleep apnea, patients treated with CPAP were found to have a 60% relative risk reduction in mortality-cardiac transplant rate compared with the control group not using CPAP.9 Further stratification in this study showed that patients with significant Cheyne-Stokes respirations and central sleep apnea had an improved ejection fraction at 3 months and an 81% reduced mortality-cardiac transplant rate.9

 

 

ADHERENCE

Adherence to PAP therapy is a problem in terms of both frequency of use and duration of use per night. A review of randomized control trials of CPAP compliance between 2011 and 2015 found adherence varied widely from 35% to 87%.10 The average hours of PAP use per night was found to be 5 hours in APPLES.4 Patients adherent to PAP therapy at 1 month remained adherent at 1 year, suggesting patients using CPAP for 1 month were more likely to continue use at 1 year.10 Impediments to PAP use typically involve the facial interface discomfort, lack of humidity, and pressure intolerance.

FEATURES OF PAP DEVICES

Today’s PAP devices have features designed to make them easier to use and more comfortable to improve adherence to therapy. Facial interface options, heated humidifiers, tubing accessories, cleaning devices, reporting of compliance data via telecommunication, and pressure adjustment features of PAP devices may improve patient adherence and comfort, as highlighted in the case scenarios presented below.

Interfaces

Case scenario #1

A 32-year-old woman with moderate sleep apnea complains that her PAP nasal mask is making very loud noises and is disturbing her bed partner. She is a side sleeper and also reports that she wakes with an extremely dry mouth.

Management of the leak could include which of the following?

  1. Chin strap
  2. Avoidance of facial creams before bedtime
  3. CPAP pillow
  4. Clean the mask daily
  5. All of the above

Answer: All of the above.

lance_papimpactonapnea_f2.jpg
%3Cp%3EFigure%202.%20Download%20of%20positive%20airway%20pressure%20use%20data%20for%20a%20month%20(A)%20and%20leak%20data%20for%20a%20night%20(B).%3C%2Fp%3E
Figure 2 shows an overview of data from the patient’s machine for the past month and 1 night of leak data. Both the month-use data and single-night leak data show mask leakage.

There are many types of PAP interfaces such as nasal masks, nasal pillows, nasal cushions, full-face masks, and less frequently used oral and total face masks. The mask interface is a common impediment to use of PAP therapy often due to poor mask fit or leakage.

Nasal masks cover only the nose and require that the mouth remains closed, which can be achieved with the addition of a chin strap. Nasal masks are available in a variety of materials including cloth. Nasal pillows actually go into the nostrils whereas the nasal mask is positioned under the nose. A nasal cushion mask sits under the nose but does not go into the nostrils.

A study by Lanza and colleagues11 evaluated patient comfort with PAP therapy based on the type of nasal interface mask. Patients using nasal pillows had improvement with respect to swollen eyes, discomfort, skin breakdown, and marks on the face compared with patients using nasal masks; however, nasal pillows can cause nostril pain.

Several types of full-face masks are available, some that fit over the bridge of the nose and some that fit just under the nose. A variety of head straps are available to secure full-face masks. One benefit of full-face masks is that air pressure is delivered to both the nose and the mouth, so the mouth can be open or closed. However, the larger surface area of the full-face mask increases the potential for leaks. A study of adherence in 20 patients using CPAP with nasal masks or full-face masks evaluated hours per use, adherence at 12 months, and comfort.12 Patients using full-face masks had more hours per use, better adherence at 12 months, and more comfort than patients using nasal masks.

Interface skin irritation and leak management. To help combat skin irritation, particularly for patients with rosacea, cloth products are available for use beneath the mask and headgear. Silicone pads for masks that cause pressure on the bridge of the nose can help protect against skin breakdown. Sleeping positions other than the supine position can contribute to mask leak. CPAP pillows are designed to allow patients to sleep in their desired position while maintaining an adequate mask seal. The pillows are shaped or have cutouts that prevent the mask from pushing on the pillow and creating a leak.

Humidification

Case scenario #2

A 54-year-old man with severe sleep apnea recently initiated CPAP therapy. He quickly discontinued use due to nasal congestion.

Which of the following is NOT recommended?

  1. Assure adequate heated humidification
  2. Assure that the apnea is adequately treated
  3. Use of a full-face mask
  4. Use of short-acting nasal decongestants
  5. Use of a topical nasal steroid

Answer: Use of short-acting nasal decongestants.

Nasal congestion is a common reason for nonadherence to CPAP therapy.13 Pressurized air is very drying and can be very uncomfortable. Residual apneic events can even precipitate further congestion. The use of humidification with CPAP can improve patient comfort and compliance. The vast majority of patients use CPAP devices with heated humidifiers. Heated humidification has been found to increase CPAP use and improve daytime sleepiness and feelings of satisfaction and being refreshed compared with cold humidity or no humidity.14 Cold humidification improved daytime sleepiness and satisfaction, but not to the degree found with heated humidification.

Heated humidifiers are incorporated in the CPAP machine or attach to it. Heated in-line tubing helps with “rain out,” which refers to water condensation inside the tubing and mask associated with CPAP humidification.

Topical decongestants can actually worsen congestion and cause a reflex vasodilation. Topical nasal steroids can be used for nasal congestion and may be beneficial.

 

 

Tubing

The tubing from the PAP device to the facial interface can be a source of irritation to patients due to rubbing against the skin or entanglement. Products to cover the tubing to reduce irritation and avoid entanglement are available. Extra-long tubing is also available.

Cleaning

Some people find cleaning CPAP equipment daunting. Cleaning devices are available and recommended to patients looking for reassurance about how to keep their CPAP equipment clean. There are also CPAP wipes to clean the mask of oils and creams from the skin to improve the mask seal and reduce leaks.

Pressure control

Advanced modalities are available to adjust how pressure is delivered by PAP devices, including ramp, APAP, pressure relief, and BiPAP. Ramp is a feature that delivers a lower pressure at the beginning of the sleep cycle and slowly increases pressure to therapeutic levels. The lower pressure makes it easier for the user to fall asleep and builds to therapeutic pressure once asleep. APAP adjusts the pressure automatically when needed and reduces the pressure when not needed. Pressure relief is a feature that allows the PAP pressure to decrease at the point of expiration. BiPAP gives a distinct pressure on inspiration and a distinctively different and lower pressure at the point of expiration.

Auto-PAP

Case scenario #3

A 52-year-old woman with hypertension and mild sleep apnea has a polysomnogram with an apnea–hypopnea index of 7 events per hour that increase to 32 events per hour in rapid eye movement (REM) sleep. She is on CPAP at 5 cm of water, but complains of waking every 2 hours with a sense of panic and hot flashes.

Which of the following is the most likely cause of her symptoms?

  1. An underlying anxiety disorder
  2. An underlying heart condition
  3. Perimenopausal symptoms
  4. Undertreated REM-related apnea
  5. None of the above

Answer: While all of these choices can occur, the most likely cause is undertreated REM-related apnea.

lance_papimpactonapnea_f3.jpg
%3Cp%3EFigure%203.%20Sleep%20study%20overview%20showing%20rapid%20eye%20movement%20sleep%20(arrow%2Fblack%20bar)%20associated%20with%20increased%20arousals%20and%20apneic%20events%20and%20decreased%20oxygen%20levels.%3C%2Fp%3E
The sleep study overview for this patient is shown in Figure 3. During REM sleep, arousals and apneas are clustered and associated with a severe drop in oxygen levels. While doing well on CPAP at 5 cm of water, when the patient dreams, the apnea may become worse and more pressure may be needed.

What would be the best next step in treatment for this patient?

  1. Hormonal replacement therapy
  2. Positional therapy in addition to CPAP
  3. APAP
  4. Anxiolytic medication
  5. All of the above

Answer: APAP.

APAP incorporates an algorithm that detects and adjusts to airflow, pressure fluctuations, and airway resistance. The consensus from the American Academy of Sleep Medicine is that APAP is useful in the case of:

  • Pressure intolerance
  • REM apnea or positional apnea
  • Inadequate in lab PAP titration
  • Planned weight loss (bariatric surgery)
  • Recurrent symptoms after long-term CPAP use.15

Pressure relief

Case scenario #4

A 45-year-old man with severe sleep apnea uses CPAP at 10 cm of water. He complains of the inability to exhale against the pressure from the device.

What would be the best next step?

  1. Set the pressure relief to a maximum of 3
  2. Lower the pressure of CPAP and check a download use at a lower pressure
  3. BiPAP titration study in the laboratory
  4. Switch to BiPAP if insurance allows
  5. Change to a different mask

Answer: Set the pressure relief to a maximum of 3.

The CPAP device delivers pressure in conjunction with the patient’s inspiration and expiration. At the point of expiration, there is a decrease in the pressure delivered by the device to make it easier for the user to exhale. Three selectable settings provide flow-based pressure relief with a setting of 1 for the least degree of pressure reduction and a setting of 3 for the greatest degree of pressure reduction.16

In a study of the effect of PAP with pressure relief, 93 patients were assigned to use APAP without pressure relief, CPAP with pressure relief (C-Flex), or APAP with pressure relief (A-Flex).16 At 3 and 6 months, patients using A-Flex had the best adherence to therapy.

Quality of life was also examined in this same study.16 For patients using APAP alone, there was no statistically significant difference in the Epworth Sleepiness Scale measuring daytime sleepiness or the Pittsburgh Sleep Quality Index. However, in patients using A-Flex, daytime sleepiness improved, as did sleep quality, with statistically significant improvement at 3 months.

Bilevel PAP

Case scenario #5

A 62-year-old man with severe sleep apnea uses CPAP set at 17 cm of water and pressure relief set at 3. He stopped using CPAP due to abdominal pain, extreme belching, and pressure intolerance.

What would be the appropriate next step?

  1. Use of simethicone
  2. Elevate the head while using PAP therapy
  3. BiPAP titration study in the laboratory
  4. Switching directly to BiPAP if insurance allows
  5. All of the above

Answer: All of the above.

BiPAP devices provide 2 distinct pressures, one for inhalation and one for exhalation. BiPAP also has the ability to deliver a higher overall pressure. A CPAP device typically has a maximum pressure of 20 cm of water, but BiPAP has a maximum pressure of 25 cm of water on inspiration. BiPAP may be helpful in patients with air aphasia and extreme belching. If a patient cannot tolerate CPAP because of the pressure, and if C-Flex has not alleviated the problem, BiPAP would be the next step.

The effectiveness and level of comfort of BiPAP compared with CPAP for the treatment of OSA was evaluated by the American Academy of Sleep Medicine.2 The analysis of 7 randomized control trials reporting level I and II evidence found that BiPAP was as effective as CPAP in the treatment of OSA in patients with no comorbidities. For patients with OSA and comorbidities, a level III evidence study reported an increased level of comfort in patients using BiPAP.

 

 

PATIENT-CENTERED STRATEGIES TO IMPROVE ADHERENCE

Innovative strategies and approaches focusing on patient factors affecting PAP adherence include motivational interviewing, motivational enhancement, telemedicine, and desensitization techniques.

Motivational interviews and enhancement

Motivational interviewing and motivational enhancement were first used to help with alcohol abuse.17 Motivational interviewing is goal-oriented, patient-centered counseling to elicit a particular behavioral change. The goal is to explore and resolve ambivalence, increase engagement, and evoke a positive response and perspective that builds momentum and results in action.

Motivational enhancement and motivational engagement in the use of PAP therapy were evaluated in the Patient Engagement Study.18 Patients were assigned to usual care (n = 85,358) or active patient engagement (APE) (n = 42,679). Usual care involved diagnosis of apnea, initiation of CPAP, and follow-up, whereas APE included daily feedback (ie, daily scores of apnea–hypopnea index, mask leaks, hours used), positive praise messages, and personal coaching assistance. Overall adherence for patients assigned to APE was 87% compared with 70% in the usual-care group. The hours of use per night also increased for patients in the APE group.

The Best Apnea Interventions for Research trial randomized patients with or at risk of cardiovascular disease (N = 169) to CPAP alone or CPAP with motivational enhancements for 6 months.19 Motivational enhancements and interventions included brief in-person and phone interventions. An overall average difference of 99 minutes per night improvement in CPAP use was reported in the motivational enhancement group compared with CPAP alone.

The Motivational Interviewing Nurse Therapy study trained nurses in motivational interviewing and randomized 106 patients with newly diagnosed OSA to CPAP alone or CPAP plus motivational interviews.20 Motivational interviews involved 3 sessions: 1 to build motivation prior to the CPAP titration, 1 to strengthen the commitment to achieve the prescribed time, and a booster session 1 month after CPAP setup. Adherence was found to improve at 1, 2, and 3 months in the motivational interview group; however, no difference between the 2 groups in adherence was noted at 12 months.

Telemedicine

The role of telemedicine in improving adherence with CPAP therapy was evaluated in 75 patients with moderate to severe apnea randomized to APAP alone or with phone call support from a research coordinator.21 Phone calls occurred 2 days after device setup, and daily monitoring of several factors was done via modem. Patients were contacted if the mask was leaking more than 30% of the night, use was less than 4 hours per night on 2 consecutive nights, the apnea–hypopnea index was greater than 10, or the average pressure needed was higher than 16 cm of water. Statistically significant improvement was found in the telemedicine group in mean adherence, minutes used per day, and mean amount of time spent with the patients.

Desensitization to PAP therapy

Case scenario #6

A 33-year-old woman with a history of anxiety and depression and a remote history of abuse as a child was diagnosed with severe apnea. When she tries to use her CPAP, she has a sense of panic and cannot proceed.

Which of the following has NOT been shown to be beneficial in this situation?

  1. Psychologist for behavioral therapy
  2. Desensitization protocol
  3. PAP “NAP”
  4. Short-acting hypnotics
  5. None of the above

Answer: Short-acting hypnotics.

The short-acting hypnotic zaleplon (Sonata) was evaluated in a 1-month study of 88 patients compared with placebo control, and no difference was found between the 2 groups in measures of adherence to therapy or symptoms.22

lance_papimpactonapnea_t1.jpg
A protocol for desensitization to CPAP use is helpful to assist patients in acclimating to therapy. An example of the steps in a desensitization protocol that patients can do at home is provided in Table 1.

PAP NAP. For some people, at-home desensitization is not enough, and a sleep lab session may be needed. A PAP NAP is a daytime study conducted in the sleep lab. Patients do not necessarily sleep, but work with a technologist with a minimal hookup to polysomnography equipment on mask desensitization, as well as biofeedback techniques. PAP NAPs are indicated for patients with claustrophobia, anxiety surrounding PAP therapy, or pressure intolerance.

A study of 99 patients with moderate to severe apnea and insomnia and concomitant psychiatric disorders resistant to CPAP evaluated adherence in a group receiving a PAP NAP (n = 39) compared with a control group (n = 60).23 The PAP NAP group had marked improvement in completion of CPAP titration in the lab, filling the CPAP prescription, and using CPAP more than 5 days a week and more than 4 hours a night.

A new innovative concept called the Sleep Apnea Patient-Centered Outcomes Network is a collaborative group that includes patients, researchers, and clinicians.24 The group addresses issues such as cost, outcomes, and value in the diagnosis and treatment of sleep apnea. A patient-centered website provides forums, education, and data collection capability for researchers (myapnea.org).

SUMMARY

PAP therapy is the gold standard for treatment of patients with moderate to severe OSA, though poor adherence to PAP therapy is a persistent problem. Advanced features in PAP devices such as APAP and other innovative strategies like motivational enhancement and desensitization protocols and PAP NAP are being used to address poor adherence. More randomized controlled trials are needed to evaluate PAP for sleep apnea.

lance_papimpactonapnea_f1.jpg
%3Cp%3EFigure%201.%20Obstructed%20airway%20(left)%20is%20opened%20with%20a%20column%20of%20air%20delivered%20using%20positive%20airway%20pressure%20therapy%20(right).%3C%2Fp%3E
Positive airway pressure (PAP) therapy is used to open an obstructed upper airway (Figure 1). PAP therapy consists of a small bedside unit that creates a pressurized column of air that is delivered through tubing to a facial interface, which can be nasal, oral, or both. Collin Sullivan, MD, created the nasal continuous PAP (CPAP) in 1982 using parts of a vacuum cleaner to create positive pressure that successfully resolved hypoxemia in a patient.1 Today, the various forms of PAP therapy include CPAP, the most common, auto-PAP (APAP), and bilevel PAP (BiPAP).

EFFICACY OF PAP THERAPY

The American Academy of Sleep Medicine practice guidelines for PAP are based on 342 articles, most rated as evidence levels I and II, concluding that CPAP is superior to conservative treatment to:

  • Eliminate respiratory disturbances
  • Reduce the apnea–hypopnea index
  • Decrease the arousal index on electroencephalogram
  • Increase in the total amount of slow-wave or N3 sleep
  • Reduce daytime sleepiness.2

These practice parameters are based on evidence of improved daytime sleepiness and reduced incidence of cardiovascular events in patients with moderate to severe obstructive sleep apnea (OSA) treated with PAP. The evidence is less clear for neurocognitive markers and cardiovascular events in the treatment of patients with mild sleep apnea.

Sleepiness

A study evaluated sleepiness outcomes in 149 patients with severe sleep apnea with an average apnea–hypopnea index of 69 relative to the duration of nightly CPAP use. Sleepiness was measured using the Functional Outcomes of Sleep Questionnaire, Epworth Sleepiness Scale, and Multiple Sleep Latency Test. Results suggest that a greater percentage of patients had improved daytime sleepiness as the total hours of sleep using CPAP increased.3

The Apnea Positive Pressure Long-term Efficacy Study (APPLES) was a 6-month, multicenter, randomized study of neurocognitive function in patients with OSA (N = 1,098).4 Subjective sleepiness as measured by the Epworth Sleepiness Scale showed statistically significant improvement at 2 and 6 months for patients with moderate to severe OSA using CPAP. Objective sleepiness as measured by the Maintenance of Wakefulness Test showed statistically significant improvement (ie, improved daytime alertness) at 2 and 6 months for patients with severe OSA using CPAP.

Neurocognitive function

APPLES also tested for attention and psycho­motor function as well as verbal learning and memory, though no statistically significant improvements were found in these parameters.4 Executive function and frontal lobe function showed transient improvement at 2 months in patients with severe sleep apnea using CPAP, but the improvement was not statistically significant at 6 months.

Cardiovascular outcomes

Hypertension and cardiovascular disease. Use of CPAP therapy reduces blood pressure in individuals with hypertension. A study of 32 patients who had a baseline polysomnography with 19 hours of continuous mean arterial blood pressure monitoring were treated with therapeutic CPAP (n = 16) or subtherapeutic CPAP (n = 16).5 Therapeutic treatment with CPAP for patients with moderate to severe OSA resulted in statistically significant reductions in mean arterial pressure for both systolic and diastolic pressures. The blood pressure reductions achieved are estimated to reduce coronary artery diseases by 37% and stroke by 56%.5

The risk of cardiovascular events in men with severe sleep apnea is high but mitigated by the use of CPAP. In a cohort of 1,651 men, untreated severe sleep apnea resulted in a threefold increase in the rate of cardiovascular events per 1,000 patient-years compared with 4 other groups: a control group, men who snore, men with untreated mild to moderate sleep apnea, and men with OSA using CPAP.6 However, when men with severe sleep apnea use CPAP, the risk of cardiovascular events is reduced to the rate in men who snore.

Atrial fibrillation. In patients with atrial fibrillation treated with direct-current cardioversion-
defibrillation, the recurrence of atrial fibrillation at 12 months was greater in patients with untreated OSA (82%) compared with a control group (53%) and patients treated for OSA (42%).7

Heart failure. In a study of 24 patients with heart failure, an ejection fraction less than 45%, and OSA, patients were randomized to a control group for medical treatment or medical treatment and nasal CPAP for 1 month.8 In the CPAP group, mean systolic blood pressure and heart rate were reduced, resulting in an improved ejection fraction compared with baseline, as well as compared with patients in the control group.

In patients with heart failure (N = 66) with and without Cheyne-Stokes respirations in central sleep apnea, patients treated with CPAP were found to have a 60% relative risk reduction in mortality-cardiac transplant rate compared with the control group not using CPAP.9 Further stratification in this study showed that patients with significant Cheyne-Stokes respirations and central sleep apnea had an improved ejection fraction at 3 months and an 81% reduced mortality-cardiac transplant rate.9

 

 

ADHERENCE

Adherence to PAP therapy is a problem in terms of both frequency of use and duration of use per night. A review of randomized control trials of CPAP compliance between 2011 and 2015 found adherence varied widely from 35% to 87%.10 The average hours of PAP use per night was found to be 5 hours in APPLES.4 Patients adherent to PAP therapy at 1 month remained adherent at 1 year, suggesting patients using CPAP for 1 month were more likely to continue use at 1 year.10 Impediments to PAP use typically involve the facial interface discomfort, lack of humidity, and pressure intolerance.

FEATURES OF PAP DEVICES

Today’s PAP devices have features designed to make them easier to use and more comfortable to improve adherence to therapy. Facial interface options, heated humidifiers, tubing accessories, cleaning devices, reporting of compliance data via telecommunication, and pressure adjustment features of PAP devices may improve patient adherence and comfort, as highlighted in the case scenarios presented below.

Interfaces

Case scenario #1

A 32-year-old woman with moderate sleep apnea complains that her PAP nasal mask is making very loud noises and is disturbing her bed partner. She is a side sleeper and also reports that she wakes with an extremely dry mouth.

Management of the leak could include which of the following?

  1. Chin strap
  2. Avoidance of facial creams before bedtime
  3. CPAP pillow
  4. Clean the mask daily
  5. All of the above

Answer: All of the above.

lance_papimpactonapnea_f2.jpg
%3Cp%3EFigure%202.%20Download%20of%20positive%20airway%20pressure%20use%20data%20for%20a%20month%20(A)%20and%20leak%20data%20for%20a%20night%20(B).%3C%2Fp%3E
Figure 2 shows an overview of data from the patient’s machine for the past month and 1 night of leak data. Both the month-use data and single-night leak data show mask leakage.

There are many types of PAP interfaces such as nasal masks, nasal pillows, nasal cushions, full-face masks, and less frequently used oral and total face masks. The mask interface is a common impediment to use of PAP therapy often due to poor mask fit or leakage.

Nasal masks cover only the nose and require that the mouth remains closed, which can be achieved with the addition of a chin strap. Nasal masks are available in a variety of materials including cloth. Nasal pillows actually go into the nostrils whereas the nasal mask is positioned under the nose. A nasal cushion mask sits under the nose but does not go into the nostrils.

A study by Lanza and colleagues11 evaluated patient comfort with PAP therapy based on the type of nasal interface mask. Patients using nasal pillows had improvement with respect to swollen eyes, discomfort, skin breakdown, and marks on the face compared with patients using nasal masks; however, nasal pillows can cause nostril pain.

Several types of full-face masks are available, some that fit over the bridge of the nose and some that fit just under the nose. A variety of head straps are available to secure full-face masks. One benefit of full-face masks is that air pressure is delivered to both the nose and the mouth, so the mouth can be open or closed. However, the larger surface area of the full-face mask increases the potential for leaks. A study of adherence in 20 patients using CPAP with nasal masks or full-face masks evaluated hours per use, adherence at 12 months, and comfort.12 Patients using full-face masks had more hours per use, better adherence at 12 months, and more comfort than patients using nasal masks.

Interface skin irritation and leak management. To help combat skin irritation, particularly for patients with rosacea, cloth products are available for use beneath the mask and headgear. Silicone pads for masks that cause pressure on the bridge of the nose can help protect against skin breakdown. Sleeping positions other than the supine position can contribute to mask leak. CPAP pillows are designed to allow patients to sleep in their desired position while maintaining an adequate mask seal. The pillows are shaped or have cutouts that prevent the mask from pushing on the pillow and creating a leak.

Humidification

Case scenario #2

A 54-year-old man with severe sleep apnea recently initiated CPAP therapy. He quickly discontinued use due to nasal congestion.

Which of the following is NOT recommended?

  1. Assure adequate heated humidification
  2. Assure that the apnea is adequately treated
  3. Use of a full-face mask
  4. Use of short-acting nasal decongestants
  5. Use of a topical nasal steroid

Answer: Use of short-acting nasal decongestants.

Nasal congestion is a common reason for nonadherence to CPAP therapy.13 Pressurized air is very drying and can be very uncomfortable. Residual apneic events can even precipitate further congestion. The use of humidification with CPAP can improve patient comfort and compliance. The vast majority of patients use CPAP devices with heated humidifiers. Heated humidification has been found to increase CPAP use and improve daytime sleepiness and feelings of satisfaction and being refreshed compared with cold humidity or no humidity.14 Cold humidification improved daytime sleepiness and satisfaction, but not to the degree found with heated humidification.

Heated humidifiers are incorporated in the CPAP machine or attach to it. Heated in-line tubing helps with “rain out,” which refers to water condensation inside the tubing and mask associated with CPAP humidification.

Topical decongestants can actually worsen congestion and cause a reflex vasodilation. Topical nasal steroids can be used for nasal congestion and may be beneficial.

 

 

Tubing

The tubing from the PAP device to the facial interface can be a source of irritation to patients due to rubbing against the skin or entanglement. Products to cover the tubing to reduce irritation and avoid entanglement are available. Extra-long tubing is also available.

Cleaning

Some people find cleaning CPAP equipment daunting. Cleaning devices are available and recommended to patients looking for reassurance about how to keep their CPAP equipment clean. There are also CPAP wipes to clean the mask of oils and creams from the skin to improve the mask seal and reduce leaks.

Pressure control

Advanced modalities are available to adjust how pressure is delivered by PAP devices, including ramp, APAP, pressure relief, and BiPAP. Ramp is a feature that delivers a lower pressure at the beginning of the sleep cycle and slowly increases pressure to therapeutic levels. The lower pressure makes it easier for the user to fall asleep and builds to therapeutic pressure once asleep. APAP adjusts the pressure automatically when needed and reduces the pressure when not needed. Pressure relief is a feature that allows the PAP pressure to decrease at the point of expiration. BiPAP gives a distinct pressure on inspiration and a distinctively different and lower pressure at the point of expiration.

Auto-PAP

Case scenario #3

A 52-year-old woman with hypertension and mild sleep apnea has a polysomnogram with an apnea–hypopnea index of 7 events per hour that increase to 32 events per hour in rapid eye movement (REM) sleep. She is on CPAP at 5 cm of water, but complains of waking every 2 hours with a sense of panic and hot flashes.

Which of the following is the most likely cause of her symptoms?

  1. An underlying anxiety disorder
  2. An underlying heart condition
  3. Perimenopausal symptoms
  4. Undertreated REM-related apnea
  5. None of the above

Answer: While all of these choices can occur, the most likely cause is undertreated REM-related apnea.

lance_papimpactonapnea_f3.jpg
%3Cp%3EFigure%203.%20Sleep%20study%20overview%20showing%20rapid%20eye%20movement%20sleep%20(arrow%2Fblack%20bar)%20associated%20with%20increased%20arousals%20and%20apneic%20events%20and%20decreased%20oxygen%20levels.%3C%2Fp%3E
The sleep study overview for this patient is shown in Figure 3. During REM sleep, arousals and apneas are clustered and associated with a severe drop in oxygen levels. While doing well on CPAP at 5 cm of water, when the patient dreams, the apnea may become worse and more pressure may be needed.

What would be the best next step in treatment for this patient?

  1. Hormonal replacement therapy
  2. Positional therapy in addition to CPAP
  3. APAP
  4. Anxiolytic medication
  5. All of the above

Answer: APAP.

APAP incorporates an algorithm that detects and adjusts to airflow, pressure fluctuations, and airway resistance. The consensus from the American Academy of Sleep Medicine is that APAP is useful in the case of:

  • Pressure intolerance
  • REM apnea or positional apnea
  • Inadequate in lab PAP titration
  • Planned weight loss (bariatric surgery)
  • Recurrent symptoms after long-term CPAP use.15

Pressure relief

Case scenario #4

A 45-year-old man with severe sleep apnea uses CPAP at 10 cm of water. He complains of the inability to exhale against the pressure from the device.

What would be the best next step?

  1. Set the pressure relief to a maximum of 3
  2. Lower the pressure of CPAP and check a download use at a lower pressure
  3. BiPAP titration study in the laboratory
  4. Switch to BiPAP if insurance allows
  5. Change to a different mask

Answer: Set the pressure relief to a maximum of 3.

The CPAP device delivers pressure in conjunction with the patient’s inspiration and expiration. At the point of expiration, there is a decrease in the pressure delivered by the device to make it easier for the user to exhale. Three selectable settings provide flow-based pressure relief with a setting of 1 for the least degree of pressure reduction and a setting of 3 for the greatest degree of pressure reduction.16

In a study of the effect of PAP with pressure relief, 93 patients were assigned to use APAP without pressure relief, CPAP with pressure relief (C-Flex), or APAP with pressure relief (A-Flex).16 At 3 and 6 months, patients using A-Flex had the best adherence to therapy.

Quality of life was also examined in this same study.16 For patients using APAP alone, there was no statistically significant difference in the Epworth Sleepiness Scale measuring daytime sleepiness or the Pittsburgh Sleep Quality Index. However, in patients using A-Flex, daytime sleepiness improved, as did sleep quality, with statistically significant improvement at 3 months.

Bilevel PAP

Case scenario #5

A 62-year-old man with severe sleep apnea uses CPAP set at 17 cm of water and pressure relief set at 3. He stopped using CPAP due to abdominal pain, extreme belching, and pressure intolerance.

What would be the appropriate next step?

  1. Use of simethicone
  2. Elevate the head while using PAP therapy
  3. BiPAP titration study in the laboratory
  4. Switching directly to BiPAP if insurance allows
  5. All of the above

Answer: All of the above.

BiPAP devices provide 2 distinct pressures, one for inhalation and one for exhalation. BiPAP also has the ability to deliver a higher overall pressure. A CPAP device typically has a maximum pressure of 20 cm of water, but BiPAP has a maximum pressure of 25 cm of water on inspiration. BiPAP may be helpful in patients with air aphasia and extreme belching. If a patient cannot tolerate CPAP because of the pressure, and if C-Flex has not alleviated the problem, BiPAP would be the next step.

The effectiveness and level of comfort of BiPAP compared with CPAP for the treatment of OSA was evaluated by the American Academy of Sleep Medicine.2 The analysis of 7 randomized control trials reporting level I and II evidence found that BiPAP was as effective as CPAP in the treatment of OSA in patients with no comorbidities. For patients with OSA and comorbidities, a level III evidence study reported an increased level of comfort in patients using BiPAP.

 

 

PATIENT-CENTERED STRATEGIES TO IMPROVE ADHERENCE

Innovative strategies and approaches focusing on patient factors affecting PAP adherence include motivational interviewing, motivational enhancement, telemedicine, and desensitization techniques.

Motivational interviews and enhancement

Motivational interviewing and motivational enhancement were first used to help with alcohol abuse.17 Motivational interviewing is goal-oriented, patient-centered counseling to elicit a particular behavioral change. The goal is to explore and resolve ambivalence, increase engagement, and evoke a positive response and perspective that builds momentum and results in action.

Motivational enhancement and motivational engagement in the use of PAP therapy were evaluated in the Patient Engagement Study.18 Patients were assigned to usual care (n = 85,358) or active patient engagement (APE) (n = 42,679). Usual care involved diagnosis of apnea, initiation of CPAP, and follow-up, whereas APE included daily feedback (ie, daily scores of apnea–hypopnea index, mask leaks, hours used), positive praise messages, and personal coaching assistance. Overall adherence for patients assigned to APE was 87% compared with 70% in the usual-care group. The hours of use per night also increased for patients in the APE group.

The Best Apnea Interventions for Research trial randomized patients with or at risk of cardiovascular disease (N = 169) to CPAP alone or CPAP with motivational enhancements for 6 months.19 Motivational enhancements and interventions included brief in-person and phone interventions. An overall average difference of 99 minutes per night improvement in CPAP use was reported in the motivational enhancement group compared with CPAP alone.

The Motivational Interviewing Nurse Therapy study trained nurses in motivational interviewing and randomized 106 patients with newly diagnosed OSA to CPAP alone or CPAP plus motivational interviews.20 Motivational interviews involved 3 sessions: 1 to build motivation prior to the CPAP titration, 1 to strengthen the commitment to achieve the prescribed time, and a booster session 1 month after CPAP setup. Adherence was found to improve at 1, 2, and 3 months in the motivational interview group; however, no difference between the 2 groups in adherence was noted at 12 months.

Telemedicine

The role of telemedicine in improving adherence with CPAP therapy was evaluated in 75 patients with moderate to severe apnea randomized to APAP alone or with phone call support from a research coordinator.21 Phone calls occurred 2 days after device setup, and daily monitoring of several factors was done via modem. Patients were contacted if the mask was leaking more than 30% of the night, use was less than 4 hours per night on 2 consecutive nights, the apnea–hypopnea index was greater than 10, or the average pressure needed was higher than 16 cm of water. Statistically significant improvement was found in the telemedicine group in mean adherence, minutes used per day, and mean amount of time spent with the patients.

Desensitization to PAP therapy

Case scenario #6

A 33-year-old woman with a history of anxiety and depression and a remote history of abuse as a child was diagnosed with severe apnea. When she tries to use her CPAP, she has a sense of panic and cannot proceed.

Which of the following has NOT been shown to be beneficial in this situation?

  1. Psychologist for behavioral therapy
  2. Desensitization protocol
  3. PAP “NAP”
  4. Short-acting hypnotics
  5. None of the above

Answer: Short-acting hypnotics.

The short-acting hypnotic zaleplon (Sonata) was evaluated in a 1-month study of 88 patients compared with placebo control, and no difference was found between the 2 groups in measures of adherence to therapy or symptoms.22

lance_papimpactonapnea_t1.jpg
A protocol for desensitization to CPAP use is helpful to assist patients in acclimating to therapy. An example of the steps in a desensitization protocol that patients can do at home is provided in Table 1.

PAP NAP. For some people, at-home desensitization is not enough, and a sleep lab session may be needed. A PAP NAP is a daytime study conducted in the sleep lab. Patients do not necessarily sleep, but work with a technologist with a minimal hookup to polysomnography equipment on mask desensitization, as well as biofeedback techniques. PAP NAPs are indicated for patients with claustrophobia, anxiety surrounding PAP therapy, or pressure intolerance.

A study of 99 patients with moderate to severe apnea and insomnia and concomitant psychiatric disorders resistant to CPAP evaluated adherence in a group receiving a PAP NAP (n = 39) compared with a control group (n = 60).23 The PAP NAP group had marked improvement in completion of CPAP titration in the lab, filling the CPAP prescription, and using CPAP more than 5 days a week and more than 4 hours a night.

A new innovative concept called the Sleep Apnea Patient-Centered Outcomes Network is a collaborative group that includes patients, researchers, and clinicians.24 The group addresses issues such as cost, outcomes, and value in the diagnosis and treatment of sleep apnea. A patient-centered website provides forums, education, and data collection capability for researchers (myapnea.org).

SUMMARY

PAP therapy is the gold standard for treatment of patients with moderate to severe OSA, though poor adherence to PAP therapy is a persistent problem. Advanced features in PAP devices such as APAP and other innovative strategies like motivational enhancement and desensitization protocols and PAP NAP are being used to address poor adherence. More randomized controlled trials are needed to evaluate PAP for sleep apnea.

References
  1. Sullivan CE, Issa FG, Berthon-Jones M, Eves L. Reversal of obstructive sleep apnoea by continuous positive airway pressure applied through the nares. Lancet 1981; 1(8225):862–865.
  2. Gay P, Weaver T, Loube D, Iber C. Evaluation of positive airway pressure treatment for sleep related breathing disorders in adults: a review by the Positive Airway Pressure Task Force of the Standards of Practice Committee of the American Academy of Sleep Medicine. Sleep 2006; 29(3):381–401.
  3. Weaver TE, Maislin G, Dinges DF, et al. Relationship between hours of CPAP use and achieving normal levels of sleepiness and daily functioning. Sleep 2007; 30(6):711–719.
  4. Kushida CA, Nichols DA, Holmes TH, et al. Effects of continuous positive airway pressure on neurocognitive function in obstructive sleep apnea patients: the Apnea Positive Pressure Long-term Efficacy Study (APPLES). Sleep 2012; 35(12):1593–1602.
  5. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  6. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardio­vascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  7. Kanagala R, Murali NS, Friedman PA, et al. Obstructive sleep apnea and the recurrence of atrial fibrillation. Circulation 2003; 107(20):2589–2594.
  8. Kaneko Y, Floras JS, Usui K, et al. Cardiovascular effects of continuous positive airway pressure in patients with heart failure and obstructive sleep apnea. N Engl J Med 2003; 348(13):1233–1241.
  9. Sin DD, Logan AG, Fitzgerald FS, Liu PP, Bradley TD. Effects of continuous positive airway pressure on cardiovascular outcomes in heart failure patients with and without Cheyne-Stokes respiration. Circulation 2000; 102(1):61–66.
  10. Tan B, Tan A, Huak CY, Yingjuan M, Siang WH, Poh HP. Adherence to continuous positive airway pressure therapy in Singaporean patients with obstructive sleep apnea. Am J Otolaryngol 2018; 39(5):501–506.
  11. Lanza A, Mariani S, Sommariva M, et al. Continuous positive airway pressure treatment with nasal pillows in obstructive sleep apnea: long-term effectiveness and adherence. Sleep Med 2018; 41:94–99.
  12. Mortimore IL, Whittle AT, Douglas NJ. Comparison of nose and face mask CPAP therapy for sleep apnoea. Thorax 1998; 53(4):290–292.
  13. Morgenthaler TI, Kapen S, Lee-Chiong T, et al. Practice parameters for the medical therapy of obstructive sleep apnea. Sleep 2006; 29(8):1031–1035.
  14. Massie CA, Hart RW, Peralez K, Richards GN. Effects of humidification on nasal symptoms and compliance in sleep apnea patients using continuous positive airway pressure. Chest 1999; 116(2):403–408.
  15. Morgenthaler TI, Aurora RN, Brown T, et al; Standards of Practice Committee of the AASM. Practice parameters for the use of auto­titrating continuous positive airway pressure devices for titrating pressures and treating adult patients with obstructive sleep apnea syndrome: an update for 2007. Sleep 2008; 31(1):141–147.
  16. Chihara Y, Tsuboi T, Hitomi T, et al. Flexible positive airway pressure improves treatment adherence compared with auto-adjusting PAP. Sleep 2013; 36(2):229–236.
  17. Miller WR, Rollnick S. Motivational interviewing: Preparing people to change addictive behavior. New York: Guilford Press; 1991.
  18. Malhotra A, Crocker ME, Willes L, Kelly C, Lynch S, Benjafield AV. Patient engagement using new technology to improve adherence to positive airway pressure therapy: a retrospective analysis. Chest 2018; 153(4):843–850.
  19. Bakker JP, Wang R, Weng J, et al. Motivational enhancement for increasing adherence to CPAP: a randomized controlled trial. Chest 2016; 150(2):337–345.
  20. Olsen S, Smith SS, Oei TP, Douglas J. Motivational interviewing (MINT) improves continuous positive airway pressure (CPAP) acceptance and adherence: a randomized controlled trial. J Consult Clin Psychol 2012; 80(1):151–163.
  21. Fox N, Hirsch-Allen AJ, Goodfellow E, et al. The impact of a telemedicine monitoring system on positive airway pressure adherence in patients with obstructive sleep apnea: a randomized controlled trial. Sleep 2012; 35(4):477–481.
  22. Park JG, Olson EJ, Morgenthaler TI. Impact of zaleplon on continuous positive airway pressure therapy compliance. J Clin Sleep Med 2013; 9(5):439–444.
  23. Krakow B, Ulibarri V, Melendrez D, Kikta S, Togami L, Haynes P. A daytime, abbreviated cardio-respiratory sleep study (CPT 95807-52) to acclimate insomnia patients with sleep disordered breathing to positive airway pressure (PAP-NAP). J Clin Sleep Med 2008; 4(3):212–222.
  24. Redline S, Baker-Goodwin S, Bakker JP, et al; for the Sleep Apnea Patient-Centered Outcomes Network. Patient partnerships transforming sleep medicine research and clinical care: perspectives from the sleep apnea patient-centered outcomes network. J Clin Sleep Med 2016; 12(7):1053–1058.
References
  1. Sullivan CE, Issa FG, Berthon-Jones M, Eves L. Reversal of obstructive sleep apnoea by continuous positive airway pressure applied through the nares. Lancet 1981; 1(8225):862–865.
  2. Gay P, Weaver T, Loube D, Iber C. Evaluation of positive airway pressure treatment for sleep related breathing disorders in adults: a review by the Positive Airway Pressure Task Force of the Standards of Practice Committee of the American Academy of Sleep Medicine. Sleep 2006; 29(3):381–401.
  3. Weaver TE, Maislin G, Dinges DF, et al. Relationship between hours of CPAP use and achieving normal levels of sleepiness and daily functioning. Sleep 2007; 30(6):711–719.
  4. Kushida CA, Nichols DA, Holmes TH, et al. Effects of continuous positive airway pressure on neurocognitive function in obstructive sleep apnea patients: the Apnea Positive Pressure Long-term Efficacy Study (APPLES). Sleep 2012; 35(12):1593–1602.
  5. Becker HF, Jerrentrup A, Ploch T, et al. Effect of nasal continuous positive airway pressure treatment on blood pressure in patients with obstructive sleep apnea. Circulation 2003; 107(1):68–73.
  6. Marin JM, Carrizo SJ, Vicente E, Agusti AGN. Long-term cardio­vascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet 2005; 365(9464):1046–1053.
  7. Kanagala R, Murali NS, Friedman PA, et al. Obstructive sleep apnea and the recurrence of atrial fibrillation. Circulation 2003; 107(20):2589–2594.
  8. Kaneko Y, Floras JS, Usui K, et al. Cardiovascular effects of continuous positive airway pressure in patients with heart failure and obstructive sleep apnea. N Engl J Med 2003; 348(13):1233–1241.
  9. Sin DD, Logan AG, Fitzgerald FS, Liu PP, Bradley TD. Effects of continuous positive airway pressure on cardiovascular outcomes in heart failure patients with and without Cheyne-Stokes respiration. Circulation 2000; 102(1):61–66.
  10. Tan B, Tan A, Huak CY, Yingjuan M, Siang WH, Poh HP. Adherence to continuous positive airway pressure therapy in Singaporean patients with obstructive sleep apnea. Am J Otolaryngol 2018; 39(5):501–506.
  11. Lanza A, Mariani S, Sommariva M, et al. Continuous positive airway pressure treatment with nasal pillows in obstructive sleep apnea: long-term effectiveness and adherence. Sleep Med 2018; 41:94–99.
  12. Mortimore IL, Whittle AT, Douglas NJ. Comparison of nose and face mask CPAP therapy for sleep apnoea. Thorax 1998; 53(4):290–292.
  13. Morgenthaler TI, Kapen S, Lee-Chiong T, et al. Practice parameters for the medical therapy of obstructive sleep apnea. Sleep 2006; 29(8):1031–1035.
  14. Massie CA, Hart RW, Peralez K, Richards GN. Effects of humidification on nasal symptoms and compliance in sleep apnea patients using continuous positive airway pressure. Chest 1999; 116(2):403–408.
  15. Morgenthaler TI, Aurora RN, Brown T, et al; Standards of Practice Committee of the AASM. Practice parameters for the use of auto­titrating continuous positive airway pressure devices for titrating pressures and treating adult patients with obstructive sleep apnea syndrome: an update for 2007. Sleep 2008; 31(1):141–147.
  16. Chihara Y, Tsuboi T, Hitomi T, et al. Flexible positive airway pressure improves treatment adherence compared with auto-adjusting PAP. Sleep 2013; 36(2):229–236.
  17. Miller WR, Rollnick S. Motivational interviewing: Preparing people to change addictive behavior. New York: Guilford Press; 1991.
  18. Malhotra A, Crocker ME, Willes L, Kelly C, Lynch S, Benjafield AV. Patient engagement using new technology to improve adherence to positive airway pressure therapy: a retrospective analysis. Chest 2018; 153(4):843–850.
  19. Bakker JP, Wang R, Weng J, et al. Motivational enhancement for increasing adherence to CPAP: a randomized controlled trial. Chest 2016; 150(2):337–345.
  20. Olsen S, Smith SS, Oei TP, Douglas J. Motivational interviewing (MINT) improves continuous positive airway pressure (CPAP) acceptance and adherence: a randomized controlled trial. J Consult Clin Psychol 2012; 80(1):151–163.
  21. Fox N, Hirsch-Allen AJ, Goodfellow E, et al. The impact of a telemedicine monitoring system on positive airway pressure adherence in patients with obstructive sleep apnea: a randomized controlled trial. Sleep 2012; 35(4):477–481.
  22. Park JG, Olson EJ, Morgenthaler TI. Impact of zaleplon on continuous positive airway pressure therapy compliance. J Clin Sleep Med 2013; 9(5):439–444.
  23. Krakow B, Ulibarri V, Melendrez D, Kikta S, Togami L, Haynes P. A daytime, abbreviated cardio-respiratory sleep study (CPT 95807-52) to acclimate insomnia patients with sleep disordered breathing to positive airway pressure (PAP-NAP). J Clin Sleep Med 2008; 4(3):212–222.
  24. Redline S, Baker-Goodwin S, Bakker JP, et al; for the Sleep Apnea Patient-Centered Outcomes Network. Patient partnerships transforming sleep medicine research and clinical care: perspectives from the sleep apnea patient-centered outcomes network. J Clin Sleep Med 2016; 12(7):1053–1058.
Page Number
26-33
Page Number
26-33
Publications
Publications
Topics
Article Type
Display Headline
Positive airway pressure: Making an impact on sleep apnea
Display Headline
Positive airway pressure: Making an impact on sleep apnea
Legacy Keywords
Positive airway pressure, PAP, continuous positive airway pressure, CPAP, auto-positive airway pressure, APAP, bilevel positive airway pressure, BiPAP, colleen lance, nancy foldvary-schaefer
Legacy Keywords
Positive airway pressure, PAP, continuous positive airway pressure, CPAP, auto-positive airway pressure, APAP, bilevel positive airway pressure, BiPAP, colleen lance, nancy foldvary-schaefer
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2019 September;86(9 suppl 1):26-33
Inside the Article

KEY POINTS

  • PAP therapy is the gold standard treatment for moderate to severe sleep apnea.
  • Adherence to PAP therapy remains a challenge due to the PAP device itself and various patient comfort factors.
  • Features of PAP devices that may improve adherence are advanced pressure control, including ramp, auto and bilevel, heated humidification, and compliance data reporting.
  • Strategies to motivate patients to use PAP therapy include motivational interviewing, desensitization, and PAP “NAPs.”
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 07:30
Un-Gate On Date
Wed, 08/28/2019 - 07:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 07:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Alternative interventions for obstructive sleep apnea

Article Type
Changed
Tue, 09/03/2019 - 12:11
Display Headline
Alternative interventions for obstructive sleep apnea

The most widely used treatment for patients with obstructive sleep apnea (OSA) is positive airway pressure (PAP) therapy. Improved quality of life and cardiovascular outcomes for patients with OSA using PAP have been demonstrated. However, for some patients with OSA, PAP therapy is difficult to use or tolerate. Fortunately, there are other available treatment interventions for patients with OSA such as lifestyle interventions, surgical interventions, hypoglossal nerve stimulation (HNS), oral appliance therapy (OAT), and expiratory PAP (EPAP) devices. These alternative treatments can also improve symptoms of OSA though data regarding cardiovascular outcomes are lacking.

LIFESTYLE INTERVENTIONS

Weight loss

Because a higher body mass index (BMI) increases the risk for OSA, weight loss should be recommended for patients with OSA who are overweight. Much of the research evaluating the effect of weight loss on OSA has methodologic limitations such as lack of randomization or controls, potential confounding variables, and limited follow-up. A randomized controlled trial of 72 overweight patients with mild OSA (apnea–hypopnea index [AHI] of 5 to 15) compared a group assigned to a very low calorie diet and lifestyle counseling with a control group.1 At 1 year, weight loss of 15 kg or more resulted in a statistically significant reduction in their AHI to normal, resolving their OSA. A 15 kg weight loss in this study was associated with an overall reduction in the AHI of at least 2 units.

Exercise

Exercise is also recommended for patients with OSA, and it can lessen the severity of symptoms even without weight loss. A meta-analysis of 5 randomized trials of 129 patients reported a reduction in the AHI of as much as 6 events per hour in individuals assigned to a strict exercise regimen.2 The reduction in the AHI occurred despite a slight reduction in BMI (1.37 kg/m2).

Sleep position

For some patients, sleeping in the supine position may worsen their OSA, in which case avoiding the supine sleep position is recommended. A sleep study such as polysomnography should be performed to confirm the resolution of OSA in the nonsupine position.3 Products such as pillows or vibratory feedback devices can help the patient avoid sleeping on the back. The ability to monitor patient adherence to sleep position therapy alone is very limited.

Alcohol avoidance

Alcohol consumption depresses the central nervous system, promotes waking, and increases daytime sleepiness, thus exacerbating OSA. Patients with untreated OSA should avoid alcohol because it worsens the duration and frequency of obstructive respiratory events during sleep, and it can worsen the degree of oxygen desaturation that occurs during abnormal respiratory events.4

Concomitant medications

A review of medications in patients with OSA is warranted. Use of benzodiazepines, benzodiazepine-receptor agonists, barbiturates, and opiates in patients with OSA should be avoided especially if OSA is untreated. If these medications are necessary, careful monitoring is recommended. Medications that can cause weight gain such as some antidepressants should also be avoided.

SURGICAL INTERVENTIONS

Surgical interventions for OSA target the location of the obstruction in the upper airway. The upper airway consists of 3 regions: the palate, oropharynx, and larynx.5 More than 30 surgical soft-tissue and skeletal interventions for OSA are reported in the literature.6

Evaluating the outcomes of various surgical interventions for OSA is hindered by differences in the definition of surgical success or cure. As such, surgical interventions for OSA remain controversial. The practice parameters from 2010 reviewed surgical modifications of the upper airway for adults with OSA.7,8 Success is defined as a greater than 50% reduction in the AHI to fewer than 20 events per hour, whereas surgical cure is defined as a reduction in the AHI to fewer than 5 events per hour.7

waters_osaalternativeinterventions_t1.jpg
Table 1 lists commonly used surgical procedures for OSA and reported outcomes, though the quality of evidence to evaluate these procedures is low.8

Uvulopalatopharyngoplasty

Uvulopalatopharyngoplasty (UPPP) is a surgical procedure that remodels the throat via removal of the tonsils and the posterior surface of the soft palate and uvula and closure of the tonsillar pillars, and thus addresses retropalatal collapse. UPPP rarely achieves a surgical cure (ie, AHI < 5) and has been shown to have a 33% reduction in the AHI, with a postoperative average AHI remaining elevated at 29.8 (ie, moderate to severe OSA).8 In general, 50% of patients have a 50% reduction in AHI.9 The 4-year responder rate for UPPP is 44% to 50%.10 Factors limiting the long-term success of this procedure include weight gain, assessment of surgical candidates,11 and decreased adherence to PAP therapy after the procedure.

The use of UPPP in combination with other surgical procedures has also been evaluated.8 The AHI in patients with OSA improved postoperatively when UPPP was done simultaneously or in a multiphase approach with radiofrequency ablation, midline glossectomy, tongue advancement, hyoid suspension, or maxillomandibular advancement, though greater improvement was noted with the multiphase approach.

waters_osaalternativeinterventions_dise_sidebar.jpg

Maxillomandibular advancement

Maxillomandibular advancement is a surgical procedure that moves the maxilla and mandible forward and expands the facial skeletal framework via LeFort I maxillary and sagittal split mandibular osteotomies. Maxillomandibular advancement achieves enlargement of the nasopharyngeal, retropalatal, and hypopharyngeal airway. This increases tension on the pharyngeal soft tissue, which enlarges the medial-lateral and anteroposterior dimensions of the upper airway.14

A meta-analysis of 45 studies evaluated the change in the AHI after maxillomandibular advancement in 518 patients.15 Secondary outcomes were surgical success (> 50% reduction in AHI to < 20 events per hour) and surgical cure (AHI < 5). Patients with a higher preoperative AHI achieved the greatest magnitude reduction in AHI but were less likely to achieve surgical success or cure. Patients with a lower preoperative AHI had a greater likelihood of surgical success and cure.

Bariatric surgery

Bariatric surgery is increasingly used for treatment of OSA in individuals with morbid obesity. A systematic review of bariatric surgery including the roux-en-Y gastric bypass, laparoscopic sleeve gastrectomy, and biliopancreatic diversion evaluated 69 studies with 13,900 patients with OSA.16 OSA was found to be improved or eliminated in 75% of patients for all bariatric surgery procedures.

 

 

HYPOGLOSSAL NERVE STIMULATION

waters_osaalternativeinterventions_f1.jpg
%3Cp%3EFigure%201.%20Hypoglossal%20nerve%20stimulation%20consists%20of%20an%20implanted%20pulse%20generator%2C%20sensing%20lead%2C%20and%20stimulation%20lead.%3C%2Fp%3E
HNS, or upper airway stimulation, is a new, fully implantable treatment for patients with OSA. The system consists of an implanted pulse generator (IPG), sensing lead, and stimulation lead.17 The device is implanted unilaterally via incisions under the clavicle for the IPG, on the chest for the sensing lead, and on the neck for the stimulation lead (Figure 1).

waters_osaalternativeinterventions_t2.jpg
The IPG contains a battery, computer, and lead connector block. It receives information from the sensing lead, operates timing and output algorithms, conveys energy to the stimulation lead, and also serves as a return electrode for advanced stimulation configurations. The sensing lead monitors breathing during sleep and detects pressure changes in the respiratory cycle and conveys this information to the IPG. The stimulation lead encircles the medial branch of the hypoglossal nerve (cranial nerve XII) with an electrode cuff. Stimulation as generated from the IPG is delivered to key airway muscles, which are controlled by the hypoglossal nerve, primarily the genioglossus muscle responsible for tongue protrusion. The device can be turned on and off with a handheld sleep remote.

Indications and contraindications

The indications and contraindications for HNS are shown in Table 2.

waters_osaalternativeinterventions_hns_sidebar.jpg

Efficacy and outcomes

Stimulation of the hypoglossal nerve results in a multilevel mechanism of action: activation and protrusion of the tongue opens the oropharyngeal airway directly but also affects the retropalatal airway by a palatoglossal coupling action.19 Sleep lab testing with polysomnography is used to titrate the voltage of HNS to achieve an open airway that resolves apneic events and normalizes airflow, breathing patterns, and oxygen saturation levels.

Approval of HNS for OSA by the US Food and Drug Administration was based on findings in the Stimulation Therapy for Apnea Reduction (STAR) trial,17 a prospective trial of 126 patients at 22 centers in the United States and Europe with the primary outcomes of AHI and oxygen desaturation index. Secondary outcomes included quality of life as measured by the Functional Outcomes of Sleep Questionnaire and Epworth Sleepiness Scale (ESS). Patient demographics included mean age 54.5, 83% men, mean BMI of 28 kg/m2, and mean baseline AHI of 34 (ie, severe OSA).

Data at 5 years for 97 of the 126 patients on HNS in the STAR trial is available.20 The AHI was reduced an average of 70% to levels in the mild OSA range.20,21 Overall, 85% of the patients had improved quality-of-life measures after HNS implantation, with increased Functional Outcomes of Sleep Questionnaire scores and ESS scores in the normal range over time. Consistent HNS therapy demonstrated sustained benefits at 5 years. The AHI improved by 50% or to less than 20 in 75% of patients, with 44% having resolved OSA and 78% improved to mild OSA (AHI < 15). Device-related adverse events occurred in 6% (9 of 126) of patients requiring replacement or repositioning of the stimulator or leads.20

Moderate to severe snoring was prevalent at baseline in the STAR trial, but over the course of 5 years, 85% of bed partners of patients on HNS reported no or soft snoring.17,21 Nightly use averaged 80% over 60 months based on patient reporting, with 87% reporting use at least 5 nights per week at 36 weeks.20

In terms of predictors of response to HNS therapy, the oxygen desaturation index is the only characteristic that reached a level of statistical significance; patients with higher levels of oxygen desaturation tended to improve and tolerate therapy better long-term.20 A randomized controlled trial of withdrawal of HNS therapy demonstrated increased AHI and oxygen desaturation index when therapy was withdrawn, followed by improvement when therapy resumed.22

A clinical trial of 20 patients implanted with HNS after its approval in 2014 reported that the mean AHI decreased from 33 before implant to 5.1 after implant.23 The ESS also improved from 10.3 before implant to 6 after implant. Mean adherence to device use was 7 (± 2) hours per night. The average stimulation amplitude was 1.89 (± 0.5) volts after the titration sleep study was completed. Similar reductions in AHI were reported by Huntley et al24 for patients receiving HNS implant at 2 academic centers, with no differences between the 2 cohorts in postoperative AHI.

Adverse events

The adverse events reported with HNS are related to the implant procedure or the device.21 Procedure-related adverse events are incision discomfort, temporary tongue weakness, headache, and mild infection of incisions. The most common device-related adverse event is discomfort from the electrical stimulation. Tongue abrasion can also occur if the tongue protrudes and rubs against a sharp tooth. Dry mouth is also commonly reported.

HNS compared with UPPP

Outcomes in patients with moderate to severe OSA matched for BMI, demographics, and preoperative AHI were evaluated comparing patients undergoing HNS (n = 20) with patients receiving UPPP (n = 20).25 The AHI decreased 29% postoperatively in patients with UPPP compared with 88% in patients with HNS, 65% of which had normalization of their AHI. Surgical success was achieved in 40% of patients in the UPPP group compared with 100% in the HNS group. Greater improvement in daytime sleepiness was noted in patients in the HNS group compared with the UPPP group.

 

 

ORAL APPLIANCE THERAPY

OAT devices help protrude the mandible forward and stabilize it to maintain a more patent airway during sleep. Oral appliances can be custom-made or prefabricated. Oral appliances can be titratable or nontitratable: titration provides a mechanism to adjust mandibular protrusion analogous to PAP titration, whereas the absence of titration holds the mandible in a single position. The most effective oral appliances are custom-made and titratable.

Types of OAT devices

Custom oral appliances. Custom oral appliances are fabricated using digital or physical impressions of the patient’s oral structures. Custom oral appliances are made of biocompatible materials and engage both the maxillary and mandibular arches.

Custom oral appliances are made by a qualified dentist who takes maxillary and mandibular impressions with a bite registration using the George Gauge with 40% to 50% of maximum protrusion. The appliance is fabricated in a laboratory and then fitted to the patient, who is instructed to titrate the device 0.5 mm to 1 mm per week and follow-up with the dentist at 2-week intervals. Once the patient has titrated the device to the point of comfort or improved sleep quality or snoring, polysomnography should be done with the device in place and titrated to improve the AHI as much as possible. Follow-up is recommended at 6 months and annually thereafter.

Prefabricated oral appliances. Prefabricated oral appliances are of the boil-and-bite type, only partially modified to the patient’s oral structures.

Tongue-retaining devices. Another type of oral appliance is a tongue-retaining device, which is designed to hold the tongue forward and can be custom-made or prefabricated.

waters_osaalternativeinterventions_oat_sidebar.jpg

Patient considerations for OAT

waters_osaalternativeinterventions_t3.jpg
OAT is not appropriate for all patients with OSA, and the indications and contraindications for use of OAT are presented in Table 3. If OAT is indicated, several considerations may influence the type of device that is most appropriate for the patient (Table 4).

Practice recommendations

waters_osaalternativeinterventions_t4.jpg
The American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine established clinical practice guidelines and recommendations for OAT in patients with OSA:

  • Prescribed OAT should be done by a qualified dentist, and a custom, titratable appliance is preferred
  • OAT is preferred over no therapy for adults with OSA who are intolerant to PAP or prefer alternative therapies
  • A qualified dentist should provide oversight for dental-related side effects or occlusal changes
  • Follow-up sleep testing should be conducted to confirm efficacy or titrate treatment
  • Periodic office visits with the sleep physician and qualified dentist are recommended.26

The quality of evidence for these recommendations is low, with the exception of use of OAT rather than no therapy, which is considered of moderate quality.

Effects of OAT

Anatomic and physiologic effects. With OAT in place in the mouth, the airway caliber in the lateral dimension are increased, and the airway size at the retropalatal level is increased.27–30 With respect to the tongue, increased genioglossus muscle activity has been reported with OAT.

Side effects. Side effects of OAT include excess salivation, dry mouth, tooth tenderness, soft-tissue changes, jaw discomfort, tooth movement, and occlusal changes such as difficulty chewing in the morning. Feelings of suffocation, vivid dreams, and anxiety have also been reported with OAT.31–33

Efficacy and outcomes

Review of the data on the efficacy of OAT did not illuminate factors that predict treatment success.26 Data indicate that in patients with mild OSA using OAT or PAP therapy, there was no significant difference in the percentage achieving their target AHI; however, patients with moderate to severe OSA had a statistically significant greater odds of achieving their target AHI using PAP therapy compared with OAT. Therefore, OAT should be reserved for patients with severe OSA who cannot use or are intolerant to PAP.

Moderate-grade quality of evidence was reviewed for the established OAT practice recommendations for OSA outcomes before and after use of custom, titratable OAT devices.26 Use of a custom OAT device reduced the mean AHI, increased mean oxygen saturation, decreased the mean oxygen desaturation, decreased the arousal index, decreased the ESS, and increased quality of life compared with values prior to use of OAT.

With respect to adherence and discontinuation, patients using OAT had higher mean adherence and lower discontinuation because of side effects compared with patients using continuous PAP.26

 

 

NASAL EPAP THERAPY

Nasal EPAP is a new treatment for OSA that consists of a mechanical valve worn in each naris at night. The valves have a low inspiratory resistance and a high expiratory resistance thus increased pressure occurs at exhalation.

Pressure at exhalation may counter the airway collapse in OSA. With the mouth closed and use of the nasal valves, the positive pressure during the normal respiratory cycle is utilized to maintain an open airway.34 At the onset and throughout inspiration, the activity of the airway dilator muscles increases. At maximum expiration, right before the end of the expiratory pause, the dilator muscle stops abruptly and the airway is of its smallest caliber. The presence of the nasal valve at this point is thought to act as a pneumatic splint to the airway, and the nasal EPAP helps keep the airway patent during the next inspiratory phase.

Nasal EPAP valves are available in a 30-day starter kit. Intended for single-night use, the kit includes valves of increasing levels of expiration resistance: low (nights 1 and 2), medium (nights 3 and 4), and normal (nights 5–30).

Outcomes of nasal EPAP therapy

A multicenter 30-day in-home trial evaluated efficacy and compliance of nasal EPAP therapy.35 The AHI was reduced by 50% or more in 14 of 34 (41%) patients using nasal EPAP compared with the control group at the 30-day follow-up. The patient-reported compliance with nasal EPAP was 94%. Patients in this study had mild to moderate OSA and did not have obesity or other comorbidities such as pulmonary hypertension or cardiovascular disease.

A randomized controlled trial compared nasal EPAP with a sham device in patients with newly diagnosed or untreated OSA (N = 250) for 3 months.36 A median reduction of 52% in the AHI was noted in the intention-to-treat group (N = 229) during rapid eye movement (REM) and non-REM sleep, though it was statistically significant only during REM sleep and supine sleep. At 3 months, improved OSA was maintained in 42% of the patients using nasal EPAP compared with 10% of patients using a sham device. Improvements in daytime sleepiness and adherence with 88% using EPAP the entire night were also noted.

In a 12-month study of nasal EPAP, 67% of patients (34 of 51) used nasal EPAP for the full trial duration.37 Of patients using nasal EPAP for 12 months, the median AHI was reduced by 71%, the ESS improved, and adherence to full-night use was 89%.

Patient considerations for nasal EPAP

In clinical practice, nasal EPAP therapy requires nasal patency and use of a chin strap in patients with mouth leakage. Nasal EPAP may be recommended for patients who travel frequently and can go without continuous PAP or bilevel PAP for short periods of time, and for patients who do not have significant medical comorbidities.

Side effects and limitations of nasal EPAP

Reported side effects of nasal EPAP include difficulty with exhalation, nasal discomfort, dry mouth, and headache. Nasal EPAP therapy is of limited use in patients with severe OSA and severe oxygen desaturation. The efficacy of nasal EPAP beyond 12 months is unknown. Use of nasal EPAP in patients with prior upper-airway surgery and in combination with other therapies is yet to be evaluated.

References
  1. Tuomilehto HPI, Seppä JM, Partinen MM, et al; Kuopio Sleep Apnea Group. Lifestyle intervention with weight reduction: first-line treatment in mild obstructive sleep apnea. Am J Respir Crit Care Med 2009; 179(4):320–327.
  2. Iftikhar IH, Kline CE, Youngstedt SD. Effects of exercise training on sleep apnea: a meta-analysis. Lung 2014; 192(1):175–184.
  3. de Vries GE, Hoekema A, Doff MHJ, et al. Usage of positional therapy in adults with obstructive sleep apnea. J Clin Sleep Med 2015; 11(2):131–137.
  4. Issa FG, Sullivan CE. Alcohol, snoring and sleep apnea. J Neurol Neurosurg Psychiatry 1982; 45(4):353–359.
  5. Rowley JA, Badr MS. Anatomy and physiology of upper airway obstruction. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th edition. Philadelphia, PA: Elsevier; 2017:1076–1087.
  6. Camacho M, Certal V, Capasso R. Comprehensive review of surgeries for obstructive sleep apnea syndrome. Braz J Otorhinolaryngol 2013; 79(6):780–788.
  7. Aurora RN, Casey KR, Kristo D, et al. Practice parameters for the surgical modifications of the upper airway for obstructive sleep apnea in adults. Sleep 2010; 33(10):1408–1413.
  8. Caples SM, Rowley JA, Prinsell JR, et al. Surgical modifications of the upper airway for obstructive sleep apnea in adults: a systematic review and meta-analysis. Sleep 2010; 33(10):1396–1407.
  9. Khan A, Ramar K, Maddirala S, Friedman O, Pallanch JF, Olson EJ. Uvulopalatopharyngoplasty in the management of obstructive sleep apnea: the Mayo Clinic experience. Mayo Clin Proc 2009; 84(9):795–800.
  10. Larson LH, Carlsson-Nordlander B, Svanborg E. Four-year follow-up after uvulopalatopharyngoplasty in 50 unselected patients with obstructive sleep apnea syndrome. Laryngoscope 1994; 104(11 Pt 1):1362–1368.
  11. Aboussouan LS, Golish JA, Wood BG, Mehta AC, Wood DE, Dinner DS. Dynamic pharyngoscopy in predicting outcome of uvulopalatopharyngoplasty for moderate and severe obstructive sleep apnea. Chest 1995; 107(4):946–951.
  12. Vanderveken OM, Maurer JT, Hohenhorst W, et al. Evaluation of drug-induced sleep endoscopy as a patient selection tool for implanted upper airway stimulation for obstructive sleep apnea. J Clin Sleep Med 2013; 9(5):433–438.
  13. Vroegop AV, Vanderveken OM, Boudewyns AN, et al. Drug-induced sleep endoscopy in sleep-disordered breathing: report on 1,249 cases. Laryngoscope 2014; 124(3):797–802.
  14. Gokce SM, Gorgulu S, Gokce HS, Bengi AO, Karacayli U, Ors F. Evaluation of pharyngeal airway space changes after bimaxillary orthognathic surgery with a 3-dimensional simulation and modeling program. Am J Orthod Dentofacial Orthop 2014; 146(4):477–492.
  15. Zaghi S, Holty J-EC, Certal V, et al. Maxillomandibular advancement for treatment of obstructive sleep apnea: a meta-analysis. JAMA Otolaryngol Head Neck Surg 2016; 142(1):58–66.
  16. Sarkhosh K, Switzer NJ, El-Hadi M, Birch DW, Shi X, Karmali S. The impact of bariatric surgery on obstructive sleep apnea: a systematic review. Obes Surg 2013; 23(3):414–423.
  17. Strollo PJ Jr, Soose RJ, Maurer JT, et al; STAR Trial Group. Upper-airway stimulation for obstructive sleep apnea. N Engl J Med 2014; 370(2):139–149.
  18. Ong AA, Murphey AW, Nguyen SA, et al. Efficacy of upper airway stimulation on collapse patterns observed during drug-induced sedation endoscopy. Otolaryngol Head Neck Surg 2016; 154(5):970–977.
  19. Safiruddin F, Vanderveken OM, de Vries N, et al. Effect of upper-airway stimulation for obstructive sleep apnoea on airway dimensions. Eur Respir J 2015; 45(1):129–138.
  20. Woodson BT, Strohl KP, Soose RJ, et al. Upper airway stimulation for obstructive sleep apnea: 5-year outcomes. Otolaryngol Head Neck Surg 2018; 159(1):194–202.
  21. Woodson BT, Soose RJ, Gillespie MB; STAR Trial Investigators. Three-year outcomes of cranial nerve stimulation for obstructive sleep apnea: the STAR Trial. Otolaryngol Head Neck Surg 2016; 154(1):181–188.
  22. Woodson BT, Gillespie MB, Soose RJ, et al; STAR Trial Investigators. Randomized controlled withdrawal study of upper airway stimulation on OSA: short-and long-term effect. Otolaryngol Head Neck Surg 2014; 151(5):880–887.
  23. Kent DT, Lee JJ, Strollo PJ Jr, Soose RJ. Upper airway stimulation for OSA: early adherence and outcome results of one center. Otolaryngol Head Neck Surg 2016; 155(1):188–193.
  24. Huntley C, Kaffenberger T, Doghramji K, Soose R, Boon M. Upper airway stimulation for treatment of obstructive sleep apnea: an evaluation and comparison of outcomes at two academic centers. J Clin Sleep Med 2017; 13(9):1075–1079.
  25. Shah J, Russell JO, Waters T, Kominsky AH, Trask D. Uvulopalatopharyngoplasty vs CN XII stimulation for treatment of obstructive sleep apnea: a single institution experience. Am J Otolaryngol 2018; 39(3):266–270.
  26. Ramar K, Dort LC, Katz SG, et al. Clinical practice guideline for the treatment of obstructive sleep apnea and snoring with oral appliance therapy: an update for 2015—an American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2015; 11(7):773–827.
  27. Sutherland K, Deane SA, Chan ASL, et al. Comparative effects of two oral appliances on upper airway structure in obstructive sleep apnea. Sleep 2011; 34(4):469–477.
  28. Ryan CF, Love LL, Peat D, Fleetham JA, Lowe AA. Mandibular advancement oral appliance therapy for obstructive sleep apnoea: effect on awake caliber of the velopharynx. Thorax 1999; 54(11):972–977.
  29. Tsuiki S, Ono T, Kuroda T. Mandibular advancement modulates respiratory-related genioglossus electromyographic activity. Sleep Breath 2000; 4(2):53–58.
  30. Lowe AA. Oral appliances for sleep breathing disorders. Principles and Practice of Sleep Medicine. 3rd edition. In: Kryger MH, Roth T, Dement WE, eds. Philadelphia: Saunders; 2000:929–939.
  31. Marklund M. Predictors of long-term orthodontic side effects from mandibular advancement devices in patients with snoring and obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2006; 129(2):214–221.
  32. Hammond RJ, Gotsopoulos H, Shen G, Petocz P, Cistulli PA, Darendeliler MA. A follow-up study of dental and skeletal changes associated with mandibular advancement splint use in obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2007; 132(6):806–814.
  33. Pantin CC, Hillman DR, Tennant M. Dental side effects of an oral device to treat snoring and obstructive sleep apnea. Sleep 1999; 22(2):237–240.
  34. Colrain IM, Brooks S, Black J. A pilot evaluation of a nasal expiratory resistance device for the treatment of obstructive sleep apnea. J Clin Sleep Med 2008; 4(5):426–433.
  35. Rosenthal L, Massie CA, Dolan DC, Loomas B, Kram J, Hart RW. A multicenter, prospective study of a novel nasal EPAP device in the treatment of obstructive sleep apnea: efficacy and 30-day adherence. J Clin Sleep Med 2009; 5(6):532–537.
  36. Berry RB, Kryger MH, Massie CA. A novel nasal expiratory positive airway pressure (EPAP) device for the treatment of obstructive sleep apnea: a randomized controlled trial. Sleep 2011; 34(4):479–485.
  37. Kryger MH, Berry RB, Massie CA. Long-term use of a nasal expiratory positive airway pressure (EPAP) device as a treatment for obstructive sleep apnea (OSA). J Clin Sleep Med 2011; 7(5):449–453.
Article PDF
Author and Disclosure Information

Tina Waters, MD
Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Tina Waters, MD, Medical Principal, Cigna; tina.waters@cigna.com

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

This article is based on Dr. Waters’ “Alternative Interventions for Obstructive Sleep Apnea” webcast released May 23, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Waters.

Publications
Topics
Page Number
34-41
Legacy Keywords
Osa, obstructive sleep apnea, osa surgery, osa conservative interventions, osa lifestyle interventions, osa uvulopalatopharyngoplasty, osa maxillomandibular advancement, osa bariatric surgery, drug-induced sleep endoscopy with osa, hypoglossal nerve stimulation, hns, oral appliance therapy, oat, expiratory positive airway pressure, epap, tina waters, nancy foldvary-schaefer
Sections
Author and Disclosure Information

Tina Waters, MD
Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Tina Waters, MD, Medical Principal, Cigna; tina.waters@cigna.com

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

This article is based on Dr. Waters’ “Alternative Interventions for Obstructive Sleep Apnea” webcast released May 23, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Waters.

Author and Disclosure Information

Tina Waters, MD
Sleep Disorders Center, Neurological Institute, Cleveland Clinic

Correspondence: Tina Waters, MD, Medical Principal, Cigna; tina.waters@cigna.com

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

This article is based on Dr. Waters’ “Alternative Interventions for Obstructive Sleep Apnea” webcast released May 23, 2019, part of the “Obstructive Sleep Apnea: A Cleveland Clinic State-of-the-Art Review” online series (available at www.clevelandclinicmeded.com/online/sleep-apnea). The article was drafted by Cleveland Clinic Journal of Medicine staff and was then reviewed, revised, and approved by Dr. Waters.

Article PDF
Article PDF
Related Articles

The most widely used treatment for patients with obstructive sleep apnea (OSA) is positive airway pressure (PAP) therapy. Improved quality of life and cardiovascular outcomes for patients with OSA using PAP have been demonstrated. However, for some patients with OSA, PAP therapy is difficult to use or tolerate. Fortunately, there are other available treatment interventions for patients with OSA such as lifestyle interventions, surgical interventions, hypoglossal nerve stimulation (HNS), oral appliance therapy (OAT), and expiratory PAP (EPAP) devices. These alternative treatments can also improve symptoms of OSA though data regarding cardiovascular outcomes are lacking.

LIFESTYLE INTERVENTIONS

Weight loss

Because a higher body mass index (BMI) increases the risk for OSA, weight loss should be recommended for patients with OSA who are overweight. Much of the research evaluating the effect of weight loss on OSA has methodologic limitations such as lack of randomization or controls, potential confounding variables, and limited follow-up. A randomized controlled trial of 72 overweight patients with mild OSA (apnea–hypopnea index [AHI] of 5 to 15) compared a group assigned to a very low calorie diet and lifestyle counseling with a control group.1 At 1 year, weight loss of 15 kg or more resulted in a statistically significant reduction in their AHI to normal, resolving their OSA. A 15 kg weight loss in this study was associated with an overall reduction in the AHI of at least 2 units.

Exercise

Exercise is also recommended for patients with OSA, and it can lessen the severity of symptoms even without weight loss. A meta-analysis of 5 randomized trials of 129 patients reported a reduction in the AHI of as much as 6 events per hour in individuals assigned to a strict exercise regimen.2 The reduction in the AHI occurred despite a slight reduction in BMI (1.37 kg/m2).

Sleep position

For some patients, sleeping in the supine position may worsen their OSA, in which case avoiding the supine sleep position is recommended. A sleep study such as polysomnography should be performed to confirm the resolution of OSA in the nonsupine position.3 Products such as pillows or vibratory feedback devices can help the patient avoid sleeping on the back. The ability to monitor patient adherence to sleep position therapy alone is very limited.

Alcohol avoidance

Alcohol consumption depresses the central nervous system, promotes waking, and increases daytime sleepiness, thus exacerbating OSA. Patients with untreated OSA should avoid alcohol because it worsens the duration and frequency of obstructive respiratory events during sleep, and it can worsen the degree of oxygen desaturation that occurs during abnormal respiratory events.4

Concomitant medications

A review of medications in patients with OSA is warranted. Use of benzodiazepines, benzodiazepine-receptor agonists, barbiturates, and opiates in patients with OSA should be avoided especially if OSA is untreated. If these medications are necessary, careful monitoring is recommended. Medications that can cause weight gain such as some antidepressants should also be avoided.

SURGICAL INTERVENTIONS

Surgical interventions for OSA target the location of the obstruction in the upper airway. The upper airway consists of 3 regions: the palate, oropharynx, and larynx.5 More than 30 surgical soft-tissue and skeletal interventions for OSA are reported in the literature.6

Evaluating the outcomes of various surgical interventions for OSA is hindered by differences in the definition of surgical success or cure. As such, surgical interventions for OSA remain controversial. The practice parameters from 2010 reviewed surgical modifications of the upper airway for adults with OSA.7,8 Success is defined as a greater than 50% reduction in the AHI to fewer than 20 events per hour, whereas surgical cure is defined as a reduction in the AHI to fewer than 5 events per hour.7

waters_osaalternativeinterventions_t1.jpg
Table 1 lists commonly used surgical procedures for OSA and reported outcomes, though the quality of evidence to evaluate these procedures is low.8

Uvulopalatopharyngoplasty

Uvulopalatopharyngoplasty (UPPP) is a surgical procedure that remodels the throat via removal of the tonsils and the posterior surface of the soft palate and uvula and closure of the tonsillar pillars, and thus addresses retropalatal collapse. UPPP rarely achieves a surgical cure (ie, AHI < 5) and has been shown to have a 33% reduction in the AHI, with a postoperative average AHI remaining elevated at 29.8 (ie, moderate to severe OSA).8 In general, 50% of patients have a 50% reduction in AHI.9 The 4-year responder rate for UPPP is 44% to 50%.10 Factors limiting the long-term success of this procedure include weight gain, assessment of surgical candidates,11 and decreased adherence to PAP therapy after the procedure.

The use of UPPP in combination with other surgical procedures has also been evaluated.8 The AHI in patients with OSA improved postoperatively when UPPP was done simultaneously or in a multiphase approach with radiofrequency ablation, midline glossectomy, tongue advancement, hyoid suspension, or maxillomandibular advancement, though greater improvement was noted with the multiphase approach.

waters_osaalternativeinterventions_dise_sidebar.jpg

Maxillomandibular advancement

Maxillomandibular advancement is a surgical procedure that moves the maxilla and mandible forward and expands the facial skeletal framework via LeFort I maxillary and sagittal split mandibular osteotomies. Maxillomandibular advancement achieves enlargement of the nasopharyngeal, retropalatal, and hypopharyngeal airway. This increases tension on the pharyngeal soft tissue, which enlarges the medial-lateral and anteroposterior dimensions of the upper airway.14

A meta-analysis of 45 studies evaluated the change in the AHI after maxillomandibular advancement in 518 patients.15 Secondary outcomes were surgical success (> 50% reduction in AHI to < 20 events per hour) and surgical cure (AHI < 5). Patients with a higher preoperative AHI achieved the greatest magnitude reduction in AHI but were less likely to achieve surgical success or cure. Patients with a lower preoperative AHI had a greater likelihood of surgical success and cure.

Bariatric surgery

Bariatric surgery is increasingly used for treatment of OSA in individuals with morbid obesity. A systematic review of bariatric surgery including the roux-en-Y gastric bypass, laparoscopic sleeve gastrectomy, and biliopancreatic diversion evaluated 69 studies with 13,900 patients with OSA.16 OSA was found to be improved or eliminated in 75% of patients for all bariatric surgery procedures.

 

 

HYPOGLOSSAL NERVE STIMULATION

waters_osaalternativeinterventions_f1.jpg
%3Cp%3EFigure%201.%20Hypoglossal%20nerve%20stimulation%20consists%20of%20an%20implanted%20pulse%20generator%2C%20sensing%20lead%2C%20and%20stimulation%20lead.%3C%2Fp%3E
HNS, or upper airway stimulation, is a new, fully implantable treatment for patients with OSA. The system consists of an implanted pulse generator (IPG), sensing lead, and stimulation lead.17 The device is implanted unilaterally via incisions under the clavicle for the IPG, on the chest for the sensing lead, and on the neck for the stimulation lead (Figure 1).

waters_osaalternativeinterventions_t2.jpg
The IPG contains a battery, computer, and lead connector block. It receives information from the sensing lead, operates timing and output algorithms, conveys energy to the stimulation lead, and also serves as a return electrode for advanced stimulation configurations. The sensing lead monitors breathing during sleep and detects pressure changes in the respiratory cycle and conveys this information to the IPG. The stimulation lead encircles the medial branch of the hypoglossal nerve (cranial nerve XII) with an electrode cuff. Stimulation as generated from the IPG is delivered to key airway muscles, which are controlled by the hypoglossal nerve, primarily the genioglossus muscle responsible for tongue protrusion. The device can be turned on and off with a handheld sleep remote.

Indications and contraindications

The indications and contraindications for HNS are shown in Table 2.

waters_osaalternativeinterventions_hns_sidebar.jpg

Efficacy and outcomes

Stimulation of the hypoglossal nerve results in a multilevel mechanism of action: activation and protrusion of the tongue opens the oropharyngeal airway directly but also affects the retropalatal airway by a palatoglossal coupling action.19 Sleep lab testing with polysomnography is used to titrate the voltage of HNS to achieve an open airway that resolves apneic events and normalizes airflow, breathing patterns, and oxygen saturation levels.

Approval of HNS for OSA by the US Food and Drug Administration was based on findings in the Stimulation Therapy for Apnea Reduction (STAR) trial,17 a prospective trial of 126 patients at 22 centers in the United States and Europe with the primary outcomes of AHI and oxygen desaturation index. Secondary outcomes included quality of life as measured by the Functional Outcomes of Sleep Questionnaire and Epworth Sleepiness Scale (ESS). Patient demographics included mean age 54.5, 83% men, mean BMI of 28 kg/m2, and mean baseline AHI of 34 (ie, severe OSA).

Data at 5 years for 97 of the 126 patients on HNS in the STAR trial is available.20 The AHI was reduced an average of 70% to levels in the mild OSA range.20,21 Overall, 85% of the patients had improved quality-of-life measures after HNS implantation, with increased Functional Outcomes of Sleep Questionnaire scores and ESS scores in the normal range over time. Consistent HNS therapy demonstrated sustained benefits at 5 years. The AHI improved by 50% or to less than 20 in 75% of patients, with 44% having resolved OSA and 78% improved to mild OSA (AHI < 15). Device-related adverse events occurred in 6% (9 of 126) of patients requiring replacement or repositioning of the stimulator or leads.20

Moderate to severe snoring was prevalent at baseline in the STAR trial, but over the course of 5 years, 85% of bed partners of patients on HNS reported no or soft snoring.17,21 Nightly use averaged 80% over 60 months based on patient reporting, with 87% reporting use at least 5 nights per week at 36 weeks.20

In terms of predictors of response to HNS therapy, the oxygen desaturation index is the only characteristic that reached a level of statistical significance; patients with higher levels of oxygen desaturation tended to improve and tolerate therapy better long-term.20 A randomized controlled trial of withdrawal of HNS therapy demonstrated increased AHI and oxygen desaturation index when therapy was withdrawn, followed by improvement when therapy resumed.22

A clinical trial of 20 patients implanted with HNS after its approval in 2014 reported that the mean AHI decreased from 33 before implant to 5.1 after implant.23 The ESS also improved from 10.3 before implant to 6 after implant. Mean adherence to device use was 7 (± 2) hours per night. The average stimulation amplitude was 1.89 (± 0.5) volts after the titration sleep study was completed. Similar reductions in AHI were reported by Huntley et al24 for patients receiving HNS implant at 2 academic centers, with no differences between the 2 cohorts in postoperative AHI.

Adverse events

The adverse events reported with HNS are related to the implant procedure or the device.21 Procedure-related adverse events are incision discomfort, temporary tongue weakness, headache, and mild infection of incisions. The most common device-related adverse event is discomfort from the electrical stimulation. Tongue abrasion can also occur if the tongue protrudes and rubs against a sharp tooth. Dry mouth is also commonly reported.

HNS compared with UPPP

Outcomes in patients with moderate to severe OSA matched for BMI, demographics, and preoperative AHI were evaluated comparing patients undergoing HNS (n = 20) with patients receiving UPPP (n = 20).25 The AHI decreased 29% postoperatively in patients with UPPP compared with 88% in patients with HNS, 65% of which had normalization of their AHI. Surgical success was achieved in 40% of patients in the UPPP group compared with 100% in the HNS group. Greater improvement in daytime sleepiness was noted in patients in the HNS group compared with the UPPP group.

 

 

ORAL APPLIANCE THERAPY

OAT devices help protrude the mandible forward and stabilize it to maintain a more patent airway during sleep. Oral appliances can be custom-made or prefabricated. Oral appliances can be titratable or nontitratable: titration provides a mechanism to adjust mandibular protrusion analogous to PAP titration, whereas the absence of titration holds the mandible in a single position. The most effective oral appliances are custom-made and titratable.

Types of OAT devices

Custom oral appliances. Custom oral appliances are fabricated using digital or physical impressions of the patient’s oral structures. Custom oral appliances are made of biocompatible materials and engage both the maxillary and mandibular arches.

Custom oral appliances are made by a qualified dentist who takes maxillary and mandibular impressions with a bite registration using the George Gauge with 40% to 50% of maximum protrusion. The appliance is fabricated in a laboratory and then fitted to the patient, who is instructed to titrate the device 0.5 mm to 1 mm per week and follow-up with the dentist at 2-week intervals. Once the patient has titrated the device to the point of comfort or improved sleep quality or snoring, polysomnography should be done with the device in place and titrated to improve the AHI as much as possible. Follow-up is recommended at 6 months and annually thereafter.

Prefabricated oral appliances. Prefabricated oral appliances are of the boil-and-bite type, only partially modified to the patient’s oral structures.

Tongue-retaining devices. Another type of oral appliance is a tongue-retaining device, which is designed to hold the tongue forward and can be custom-made or prefabricated.

waters_osaalternativeinterventions_oat_sidebar.jpg

Patient considerations for OAT

waters_osaalternativeinterventions_t3.jpg
OAT is not appropriate for all patients with OSA, and the indications and contraindications for use of OAT are presented in Table 3. If OAT is indicated, several considerations may influence the type of device that is most appropriate for the patient (Table 4).

Practice recommendations

waters_osaalternativeinterventions_t4.jpg
The American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine established clinical practice guidelines and recommendations for OAT in patients with OSA:

  • Prescribed OAT should be done by a qualified dentist, and a custom, titratable appliance is preferred
  • OAT is preferred over no therapy for adults with OSA who are intolerant to PAP or prefer alternative therapies
  • A qualified dentist should provide oversight for dental-related side effects or occlusal changes
  • Follow-up sleep testing should be conducted to confirm efficacy or titrate treatment
  • Periodic office visits with the sleep physician and qualified dentist are recommended.26

The quality of evidence for these recommendations is low, with the exception of use of OAT rather than no therapy, which is considered of moderate quality.

Effects of OAT

Anatomic and physiologic effects. With OAT in place in the mouth, the airway caliber in the lateral dimension are increased, and the airway size at the retropalatal level is increased.27–30 With respect to the tongue, increased genioglossus muscle activity has been reported with OAT.

Side effects. Side effects of OAT include excess salivation, dry mouth, tooth tenderness, soft-tissue changes, jaw discomfort, tooth movement, and occlusal changes such as difficulty chewing in the morning. Feelings of suffocation, vivid dreams, and anxiety have also been reported with OAT.31–33

Efficacy and outcomes

Review of the data on the efficacy of OAT did not illuminate factors that predict treatment success.26 Data indicate that in patients with mild OSA using OAT or PAP therapy, there was no significant difference in the percentage achieving their target AHI; however, patients with moderate to severe OSA had a statistically significant greater odds of achieving their target AHI using PAP therapy compared with OAT. Therefore, OAT should be reserved for patients with severe OSA who cannot use or are intolerant to PAP.

Moderate-grade quality of evidence was reviewed for the established OAT practice recommendations for OSA outcomes before and after use of custom, titratable OAT devices.26 Use of a custom OAT device reduced the mean AHI, increased mean oxygen saturation, decreased the mean oxygen desaturation, decreased the arousal index, decreased the ESS, and increased quality of life compared with values prior to use of OAT.

With respect to adherence and discontinuation, patients using OAT had higher mean adherence and lower discontinuation because of side effects compared with patients using continuous PAP.26

 

 

NASAL EPAP THERAPY

Nasal EPAP is a new treatment for OSA that consists of a mechanical valve worn in each naris at night. The valves have a low inspiratory resistance and a high expiratory resistance thus increased pressure occurs at exhalation.

Pressure at exhalation may counter the airway collapse in OSA. With the mouth closed and use of the nasal valves, the positive pressure during the normal respiratory cycle is utilized to maintain an open airway.34 At the onset and throughout inspiration, the activity of the airway dilator muscles increases. At maximum expiration, right before the end of the expiratory pause, the dilator muscle stops abruptly and the airway is of its smallest caliber. The presence of the nasal valve at this point is thought to act as a pneumatic splint to the airway, and the nasal EPAP helps keep the airway patent during the next inspiratory phase.

Nasal EPAP valves are available in a 30-day starter kit. Intended for single-night use, the kit includes valves of increasing levels of expiration resistance: low (nights 1 and 2), medium (nights 3 and 4), and normal (nights 5–30).

Outcomes of nasal EPAP therapy

A multicenter 30-day in-home trial evaluated efficacy and compliance of nasal EPAP therapy.35 The AHI was reduced by 50% or more in 14 of 34 (41%) patients using nasal EPAP compared with the control group at the 30-day follow-up. The patient-reported compliance with nasal EPAP was 94%. Patients in this study had mild to moderate OSA and did not have obesity or other comorbidities such as pulmonary hypertension or cardiovascular disease.

A randomized controlled trial compared nasal EPAP with a sham device in patients with newly diagnosed or untreated OSA (N = 250) for 3 months.36 A median reduction of 52% in the AHI was noted in the intention-to-treat group (N = 229) during rapid eye movement (REM) and non-REM sleep, though it was statistically significant only during REM sleep and supine sleep. At 3 months, improved OSA was maintained in 42% of the patients using nasal EPAP compared with 10% of patients using a sham device. Improvements in daytime sleepiness and adherence with 88% using EPAP the entire night were also noted.

In a 12-month study of nasal EPAP, 67% of patients (34 of 51) used nasal EPAP for the full trial duration.37 Of patients using nasal EPAP for 12 months, the median AHI was reduced by 71%, the ESS improved, and adherence to full-night use was 89%.

Patient considerations for nasal EPAP

In clinical practice, nasal EPAP therapy requires nasal patency and use of a chin strap in patients with mouth leakage. Nasal EPAP may be recommended for patients who travel frequently and can go without continuous PAP or bilevel PAP for short periods of time, and for patients who do not have significant medical comorbidities.

Side effects and limitations of nasal EPAP

Reported side effects of nasal EPAP include difficulty with exhalation, nasal discomfort, dry mouth, and headache. Nasal EPAP therapy is of limited use in patients with severe OSA and severe oxygen desaturation. The efficacy of nasal EPAP beyond 12 months is unknown. Use of nasal EPAP in patients with prior upper-airway surgery and in combination with other therapies is yet to be evaluated.

The most widely used treatment for patients with obstructive sleep apnea (OSA) is positive airway pressure (PAP) therapy. Improved quality of life and cardiovascular outcomes for patients with OSA using PAP have been demonstrated. However, for some patients with OSA, PAP therapy is difficult to use or tolerate. Fortunately, there are other available treatment interventions for patients with OSA such as lifestyle interventions, surgical interventions, hypoglossal nerve stimulation (HNS), oral appliance therapy (OAT), and expiratory PAP (EPAP) devices. These alternative treatments can also improve symptoms of OSA though data regarding cardiovascular outcomes are lacking.

LIFESTYLE INTERVENTIONS

Weight loss

Because a higher body mass index (BMI) increases the risk for OSA, weight loss should be recommended for patients with OSA who are overweight. Much of the research evaluating the effect of weight loss on OSA has methodologic limitations such as lack of randomization or controls, potential confounding variables, and limited follow-up. A randomized controlled trial of 72 overweight patients with mild OSA (apnea–hypopnea index [AHI] of 5 to 15) compared a group assigned to a very low calorie diet and lifestyle counseling with a control group.1 At 1 year, weight loss of 15 kg or more resulted in a statistically significant reduction in their AHI to normal, resolving their OSA. A 15 kg weight loss in this study was associated with an overall reduction in the AHI of at least 2 units.

Exercise

Exercise is also recommended for patients with OSA, and it can lessen the severity of symptoms even without weight loss. A meta-analysis of 5 randomized trials of 129 patients reported a reduction in the AHI of as much as 6 events per hour in individuals assigned to a strict exercise regimen.2 The reduction in the AHI occurred despite a slight reduction in BMI (1.37 kg/m2).

Sleep position

For some patients, sleeping in the supine position may worsen their OSA, in which case avoiding the supine sleep position is recommended. A sleep study such as polysomnography should be performed to confirm the resolution of OSA in the nonsupine position.3 Products such as pillows or vibratory feedback devices can help the patient avoid sleeping on the back. The ability to monitor patient adherence to sleep position therapy alone is very limited.

Alcohol avoidance

Alcohol consumption depresses the central nervous system, promotes waking, and increases daytime sleepiness, thus exacerbating OSA. Patients with untreated OSA should avoid alcohol because it worsens the duration and frequency of obstructive respiratory events during sleep, and it can worsen the degree of oxygen desaturation that occurs during abnormal respiratory events.4

Concomitant medications

A review of medications in patients with OSA is warranted. Use of benzodiazepines, benzodiazepine-receptor agonists, barbiturates, and opiates in patients with OSA should be avoided especially if OSA is untreated. If these medications are necessary, careful monitoring is recommended. Medications that can cause weight gain such as some antidepressants should also be avoided.

SURGICAL INTERVENTIONS

Surgical interventions for OSA target the location of the obstruction in the upper airway. The upper airway consists of 3 regions: the palate, oropharynx, and larynx.5 More than 30 surgical soft-tissue and skeletal interventions for OSA are reported in the literature.6

Evaluating the outcomes of various surgical interventions for OSA is hindered by differences in the definition of surgical success or cure. As such, surgical interventions for OSA remain controversial. The practice parameters from 2010 reviewed surgical modifications of the upper airway for adults with OSA.7,8 Success is defined as a greater than 50% reduction in the AHI to fewer than 20 events per hour, whereas surgical cure is defined as a reduction in the AHI to fewer than 5 events per hour.7

waters_osaalternativeinterventions_t1.jpg
Table 1 lists commonly used surgical procedures for OSA and reported outcomes, though the quality of evidence to evaluate these procedures is low.8

Uvulopalatopharyngoplasty

Uvulopalatopharyngoplasty (UPPP) is a surgical procedure that remodels the throat via removal of the tonsils and the posterior surface of the soft palate and uvula and closure of the tonsillar pillars, and thus addresses retropalatal collapse. UPPP rarely achieves a surgical cure (ie, AHI < 5) and has been shown to have a 33% reduction in the AHI, with a postoperative average AHI remaining elevated at 29.8 (ie, moderate to severe OSA).8 In general, 50% of patients have a 50% reduction in AHI.9 The 4-year responder rate for UPPP is 44% to 50%.10 Factors limiting the long-term success of this procedure include weight gain, assessment of surgical candidates,11 and decreased adherence to PAP therapy after the procedure.

The use of UPPP in combination with other surgical procedures has also been evaluated.8 The AHI in patients with OSA improved postoperatively when UPPP was done simultaneously or in a multiphase approach with radiofrequency ablation, midline glossectomy, tongue advancement, hyoid suspension, or maxillomandibular advancement, though greater improvement was noted with the multiphase approach.

waters_osaalternativeinterventions_dise_sidebar.jpg

Maxillomandibular advancement

Maxillomandibular advancement is a surgical procedure that moves the maxilla and mandible forward and expands the facial skeletal framework via LeFort I maxillary and sagittal split mandibular osteotomies. Maxillomandibular advancement achieves enlargement of the nasopharyngeal, retropalatal, and hypopharyngeal airway. This increases tension on the pharyngeal soft tissue, which enlarges the medial-lateral and anteroposterior dimensions of the upper airway.14

A meta-analysis of 45 studies evaluated the change in the AHI after maxillomandibular advancement in 518 patients.15 Secondary outcomes were surgical success (> 50% reduction in AHI to < 20 events per hour) and surgical cure (AHI < 5). Patients with a higher preoperative AHI achieved the greatest magnitude reduction in AHI but were less likely to achieve surgical success or cure. Patients with a lower preoperative AHI had a greater likelihood of surgical success and cure.

Bariatric surgery

Bariatric surgery is increasingly used for treatment of OSA in individuals with morbid obesity. A systematic review of bariatric surgery including the roux-en-Y gastric bypass, laparoscopic sleeve gastrectomy, and biliopancreatic diversion evaluated 69 studies with 13,900 patients with OSA.16 OSA was found to be improved or eliminated in 75% of patients for all bariatric surgery procedures.

 

 

HYPOGLOSSAL NERVE STIMULATION

waters_osaalternativeinterventions_f1.jpg
%3Cp%3EFigure%201.%20Hypoglossal%20nerve%20stimulation%20consists%20of%20an%20implanted%20pulse%20generator%2C%20sensing%20lead%2C%20and%20stimulation%20lead.%3C%2Fp%3E
HNS, or upper airway stimulation, is a new, fully implantable treatment for patients with OSA. The system consists of an implanted pulse generator (IPG), sensing lead, and stimulation lead.17 The device is implanted unilaterally via incisions under the clavicle for the IPG, on the chest for the sensing lead, and on the neck for the stimulation lead (Figure 1).

waters_osaalternativeinterventions_t2.jpg
The IPG contains a battery, computer, and lead connector block. It receives information from the sensing lead, operates timing and output algorithms, conveys energy to the stimulation lead, and also serves as a return electrode for advanced stimulation configurations. The sensing lead monitors breathing during sleep and detects pressure changes in the respiratory cycle and conveys this information to the IPG. The stimulation lead encircles the medial branch of the hypoglossal nerve (cranial nerve XII) with an electrode cuff. Stimulation as generated from the IPG is delivered to key airway muscles, which are controlled by the hypoglossal nerve, primarily the genioglossus muscle responsible for tongue protrusion. The device can be turned on and off with a handheld sleep remote.

Indications and contraindications

The indications and contraindications for HNS are shown in Table 2.

waters_osaalternativeinterventions_hns_sidebar.jpg

Efficacy and outcomes

Stimulation of the hypoglossal nerve results in a multilevel mechanism of action: activation and protrusion of the tongue opens the oropharyngeal airway directly but also affects the retropalatal airway by a palatoglossal coupling action.19 Sleep lab testing with polysomnography is used to titrate the voltage of HNS to achieve an open airway that resolves apneic events and normalizes airflow, breathing patterns, and oxygen saturation levels.

Approval of HNS for OSA by the US Food and Drug Administration was based on findings in the Stimulation Therapy for Apnea Reduction (STAR) trial,17 a prospective trial of 126 patients at 22 centers in the United States and Europe with the primary outcomes of AHI and oxygen desaturation index. Secondary outcomes included quality of life as measured by the Functional Outcomes of Sleep Questionnaire and Epworth Sleepiness Scale (ESS). Patient demographics included mean age 54.5, 83% men, mean BMI of 28 kg/m2, and mean baseline AHI of 34 (ie, severe OSA).

Data at 5 years for 97 of the 126 patients on HNS in the STAR trial is available.20 The AHI was reduced an average of 70% to levels in the mild OSA range.20,21 Overall, 85% of the patients had improved quality-of-life measures after HNS implantation, with increased Functional Outcomes of Sleep Questionnaire scores and ESS scores in the normal range over time. Consistent HNS therapy demonstrated sustained benefits at 5 years. The AHI improved by 50% or to less than 20 in 75% of patients, with 44% having resolved OSA and 78% improved to mild OSA (AHI < 15). Device-related adverse events occurred in 6% (9 of 126) of patients requiring replacement or repositioning of the stimulator or leads.20

Moderate to severe snoring was prevalent at baseline in the STAR trial, but over the course of 5 years, 85% of bed partners of patients on HNS reported no or soft snoring.17,21 Nightly use averaged 80% over 60 months based on patient reporting, with 87% reporting use at least 5 nights per week at 36 weeks.20

In terms of predictors of response to HNS therapy, the oxygen desaturation index is the only characteristic that reached a level of statistical significance; patients with higher levels of oxygen desaturation tended to improve and tolerate therapy better long-term.20 A randomized controlled trial of withdrawal of HNS therapy demonstrated increased AHI and oxygen desaturation index when therapy was withdrawn, followed by improvement when therapy resumed.22

A clinical trial of 20 patients implanted with HNS after its approval in 2014 reported that the mean AHI decreased from 33 before implant to 5.1 after implant.23 The ESS also improved from 10.3 before implant to 6 after implant. Mean adherence to device use was 7 (± 2) hours per night. The average stimulation amplitude was 1.89 (± 0.5) volts after the titration sleep study was completed. Similar reductions in AHI were reported by Huntley et al24 for patients receiving HNS implant at 2 academic centers, with no differences between the 2 cohorts in postoperative AHI.

Adverse events

The adverse events reported with HNS are related to the implant procedure or the device.21 Procedure-related adverse events are incision discomfort, temporary tongue weakness, headache, and mild infection of incisions. The most common device-related adverse event is discomfort from the electrical stimulation. Tongue abrasion can also occur if the tongue protrudes and rubs against a sharp tooth. Dry mouth is also commonly reported.

HNS compared with UPPP

Outcomes in patients with moderate to severe OSA matched for BMI, demographics, and preoperative AHI were evaluated comparing patients undergoing HNS (n = 20) with patients receiving UPPP (n = 20).25 The AHI decreased 29% postoperatively in patients with UPPP compared with 88% in patients with HNS, 65% of which had normalization of their AHI. Surgical success was achieved in 40% of patients in the UPPP group compared with 100% in the HNS group. Greater improvement in daytime sleepiness was noted in patients in the HNS group compared with the UPPP group.

 

 

ORAL APPLIANCE THERAPY

OAT devices help protrude the mandible forward and stabilize it to maintain a more patent airway during sleep. Oral appliances can be custom-made or prefabricated. Oral appliances can be titratable or nontitratable: titration provides a mechanism to adjust mandibular protrusion analogous to PAP titration, whereas the absence of titration holds the mandible in a single position. The most effective oral appliances are custom-made and titratable.

Types of OAT devices

Custom oral appliances. Custom oral appliances are fabricated using digital or physical impressions of the patient’s oral structures. Custom oral appliances are made of biocompatible materials and engage both the maxillary and mandibular arches.

Custom oral appliances are made by a qualified dentist who takes maxillary and mandibular impressions with a bite registration using the George Gauge with 40% to 50% of maximum protrusion. The appliance is fabricated in a laboratory and then fitted to the patient, who is instructed to titrate the device 0.5 mm to 1 mm per week and follow-up with the dentist at 2-week intervals. Once the patient has titrated the device to the point of comfort or improved sleep quality or snoring, polysomnography should be done with the device in place and titrated to improve the AHI as much as possible. Follow-up is recommended at 6 months and annually thereafter.

Prefabricated oral appliances. Prefabricated oral appliances are of the boil-and-bite type, only partially modified to the patient’s oral structures.

Tongue-retaining devices. Another type of oral appliance is a tongue-retaining device, which is designed to hold the tongue forward and can be custom-made or prefabricated.

waters_osaalternativeinterventions_oat_sidebar.jpg

Patient considerations for OAT

waters_osaalternativeinterventions_t3.jpg
OAT is not appropriate for all patients with OSA, and the indications and contraindications for use of OAT are presented in Table 3. If OAT is indicated, several considerations may influence the type of device that is most appropriate for the patient (Table 4).

Practice recommendations

waters_osaalternativeinterventions_t4.jpg
The American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine established clinical practice guidelines and recommendations for OAT in patients with OSA:

  • Prescribed OAT should be done by a qualified dentist, and a custom, titratable appliance is preferred
  • OAT is preferred over no therapy for adults with OSA who are intolerant to PAP or prefer alternative therapies
  • A qualified dentist should provide oversight for dental-related side effects or occlusal changes
  • Follow-up sleep testing should be conducted to confirm efficacy or titrate treatment
  • Periodic office visits with the sleep physician and qualified dentist are recommended.26

The quality of evidence for these recommendations is low, with the exception of use of OAT rather than no therapy, which is considered of moderate quality.

Effects of OAT

Anatomic and physiologic effects. With OAT in place in the mouth, the airway caliber in the lateral dimension are increased, and the airway size at the retropalatal level is increased.27–30 With respect to the tongue, increased genioglossus muscle activity has been reported with OAT.

Side effects. Side effects of OAT include excess salivation, dry mouth, tooth tenderness, soft-tissue changes, jaw discomfort, tooth movement, and occlusal changes such as difficulty chewing in the morning. Feelings of suffocation, vivid dreams, and anxiety have also been reported with OAT.31–33

Efficacy and outcomes

Review of the data on the efficacy of OAT did not illuminate factors that predict treatment success.26 Data indicate that in patients with mild OSA using OAT or PAP therapy, there was no significant difference in the percentage achieving their target AHI; however, patients with moderate to severe OSA had a statistically significant greater odds of achieving their target AHI using PAP therapy compared with OAT. Therefore, OAT should be reserved for patients with severe OSA who cannot use or are intolerant to PAP.

Moderate-grade quality of evidence was reviewed for the established OAT practice recommendations for OSA outcomes before and after use of custom, titratable OAT devices.26 Use of a custom OAT device reduced the mean AHI, increased mean oxygen saturation, decreased the mean oxygen desaturation, decreased the arousal index, decreased the ESS, and increased quality of life compared with values prior to use of OAT.

With respect to adherence and discontinuation, patients using OAT had higher mean adherence and lower discontinuation because of side effects compared with patients using continuous PAP.26

 

 

NASAL EPAP THERAPY

Nasal EPAP is a new treatment for OSA that consists of a mechanical valve worn in each naris at night. The valves have a low inspiratory resistance and a high expiratory resistance thus increased pressure occurs at exhalation.

Pressure at exhalation may counter the airway collapse in OSA. With the mouth closed and use of the nasal valves, the positive pressure during the normal respiratory cycle is utilized to maintain an open airway.34 At the onset and throughout inspiration, the activity of the airway dilator muscles increases. At maximum expiration, right before the end of the expiratory pause, the dilator muscle stops abruptly and the airway is of its smallest caliber. The presence of the nasal valve at this point is thought to act as a pneumatic splint to the airway, and the nasal EPAP helps keep the airway patent during the next inspiratory phase.

Nasal EPAP valves are available in a 30-day starter kit. Intended for single-night use, the kit includes valves of increasing levels of expiration resistance: low (nights 1 and 2), medium (nights 3 and 4), and normal (nights 5–30).

Outcomes of nasal EPAP therapy

A multicenter 30-day in-home trial evaluated efficacy and compliance of nasal EPAP therapy.35 The AHI was reduced by 50% or more in 14 of 34 (41%) patients using nasal EPAP compared with the control group at the 30-day follow-up. The patient-reported compliance with nasal EPAP was 94%. Patients in this study had mild to moderate OSA and did not have obesity or other comorbidities such as pulmonary hypertension or cardiovascular disease.

A randomized controlled trial compared nasal EPAP with a sham device in patients with newly diagnosed or untreated OSA (N = 250) for 3 months.36 A median reduction of 52% in the AHI was noted in the intention-to-treat group (N = 229) during rapid eye movement (REM) and non-REM sleep, though it was statistically significant only during REM sleep and supine sleep. At 3 months, improved OSA was maintained in 42% of the patients using nasal EPAP compared with 10% of patients using a sham device. Improvements in daytime sleepiness and adherence with 88% using EPAP the entire night were also noted.

In a 12-month study of nasal EPAP, 67% of patients (34 of 51) used nasal EPAP for the full trial duration.37 Of patients using nasal EPAP for 12 months, the median AHI was reduced by 71%, the ESS improved, and adherence to full-night use was 89%.

Patient considerations for nasal EPAP

In clinical practice, nasal EPAP therapy requires nasal patency and use of a chin strap in patients with mouth leakage. Nasal EPAP may be recommended for patients who travel frequently and can go without continuous PAP or bilevel PAP for short periods of time, and for patients who do not have significant medical comorbidities.

Side effects and limitations of nasal EPAP

Reported side effects of nasal EPAP include difficulty with exhalation, nasal discomfort, dry mouth, and headache. Nasal EPAP therapy is of limited use in patients with severe OSA and severe oxygen desaturation. The efficacy of nasal EPAP beyond 12 months is unknown. Use of nasal EPAP in patients with prior upper-airway surgery and in combination with other therapies is yet to be evaluated.

References
  1. Tuomilehto HPI, Seppä JM, Partinen MM, et al; Kuopio Sleep Apnea Group. Lifestyle intervention with weight reduction: first-line treatment in mild obstructive sleep apnea. Am J Respir Crit Care Med 2009; 179(4):320–327.
  2. Iftikhar IH, Kline CE, Youngstedt SD. Effects of exercise training on sleep apnea: a meta-analysis. Lung 2014; 192(1):175–184.
  3. de Vries GE, Hoekema A, Doff MHJ, et al. Usage of positional therapy in adults with obstructive sleep apnea. J Clin Sleep Med 2015; 11(2):131–137.
  4. Issa FG, Sullivan CE. Alcohol, snoring and sleep apnea. J Neurol Neurosurg Psychiatry 1982; 45(4):353–359.
  5. Rowley JA, Badr MS. Anatomy and physiology of upper airway obstruction. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th edition. Philadelphia, PA: Elsevier; 2017:1076–1087.
  6. Camacho M, Certal V, Capasso R. Comprehensive review of surgeries for obstructive sleep apnea syndrome. Braz J Otorhinolaryngol 2013; 79(6):780–788.
  7. Aurora RN, Casey KR, Kristo D, et al. Practice parameters for the surgical modifications of the upper airway for obstructive sleep apnea in adults. Sleep 2010; 33(10):1408–1413.
  8. Caples SM, Rowley JA, Prinsell JR, et al. Surgical modifications of the upper airway for obstructive sleep apnea in adults: a systematic review and meta-analysis. Sleep 2010; 33(10):1396–1407.
  9. Khan A, Ramar K, Maddirala S, Friedman O, Pallanch JF, Olson EJ. Uvulopalatopharyngoplasty in the management of obstructive sleep apnea: the Mayo Clinic experience. Mayo Clin Proc 2009; 84(9):795–800.
  10. Larson LH, Carlsson-Nordlander B, Svanborg E. Four-year follow-up after uvulopalatopharyngoplasty in 50 unselected patients with obstructive sleep apnea syndrome. Laryngoscope 1994; 104(11 Pt 1):1362–1368.
  11. Aboussouan LS, Golish JA, Wood BG, Mehta AC, Wood DE, Dinner DS. Dynamic pharyngoscopy in predicting outcome of uvulopalatopharyngoplasty for moderate and severe obstructive sleep apnea. Chest 1995; 107(4):946–951.
  12. Vanderveken OM, Maurer JT, Hohenhorst W, et al. Evaluation of drug-induced sleep endoscopy as a patient selection tool for implanted upper airway stimulation for obstructive sleep apnea. J Clin Sleep Med 2013; 9(5):433–438.
  13. Vroegop AV, Vanderveken OM, Boudewyns AN, et al. Drug-induced sleep endoscopy in sleep-disordered breathing: report on 1,249 cases. Laryngoscope 2014; 124(3):797–802.
  14. Gokce SM, Gorgulu S, Gokce HS, Bengi AO, Karacayli U, Ors F. Evaluation of pharyngeal airway space changes after bimaxillary orthognathic surgery with a 3-dimensional simulation and modeling program. Am J Orthod Dentofacial Orthop 2014; 146(4):477–492.
  15. Zaghi S, Holty J-EC, Certal V, et al. Maxillomandibular advancement for treatment of obstructive sleep apnea: a meta-analysis. JAMA Otolaryngol Head Neck Surg 2016; 142(1):58–66.
  16. Sarkhosh K, Switzer NJ, El-Hadi M, Birch DW, Shi X, Karmali S. The impact of bariatric surgery on obstructive sleep apnea: a systematic review. Obes Surg 2013; 23(3):414–423.
  17. Strollo PJ Jr, Soose RJ, Maurer JT, et al; STAR Trial Group. Upper-airway stimulation for obstructive sleep apnea. N Engl J Med 2014; 370(2):139–149.
  18. Ong AA, Murphey AW, Nguyen SA, et al. Efficacy of upper airway stimulation on collapse patterns observed during drug-induced sedation endoscopy. Otolaryngol Head Neck Surg 2016; 154(5):970–977.
  19. Safiruddin F, Vanderveken OM, de Vries N, et al. Effect of upper-airway stimulation for obstructive sleep apnoea on airway dimensions. Eur Respir J 2015; 45(1):129–138.
  20. Woodson BT, Strohl KP, Soose RJ, et al. Upper airway stimulation for obstructive sleep apnea: 5-year outcomes. Otolaryngol Head Neck Surg 2018; 159(1):194–202.
  21. Woodson BT, Soose RJ, Gillespie MB; STAR Trial Investigators. Three-year outcomes of cranial nerve stimulation for obstructive sleep apnea: the STAR Trial. Otolaryngol Head Neck Surg 2016; 154(1):181–188.
  22. Woodson BT, Gillespie MB, Soose RJ, et al; STAR Trial Investigators. Randomized controlled withdrawal study of upper airway stimulation on OSA: short-and long-term effect. Otolaryngol Head Neck Surg 2014; 151(5):880–887.
  23. Kent DT, Lee JJ, Strollo PJ Jr, Soose RJ. Upper airway stimulation for OSA: early adherence and outcome results of one center. Otolaryngol Head Neck Surg 2016; 155(1):188–193.
  24. Huntley C, Kaffenberger T, Doghramji K, Soose R, Boon M. Upper airway stimulation for treatment of obstructive sleep apnea: an evaluation and comparison of outcomes at two academic centers. J Clin Sleep Med 2017; 13(9):1075–1079.
  25. Shah J, Russell JO, Waters T, Kominsky AH, Trask D. Uvulopalatopharyngoplasty vs CN XII stimulation for treatment of obstructive sleep apnea: a single institution experience. Am J Otolaryngol 2018; 39(3):266–270.
  26. Ramar K, Dort LC, Katz SG, et al. Clinical practice guideline for the treatment of obstructive sleep apnea and snoring with oral appliance therapy: an update for 2015—an American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2015; 11(7):773–827.
  27. Sutherland K, Deane SA, Chan ASL, et al. Comparative effects of two oral appliances on upper airway structure in obstructive sleep apnea. Sleep 2011; 34(4):469–477.
  28. Ryan CF, Love LL, Peat D, Fleetham JA, Lowe AA. Mandibular advancement oral appliance therapy for obstructive sleep apnoea: effect on awake caliber of the velopharynx. Thorax 1999; 54(11):972–977.
  29. Tsuiki S, Ono T, Kuroda T. Mandibular advancement modulates respiratory-related genioglossus electromyographic activity. Sleep Breath 2000; 4(2):53–58.
  30. Lowe AA. Oral appliances for sleep breathing disorders. Principles and Practice of Sleep Medicine. 3rd edition. In: Kryger MH, Roth T, Dement WE, eds. Philadelphia: Saunders; 2000:929–939.
  31. Marklund M. Predictors of long-term orthodontic side effects from mandibular advancement devices in patients with snoring and obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2006; 129(2):214–221.
  32. Hammond RJ, Gotsopoulos H, Shen G, Petocz P, Cistulli PA, Darendeliler MA. A follow-up study of dental and skeletal changes associated with mandibular advancement splint use in obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2007; 132(6):806–814.
  33. Pantin CC, Hillman DR, Tennant M. Dental side effects of an oral device to treat snoring and obstructive sleep apnea. Sleep 1999; 22(2):237–240.
  34. Colrain IM, Brooks S, Black J. A pilot evaluation of a nasal expiratory resistance device for the treatment of obstructive sleep apnea. J Clin Sleep Med 2008; 4(5):426–433.
  35. Rosenthal L, Massie CA, Dolan DC, Loomas B, Kram J, Hart RW. A multicenter, prospective study of a novel nasal EPAP device in the treatment of obstructive sleep apnea: efficacy and 30-day adherence. J Clin Sleep Med 2009; 5(6):532–537.
  36. Berry RB, Kryger MH, Massie CA. A novel nasal expiratory positive airway pressure (EPAP) device for the treatment of obstructive sleep apnea: a randomized controlled trial. Sleep 2011; 34(4):479–485.
  37. Kryger MH, Berry RB, Massie CA. Long-term use of a nasal expiratory positive airway pressure (EPAP) device as a treatment for obstructive sleep apnea (OSA). J Clin Sleep Med 2011; 7(5):449–453.
References
  1. Tuomilehto HPI, Seppä JM, Partinen MM, et al; Kuopio Sleep Apnea Group. Lifestyle intervention with weight reduction: first-line treatment in mild obstructive sleep apnea. Am J Respir Crit Care Med 2009; 179(4):320–327.
  2. Iftikhar IH, Kline CE, Youngstedt SD. Effects of exercise training on sleep apnea: a meta-analysis. Lung 2014; 192(1):175–184.
  3. de Vries GE, Hoekema A, Doff MHJ, et al. Usage of positional therapy in adults with obstructive sleep apnea. J Clin Sleep Med 2015; 11(2):131–137.
  4. Issa FG, Sullivan CE. Alcohol, snoring and sleep apnea. J Neurol Neurosurg Psychiatry 1982; 45(4):353–359.
  5. Rowley JA, Badr MS. Anatomy and physiology of upper airway obstruction. In: Kryger MH, Roth T, Dement WC, eds. Principles and Practice of Sleep Medicine. 6th edition. Philadelphia, PA: Elsevier; 2017:1076–1087.
  6. Camacho M, Certal V, Capasso R. Comprehensive review of surgeries for obstructive sleep apnea syndrome. Braz J Otorhinolaryngol 2013; 79(6):780–788.
  7. Aurora RN, Casey KR, Kristo D, et al. Practice parameters for the surgical modifications of the upper airway for obstructive sleep apnea in adults. Sleep 2010; 33(10):1408–1413.
  8. Caples SM, Rowley JA, Prinsell JR, et al. Surgical modifications of the upper airway for obstructive sleep apnea in adults: a systematic review and meta-analysis. Sleep 2010; 33(10):1396–1407.
  9. Khan A, Ramar K, Maddirala S, Friedman O, Pallanch JF, Olson EJ. Uvulopalatopharyngoplasty in the management of obstructive sleep apnea: the Mayo Clinic experience. Mayo Clin Proc 2009; 84(9):795–800.
  10. Larson LH, Carlsson-Nordlander B, Svanborg E. Four-year follow-up after uvulopalatopharyngoplasty in 50 unselected patients with obstructive sleep apnea syndrome. Laryngoscope 1994; 104(11 Pt 1):1362–1368.
  11. Aboussouan LS, Golish JA, Wood BG, Mehta AC, Wood DE, Dinner DS. Dynamic pharyngoscopy in predicting outcome of uvulopalatopharyngoplasty for moderate and severe obstructive sleep apnea. Chest 1995; 107(4):946–951.
  12. Vanderveken OM, Maurer JT, Hohenhorst W, et al. Evaluation of drug-induced sleep endoscopy as a patient selection tool for implanted upper airway stimulation for obstructive sleep apnea. J Clin Sleep Med 2013; 9(5):433–438.
  13. Vroegop AV, Vanderveken OM, Boudewyns AN, et al. Drug-induced sleep endoscopy in sleep-disordered breathing: report on 1,249 cases. Laryngoscope 2014; 124(3):797–802.
  14. Gokce SM, Gorgulu S, Gokce HS, Bengi AO, Karacayli U, Ors F. Evaluation of pharyngeal airway space changes after bimaxillary orthognathic surgery with a 3-dimensional simulation and modeling program. Am J Orthod Dentofacial Orthop 2014; 146(4):477–492.
  15. Zaghi S, Holty J-EC, Certal V, et al. Maxillomandibular advancement for treatment of obstructive sleep apnea: a meta-analysis. JAMA Otolaryngol Head Neck Surg 2016; 142(1):58–66.
  16. Sarkhosh K, Switzer NJ, El-Hadi M, Birch DW, Shi X, Karmali S. The impact of bariatric surgery on obstructive sleep apnea: a systematic review. Obes Surg 2013; 23(3):414–423.
  17. Strollo PJ Jr, Soose RJ, Maurer JT, et al; STAR Trial Group. Upper-airway stimulation for obstructive sleep apnea. N Engl J Med 2014; 370(2):139–149.
  18. Ong AA, Murphey AW, Nguyen SA, et al. Efficacy of upper airway stimulation on collapse patterns observed during drug-induced sedation endoscopy. Otolaryngol Head Neck Surg 2016; 154(5):970–977.
  19. Safiruddin F, Vanderveken OM, de Vries N, et al. Effect of upper-airway stimulation for obstructive sleep apnoea on airway dimensions. Eur Respir J 2015; 45(1):129–138.
  20. Woodson BT, Strohl KP, Soose RJ, et al. Upper airway stimulation for obstructive sleep apnea: 5-year outcomes. Otolaryngol Head Neck Surg 2018; 159(1):194–202.
  21. Woodson BT, Soose RJ, Gillespie MB; STAR Trial Investigators. Three-year outcomes of cranial nerve stimulation for obstructive sleep apnea: the STAR Trial. Otolaryngol Head Neck Surg 2016; 154(1):181–188.
  22. Woodson BT, Gillespie MB, Soose RJ, et al; STAR Trial Investigators. Randomized controlled withdrawal study of upper airway stimulation on OSA: short-and long-term effect. Otolaryngol Head Neck Surg 2014; 151(5):880–887.
  23. Kent DT, Lee JJ, Strollo PJ Jr, Soose RJ. Upper airway stimulation for OSA: early adherence and outcome results of one center. Otolaryngol Head Neck Surg 2016; 155(1):188–193.
  24. Huntley C, Kaffenberger T, Doghramji K, Soose R, Boon M. Upper airway stimulation for treatment of obstructive sleep apnea: an evaluation and comparison of outcomes at two academic centers. J Clin Sleep Med 2017; 13(9):1075–1079.
  25. Shah J, Russell JO, Waters T, Kominsky AH, Trask D. Uvulopalatopharyngoplasty vs CN XII stimulation for treatment of obstructive sleep apnea: a single institution experience. Am J Otolaryngol 2018; 39(3):266–270.
  26. Ramar K, Dort LC, Katz SG, et al. Clinical practice guideline for the treatment of obstructive sleep apnea and snoring with oral appliance therapy: an update for 2015—an American Academy of Sleep Medicine and American Academy of Dental Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2015; 11(7):773–827.
  27. Sutherland K, Deane SA, Chan ASL, et al. Comparative effects of two oral appliances on upper airway structure in obstructive sleep apnea. Sleep 2011; 34(4):469–477.
  28. Ryan CF, Love LL, Peat D, Fleetham JA, Lowe AA. Mandibular advancement oral appliance therapy for obstructive sleep apnoea: effect on awake caliber of the velopharynx. Thorax 1999; 54(11):972–977.
  29. Tsuiki S, Ono T, Kuroda T. Mandibular advancement modulates respiratory-related genioglossus electromyographic activity. Sleep Breath 2000; 4(2):53–58.
  30. Lowe AA. Oral appliances for sleep breathing disorders. Principles and Practice of Sleep Medicine. 3rd edition. In: Kryger MH, Roth T, Dement WE, eds. Philadelphia: Saunders; 2000:929–939.
  31. Marklund M. Predictors of long-term orthodontic side effects from mandibular advancement devices in patients with snoring and obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2006; 129(2):214–221.
  32. Hammond RJ, Gotsopoulos H, Shen G, Petocz P, Cistulli PA, Darendeliler MA. A follow-up study of dental and skeletal changes associated with mandibular advancement splint use in obstructive sleep apnea. Am J Orthod Dentofacial Orthop 2007; 132(6):806–814.
  33. Pantin CC, Hillman DR, Tennant M. Dental side effects of an oral device to treat snoring and obstructive sleep apnea. Sleep 1999; 22(2):237–240.
  34. Colrain IM, Brooks S, Black J. A pilot evaluation of a nasal expiratory resistance device for the treatment of obstructive sleep apnea. J Clin Sleep Med 2008; 4(5):426–433.
  35. Rosenthal L, Massie CA, Dolan DC, Loomas B, Kram J, Hart RW. A multicenter, prospective study of a novel nasal EPAP device in the treatment of obstructive sleep apnea: efficacy and 30-day adherence. J Clin Sleep Med 2009; 5(6):532–537.
  36. Berry RB, Kryger MH, Massie CA. A novel nasal expiratory positive airway pressure (EPAP) device for the treatment of obstructive sleep apnea: a randomized controlled trial. Sleep 2011; 34(4):479–485.
  37. Kryger MH, Berry RB, Massie CA. Long-term use of a nasal expiratory positive airway pressure (EPAP) device as a treatment for obstructive sleep apnea (OSA). J Clin Sleep Med 2011; 7(5):449–453.
Page Number
34-41
Page Number
34-41
Publications
Publications
Topics
Article Type
Display Headline
Alternative interventions for obstructive sleep apnea
Display Headline
Alternative interventions for obstructive sleep apnea
Legacy Keywords
Osa, obstructive sleep apnea, osa surgery, osa conservative interventions, osa lifestyle interventions, osa uvulopalatopharyngoplasty, osa maxillomandibular advancement, osa bariatric surgery, drug-induced sleep endoscopy with osa, hypoglossal nerve stimulation, hns, oral appliance therapy, oat, expiratory positive airway pressure, epap, tina waters, nancy foldvary-schaefer
Legacy Keywords
Osa, obstructive sleep apnea, osa surgery, osa conservative interventions, osa lifestyle interventions, osa uvulopalatopharyngoplasty, osa maxillomandibular advancement, osa bariatric surgery, drug-induced sleep endoscopy with osa, hypoglossal nerve stimulation, hns, oral appliance therapy, oat, expiratory positive airway pressure, epap, tina waters, nancy foldvary-schaefer
Sections
Citation Override
Cleveland Clinic Journal of Medicine 2019 September;86(9 suppl 1):34-41
Inside the Article

KEY POINTS

  • Alternative interventions for OSA are available for patients who cannot use PAP therapy.
  • Lifestyle interventions that may benefit patients with OSA are weight loss, exercise, change in sleep position, alcohol avoidance, and a review of concomitant medications.
  • Surgical interventions for OSA target the airway obstruction and include uvulopalatopharyngoplasty, maxillomandibular advancement, and bariatric surgery. Drug-induced sleep endoscopy is increasingly used to locate airway obstruction in patients with OSA.
  • Alternative device therapies for OSA are the implanted hypoglossal nerve stimulation system, oral appliances, and nasal expiratory PAP therapy valves.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 07:30
Un-Gate On Date
Wed, 08/28/2019 - 07:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 07:30
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Disparities in cardiovascular care: Past, present, and solutions

Article Type
Changed
Mon, 09/16/2019 - 14:17
Display Headline
Disparities in cardiovascular care: Past, present, and solutions

Cardiovascular disease became the leading cause of death in the United States in the early 20th century, and it accounts for nearly half of all deaths in industrialized nations.1 The mortality it inflicts was thought to be shared equally between both sexes and among all age groups and races.2 The cardiology community implemented innovative epidemiologic research, through which risk factors for cardiovascular disease were established.1 The development of coronary care units reduced in-hospital mortality from acute myocardial infarction from 30% to 15%.2–5 Further advances in pharmacology, revascularization, and imaging have aided in the detection and treatment of cardiovascular disease.6 Though cardiovascular disease remains the number-one cause of death worldwide, rates are on the decline.7

For several decades, health disparities have been recognized as a source of pathology in cardiovascular medicine, resulting in inequity of care administration among select populations. In this review, we examine whether the same forward thinking that has resulted in a decline in cardiovascular disease has had an impact on the pervasive disparities in cardiovascular medicine.

DISPARITIES DEFINED

Compared with whites, members of minority groups have a higher burden of chronic diseases, receive lower quality care, and have less access to medical care. Recognizing the potential public health ramifications, in 1999 the US Congress tasked the Institute of Medicine to study and assess the extent of healthcare disparities. This led to the landmark publication, Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care.8

The Institute of Medicine defines disparities in healthcare as racial or ethnic differences in the quality of healthcare that are not due to access-related factors, clinical needs, preferences, and appropriateness of intervention.8 Disparities can also exist according to socioeconomic status and sex.9

In an early study documenting the concept of disparities in cardiovascular disease, Stone and Vanzant10 concluded that heart disease was more common in African Americans than in whites, and that hypertension was the principal cause of cardiovascular disease mortality in African Americans.

youmans_disparitiesincardiovascularcare_f1.jpg
%3Cp%3EFigure%201.%20Avoidable%20deaths%20from%20heart%20disease%2C%20stroke%2C%20and%20hypertensive%20disease%2C%202001%20and%202010.%3C%2Fp%3E

Although avoidable deaths from heart disease, stroke, and hypertensive disease declined between 2001 and 2010, African Americans still have a higher mortality rate than other racial and ethnic groups (Figure 1).11

DISPARITIES AND CARDIOVASCULAR HEALTH

The concept of cardiovascular health was established by the American Heart Association (AHA) in efforts to achieve an additional 20% reduction in cardiovascular disease-related mortality by 2020.7 Cardiovascular health is defined as the absence of clinically manifest cardiovascular disease and is measured by 7 components:

  • Not smoking or abstaining from smoking for at least 1 year
  • A normal body weight, defined as a body mass index less than 25 kg/m2
  • Optimal physical activity, defined as 75 minutes of vigorous physical activity or 150 minutes of moderate-intensity physical activity per week
  • Regular consumption of a healthy diet
  • Total cholesterol below 200 mg/dL
  • Blood pressure less than 120/80 mm Hg
  • Fasting blood sugar below 100 mg/dL.

Nearly 70% of the US population can claim 2, 3, or 4 of these components, but differences exist according to race,12 and 60% of adult white Americans are limited to achieving no more than 3 of these healthy metrics, compared with 70% of adult African Americans and Hispanic Americans.

Smoking

Smoking is a major risk factor for cardiovascular disease.12–14

youmans_disparitiesincardiovascularcare_f2.jpg
%3Cp%3EFigure%202.%20Percentage%20of%20adults%20who%20are%20active%20smokers%2C%202005%20and%202014.%3C%2Fp%3E

During adolescence, white males are more likely to smoke than African American and Hispanic males,12 but this trend reverses in adulthood, when African American men have a higher prevalence of smoking than white men (21.4% vs 19%).7 Rates of lifetime use are highest among American Indian or Alaskan natives and whites (75.9%), followed by African Americans (58.4%), native Hawaiians (56.8%), and Hispanics (56.7%).15 Trends for current smoking are similar (Figure 2).16 Moreover, households with lower socioeconomic status have a higher prevalence of smoking.7

Physical activity

People with a sedentary lifestyle are more likely to die of cardiovascular disease. As many as 250,000 deaths annually in the United States are attributed to lack of regular physical activity.17

Recognizing the potential public health ramifications, the AHA and the 2018 Federal Guidelines on Physical Activity recommend that children engage in 60 minutes of daily physical activity and that adults participate in 150 minutes of moderate-intensity or 75 minutes of vigorous physical activity weekly.18,19

youmans_disparitiesincardiovascularcare_f3.jpg
%3Cp%3EFigure%203.%20Prevalence%20of%20inactivity%3Csup%3Ea%3C%2Fsup%3E%20in%20the%20United%20States%2C%202013.%3C%2Fp%3E%3Chr%3E%3Cp%3E%3Csup%3Ea%3C%2Fsup%3EPercentage%20of%20US%20adults%20eligible%20for%20intensive%20behavioral%20counseling%20for%20cardiovascular%20disease%20prevention%20and%20not%20meeting%20aerobic%20exercise%20guideline%3C%2Fp%3E

In the United States, 15.2% of adolescents reported being physically inactive, according to data published in 2016.7 Similar to most cardiovascular risk factors, minority populations and those of lower socioeconomic status had the worst profiles. The prevalence of physical inactivity was highest in African Americans and Hispanics (Figure 3).20

Studies have shown an association between screen-based sedentary behavior (computers, television, and video games) and cardiovascular disease.21–23 In the United States, 41% of adolescents used computers for activities other than homework for more than 3 hours per day on a school day.7 The pattern of use was highest in African American boys and African American girls, followed by Hispanic girls and Hispanic boys.18 Trends were similar with regard to watching television for more than 3 hours per day.

Sedentary behavior persists into adulthood, with rates of inactivity of 38.3% in African Americans, 40.1% in Hispanics, and 26.3% in white adults.7

 

 

Nutrition and obesity

Nutrition plays a major role in cardiovascular disease, specifically in the pathogenesis of atherosclerotic disease and hypertension.24 Most Americans do not meet dietary recommendations, with minority communities performing worse in specific metrics.7

Dietary patterns are reflected in the rate of obesity in this nation. Studies have shown a direct correlation between obesity and cardiovascular disease such as coronary artery disease, heart failure, and atrial fibrillation.25–28 According to data from the National Health and Nutrition Examination Survey (NHANES), 31% of children between the ages of 2 and 19 years are classified as obese or overweight. The highest rates of obesity are seen in Hispanic and African American boys and girls. The obesity epidemic is disproportionately rampant in children living in households with low income, low education, and high unemployment rates.7,29–31

Despite the risks associated with obesity, only 64.8% of obese adults report being informed by a doctor or health professional that they were overweight. The proportion of obese adults informed that they were overweight was significantly lower for African Americans and Hispanics compared with whites. Similar differences are seen based on socioeconomic status, as middle-income patients were less likely to be informed than those in the higher income strata (62.4% vs 70.6%).7,31

Blood pressure

Hypertension is a well-established risk factor for cardiovascular disease and stroke, and a blood pressure of 120/80 mm Hg or lower is identified as a component of ideal cardiovascular health.

In the United States the prevalence of hypertension in adults older than 20 is 32%.7 The prevalence of hypertension in African Americans is among the highest in the world.32,33 African Americans develop high blood pressure at earlier ages, and their average resting blood pressures are higher than in whites.34,35 For a 45-year-old without hypertension, the 40-year risk of developing hypertension is 92.7% for African Americans and 86% for whites.35 Hypertension is a major risk factor for stroke, and African Americans have a 1.8 times greater rate of fatal stroke than whites.7

In 2013 there were 71,942 deaths attributable to high blood pressure, and the 2011 death rate associated with hypertension was 18.9 per 100,000. By race, the death rate was 17.6 per 100,000 for white males and an alarming 47.1 per 100,000 for African American males; rates were 15.2 per 100,000 for white females and 35.1 per 100,000 for African American females.7

It is unclear what accounts for the racial difference in prevalence in hypertension. Studies have shown that African Americans are more likely than whites to have been told on more than 2 occasions that they have hypertension. And 85.7% of African Americans are aware that they have high blood pressure, compared with 82.7% of whites.14

African Americans and Hispanics have poorer hypertension control compared with whites.36,37 These observed differences cannot be attributed to access alone, as African Americans were more likely to be on higher-intensity blood pressure therapy, whereas Hispanics were more likely to be undertreated.36,38 In a meta-analysis of 13 trials, Peck et al39 showed that African Americans showed a lesser reduction in systolic and diastolic blood pressure when treated with angiotensin-converting enzyme (ACE) inhibitors.

The 2017 American College of Cardiology (ACC) and AHA guidelines for the prevention, detection, evaluation, and management of high blood pressure in adults40 identifies 4 drug classes as reducing cardiovascular disease morbidity and mortality: thiazide diuretics, ACE inhibitors, angiotensin II receptor blockers (ARBs), and calcium channel blockers. Of these 4 classes, thiazide diuretics and calcium channel blockers have been shown to lower blood pressure more effectively in African Americans than renin-angiotensin-aldosterone inhibition with ACE inhibitors or ARBs.

Glycemic control

Type 2 diabetes mellitus secondary to insulin resistance disproportionately affects minority groups, as the prevalence of diabetes mellitus in African Americans is almost twice as high as that in whites, and 35% higher in Hispanics compared with whites.7,41 Based on NHANES data between 1984 and 2004, the prevalence of diabetes mellitus is expected to increase by 99% in whites, 107% in African Americans, and 127% in Hispanics by 2050. Alarmingly, African Americans over age 75 are expected to experience a 606% increase by 2050.42

With regard to mortality, 21.7 deaths per 100,000 population were attributable to diabetes mellitus according to reports by the AHA in 2016. The death rate in white males was 24.3 per 100,000 compared with 44.9 per 100,000 for African Americans males. The associated mortality rate for white women was 16.2 per 100,000, and 35.8 per 100,000 for African American females.7

 

 

DISPARITIES AND CORONARY ARTERY DISEASE CARE

The management of coronary artery disease has evolved from prolonged bed rest to surgical, pharmacologic, and percutaneous revascularization.2,5 Coronary revascularization procedures are now relatively common: 950,000 percutaneous coronary interventions and 397,000 coronary artery bypass procedures were performed in 2010.7

Nevertheless, despite similar clinical presentations, African Americans with acute myocardial infarction were less likely to be referred for coronary artery bypass grafting than whites.43–46 They were also less likely to be given thrombolytics47 and less likely to undergo coronary angiography with percutaneous coronary intervention.48 Similar differences have been reported when comparing Hispanics with whites.49

Some suggest that healthcare access is a key mediator of health disparities.50 In 2009, Hispanics and African Americans accounted for more than 50% of those without health insurance.51 Improved access to healthcare might mitigate the disparity in revascularizations.

Massachusetts was one of the first states to mandate that all residents obtain health insurance. As a result, the uninsured rates declined in African Americans and Hispanics in Massachusetts, but a disparity in revascularization persisted. African Americans and Hispanics were 27% and 16% less likely to undergo revascularization procedures (coronary artery bypass grafting or percutaneous coronary intervention) than whites,51 suggesting that disparities in revascularization are not solely secondary to healthcare access.

These findings are consistent with a 2004 Veterans Administration study,52 in which healthcare access was equal among races. The study showed that African Americans received fewer cardiac procedures after an acute myocardial infarction compared with whites.

Have we made progress? The largest disparity between African Americans and whites in coronary artery disease mortality existed in 1990. The disparity persisted to 2012, and although decreased, it is projected to persist to 2030.53

DISPARITIES IN HEART FAILURE

An estimated 5.7 million Americans have heart failure, and 915,000 new cases are diagnosed annually.7 Unlike coronary artery disease, heart failure is expected to increase in prevalence by 46%, to 8 million Americans with heart failure by 2030.7,54

Our knowledge of disparities in the area of heart failure is derived primarily from epidemiologic studies. The Multi-Ethnic Study of Atherosclerosis55 showed that African Americans (4.6 per 1,000), followed by Hispanics (3.5 per 1,000) had a higher risk of developing heart failure compared with whites (2.4 per 1,000).The higher risk is in part due to disparities in socioeconomic status and prevalence of hypertension, as African Americans accounted for 75% of cases of nonischemic-related heart failure.55 African Americans also have a higher 5-year mortality rate than whites.55

Even though the 5-year mortality rate in heart failure is still 50%, the past 30 years have seen innovations in pharmacologic and device therapy and thus improved outcomes in heart failure patients. Still, significant gaps in the use of guideline-recommended therapies, quality of care, and clinical outcomes persist in contemporary practice for racial minorities with heart failure.

Disparities in inpatient care for heart failure

Patients admitted for heart failure and cared for by a cardiologist are more likely to be discharged on guideline-directed medical therapy, have fewer heart failure readmissions, and lower mortality.56,57 Breathett et al,58 in a study of 104,835 patients hospitalized in an intensive care unit for heart failure, found that primary intensive care by a cardiologist was associated with higher survival in both races. However, in the same study, white patients had a higher odds of receiving care from a cardiologist than African American patients.

Disparities and cardiac resynchronization therapy devices

In one-third of patients with heart failure, conduction delays result in dyssynchronous left ventricular contraction.59 Dyssynchrony leads to reduced cardiac performance, left ventricular remodeling, and increased mortality.56

Cardiac resynchronization therapy (CRT) was approved for clinical use in 2001, and studies have shown that it improves quality of life, exercise tolerance, cardiac performance, and morbidity and mortality rates.59–66 The 2013 ACC/AHA guidelines for the management of heart failure give a class IA recommendation (the highest) for its use in patients with a left ventricular ejection fraction of 35% or less, sinus rhythm, left bundle branch block and a QRS duration of 150 ms or greater, and New York Heart Association class II, III, or ambulatory IV symptoms while on guideline-directed medical therapy.67

Despite these recommendations, racial differences are observed. A study using the Nationwide Inpatient Sample database59 showed that between 2002 and 2010, a total of 374,202 CRT devices were implanted, averaging 41,578 annually. After adjusting for heart failure admissions, the study showed that CRT use was favored in men and in whites.

Another study, using the National Cardiovascular Data Registry,68 looked at patients who received implantable cardiac defibrillators (ICDs) and were eligible to receive CRT. It found that African Americans and Hispanics were less likely than whites to receive CRT, even though they were more likely to meet established criteria.

Disparities and left ventricular assist devices

The Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart failure (REMATCH) trial and Heart Mate II trial demonstrated that left ventricular assist devices (LVADs) were durable options for long-term support for patients with end-stage heart failure.69,70 Studies that examined the role of race and clinical outcomes after LVAD implantation have reported mixed findings.71,72 Few studies have looked at the role racial differences play in accessing LVAD therapy.

Joyce et al73 reviewed data from the Nationwide Inpatient Sample from 2002 to 2003 on patients admitted to the hospital with a primary diagnosis of heart failure or cardiogenic shock. A total of 297,866 patients were included in the study, of whom only 291 underwent LVAD implantation. A multivariate analysis found that factors such as age over 65, female sex, admission to a nonacademic center, geographic region, and African American race adversely influenced access to LVAD therapy.

Breathett et al74 evaluated racial differences in LVAD implantations from 2012 to 2015, a period that corresponds to increased health insurance expansion, and found LVAD implantations increased among African American patients with advanced heart failure, but no other racial or ethnic group.

 

 

Disparities and heart transplant

For patients with end-stage heart failure, orthotopic heart transplant is the most definitive and durable option for long-term survival. According to data from the United Network for Organ Sharing, 62,508 heart transplants were performed from January 1, 1988 to December 31, 2015. Compared with transplants of other solid organs, heart transplant occurs in significantly infrequent rates.

Barriers to transplant include lack of health insurance, considered a surrogate for low socioeconomic status. Hispanics and African Americans are less likely to have private health insurance than non-Hispanic whites, and this difference is magnified among the working poor.

Despite these perceived barriers, Kilic et al75 found that African Americans comprised 16.4% of heart transplant recipients, although they make up only approximately 13% of the US population. They also had significantly shorter wait-list times than whites. On the negative side, African Americans had a higher unadjusted mortality rate than whites (15% vs 12% P = .002). African Americans also tended to receive their transplants at centers with lower transplant volumes and higher transplant mortality rates.

Several other studies also showed that African Americans compared to whites have significantly worse outcomes after transplant.76–79 What accounts for this difference? Kilic et al75 showed that African Americans had the lowest proportion of blood type matching and lowest human leukocyte antigen matching, were younger (because African Americans develop more advanced heart failure at younger ages), had higher serum creatinine levels, and were more often bridged to transplant with an LVAD.

DISPARITIES IN CARDIOVASCULAR RESEARCH

Although the United States has the most sophisticated and robust medical system in the world, select groups have significant differences in delivery and healthcare outcomes. There are many explanations for these differences, but a contributing factor may be the paucity of research dedicated to understand racial and ethnic differences.80

Differences observed in epidemiologic studies may be secondary to pathophysiology, genetic differences, environment, and lifestyle choices. Historically, clinical trials were conducted in homogeneous populations with respect to age (middle-aged), sex (male), and race (white), and the results were generalized to heterogeneous populations.80

Disparities in research have implications in clinical practice. Overall, the primary cause of heart failure is ischemia; however, in African Americans, the primary cause is hypertensive heart disease.81 Studies in hypertension have shown that African Americans have less of a response to neurohormonal blockade with ACE inhibitors and beta-blockers than non-African Americans.82 Nevertheless, neurohormonal blockade has become the cornerstone of heart failure treatment.

Retrospective analysis of the Vasodilator-Heart Failure trials83 showed that treatment with isosorbide dinitrate plus hydralazine, compared with placebo, conferred a survival benefit for African Americans but not whites.80 No survival advantage was noted when isosorbide dinitrate/hydralazine was compared to enalapril in African Americans, although enalapril was superior to isosorbide dinitrate in whites.45 These observations were recognized 10 to 15 years after trial completion, and were only possible because the trials included sufficient numbers of African American patients to complete analysis.

In 1993, the US Congress passed the National Institutes of Health (NIH) Revitalization Act, which established guidelines requiring NIH grant applicants to include minorities in human subject research, as they were historically underrepresented in clinical research trials.84,85

In 2001, the Beta-Blocker Evaluation of Survival Trial86 reported its results investigating whether bucindolol, a nonselective beta-blocker, would reduce mortality in patients with advanced heart failure (New York Heart Association class III or IV). This was one of the first trials to prospectively investigate racial and ethnic differences in response to treatment. Though it showed no overall benefit in the use of bucindolol in the treatment of advanced heart failure, subgroup analysis showed that whites did enjoy a benefit in terms of lower mortality, whereas African Americans did not.

Results of the Vasodilator-Heart Failure trials led to further population-directed research, most notably the African American Heart Failure Trial,87 a double-blind, placebo-controlled, randomized trial in patients who identified as African American. Patients who were randomized to receive a fixed dose of hydralazine and isosorbide dinitrate had a 43% lower mortality rate, a 33% lower hospitalization rate for heart failure, and better quality of life than patients in the placebo group, leading to early termination of the trial. The outcomes suggested that the combination of isosorbide dinitrate and hydralazine treats heart failure in a manner independent of pure neurohormonal blockade.

CHALLENGES IN STUDY PARTICIPATION

Recruitment of minority participants in biomedical research is a challenging task for clinical investigators.88,89 Some of the factors thought to pose potential barriers for racial and ethnic minority participation in health research include poor access to primary medical care, failure of researchers to recruit minority populations actively, and language and cultural barriers.90

Further, it is widely claimed that African Americans are less willing than nonminority individuals to participate in clinical research trials due to general distrust of the medical community as a result of the Tuskegee Syphilis Experiment.91 That infamous study, conducted by the US Public Health Service between 1932 and 1972, sought to record the natural progression of untreated syphilis in poor African American men in Alabama. The participants were not informed of the true purpose of the study, and they were under the impression that they were simply receiving free healthcare from the US government. Further, they were denied appropriate treatment even after it became readily available, in order for researchers to observe the progression of the disease.

While the 1993 mandate did in fact increase pressure on researchers to develop strategies to overcome participation barriers, the issue of underrepresentation of racial minorities in clinical research, including cardiovascular research, has not been resolved and continues to be a problem today.

The overall goal of clinical research is to determine the best strategies to prevent and treat disease. But if the study population is not representative of the affected population at large, the results cannot be generalized to underrepresented subgroups. The implications of underrepresentation in research are far-reaching, and can further contribute to disparate care of minority patients such as African Americans, who have a higher prevalence of cardiovascular risk factors and greater burden of heart failure.

 

 

PROPOSING SOLUTIONS

Between 1986 and 2018, according to a PUBMED search, 10,462 articles highlighted the presence of a health-related disparity. Solutions to address and ultimately eradicate disparities will need to eliminate healthcare bias, increase patient access, and increase diversity and inclusion in the physician work force.

Eliminating bias

Implicit bias refers to attitudes, thoughts, and feelings that exist outside of the conscious awareness.92 These biases can be triggered by race, gender, or socioeconomic status. They have manifested in society as stereotypes that men are more competent than women, women are more verbal than men, and African Americans are more athletic than whites.93

The concept of implicit bias is important, in that the populations that experience the greatest health disparities also suffer from negative cultural stereotypes.94 Healthcare professionals are not inoculated against implicit bias.95 Studies have shown that most healthcare providers have implicit biases that reflect positive attitudes toward whites and negative attitudes toward people of color.92,94,96–98

The Implicit Association Test, introduced in 1998, is widely used to measure implicit bias. It measures response time of subjects to match particular social groups to particular attributes.99 Green et al,99 using this test, showed that although physicians reported no explicit preference for white vs African American patients or differences in perceived cooperativeness, the test revealed implicit preference favoring white Americans and implicit stereotypes of African Americans as less cooperative for medical procedures and in general. This also manifested in clinical decision-making, as white Americans were more likely, and African Americans less likely, to be treated with thrombolysis.99

Sabin et al100 showed that implicit bias was present among pediatricians, although less than in society as a whole and in other healthcare professionals.

But how does one change feelings that exist outside of the conscious awareness? Green et al99 showed that making physicians aware of their susceptibility to bias changed their behavior. A subset of physicians who were made aware that bias was a focus of the study were more likely to refer African Americans for thrombolysis even if they had a high degree of implicit pro-white bias.94,100 Perhaps mandating that all healthcare providers take a self-administered and confidentially reported Implicit Association Test will lead to awareness of implicit bias and minimize healthcare behaviors that contribute to the current state of disparities.

Improving access

Common indicators of access to healthcare include health insurance status, having a usual source of healthcare, and having a regular physician.101 Health insurance does offer protection from the costs associated with illness and health maintenance.101 It is also a major contributing factor in racial and ethnic disparities.

Chen et al102 examined the effects of the Affordable Care Act and found that it was associated with reduction in the probability of being uninsured, delaying necessary care, and forgoing necessary care, and increased probability of having a physician. However, earlier studies showed that access to health insurance by itself does not equate to equitable care.103,104

Diversifying the work force

African Americans comprise 4% of physicians and Hispanic Americans 5%, despite accounting for 13% and 16% of the US population.105 This underrepresentation has led to African American and Hispanic American patients being more likely than white patients to be treated by a physician from a dissimilar racial or ethnic background.106 Studies have shown that minority patients in a race- or ethnic-concordant relationship are more likely to use needed health services, less likely to postpone seeking care, and report greater satisfaction.106,107 Minority physicians often locate and practice in neighborhoods with high minority populations, and they disproportionately care for disadvantaged patients of lower socioeconomic status and poorer health.106,108

WE ARE STILL IN THE TUNNEL, BUT THERE IS LIGHT AT THE END

The cardiovascular community has faced tremendous challenges in the past and responded with innovative research that has led to imaging that aids in the diagnosis of subclinical cardiovascular disease and invasive and pharmacologic strategies that have improved cardiovascular outcomes. One may say that there is light at the end of the tunnel; however, the existence of disparate care reminds us that we are still in the tunnel.

Disparities in cardiovascular disease management present a unique challenge for the community. There is no drug, device, or invasive procedure to eliminate this pathology. However, by acknowledging the problem and implementing changes at the system, provider, and patient level, the cardiovascular community can achieve yet another momentous achievement: the end of cardiovascular health disparities. Cardiovascular disease makes no distinction in race, sex, age, or socioeconomic status, and neither should the medical community.

References
  1. Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial infarction. N Engl J Med 2012; 366(1):54–63. doi:10.1056/NEJMra1112570
  2. Braunwald E. Evolution of the management of acute myocardial infarction: a 20th century saga. Lancet 1998; 352(9142):1771–1774. doi:10.1016/S0140-6736(98)03212-7
  3. Julian DG. The history of coronary care units. Br Heart J 1987; 57(6):497–502. doi:10.1136/hrt.57.6.497
  4. Caswell JE. A brief history of coronary care units. Public Health Rep 1967; 82(12):1105–1111. pmid:19316519
  5. Day HW. History of coronary care units. Am J Cardiol 1972; 30(4):405–407. pmid:4560377
  6. Braunwald E. Shattuck lecture—cardiovascular medicine at the turn of the millennium: triumphs, concerns, and opportunities. N Engl J Med 1997; 337(19):1360–1369. doi:10.1056/NEJM199711063371906
  7. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee; Stroke Statistics Subcommittee. Heart disease and stroke statistics—2016 update: a report from the American Heart Association. Circulation 2016; 133(4):e38–e360. doi:10.1161/CIR.0000000000000350
  8. Nelson A. Unequal treatment: confronting racial and ethnic disparities in health care. J Natl Med Assoc 2002; 94(8):666–668. pmid:12152921
  9. McGuire TG, Alegria M, Cook BL, Wells KB, Zaslavsky AM. Implementing the Institute of Medicine definition of disparities: an application to mental health care. Health Serv Res 2006; 41(5):1979–2005. doi:10.1111/j.1475-6773.2006.00583.x
  10. Stone CT, Vanzant FR. Heart disease as seen in a southern clinic: a clinical and pathologic survey. JAMA 1927; 89(18):1473–1480. doi:10.1001/jama.1927.02690180005002
  11. Centers For Disease Control And Prevention (CDC). Vital signs: avoidable deaths from heart disease, stroke, and hypertensive disease—United States, 2001–2010. MMWR Morb Mortal Wkly Rep 2013; 62(35):727–727. pmid:PMC4585625
  12. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation 2015; 131(4):e29–e322. doi:10.1161/CIR.0000000000000152
  13. Ockene IS, Miller NH. Cigarette smoking, cardiovascular disease, and stroke: a statement for healthcare professionals from the American Heart Association. American Heart Association Task Force on Risk Reduction. Circulation 1997; 96(9):3243–3247. pmid:9386200
  14. Messner B, Bernhard D. Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis. Arterioscler Thromb Vasc Biol 2014; 34(3):509–515. doi:10.1161/ATVBAHA.113.300156
  15. Benjamin EJ, Virani SS, Callaway CW, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2018 update: a report from the American Heart Association. Circulation 2018; 137(12):e67–e492. doi:10.1161/CIR.0000000000000558
  16. Jamal A, Homa DM, O’Conner E, et al. Current cigarette smoking among adults—United States, 2005–2014. MMWR 2015; 64(44):1233–1240. doi:10.15585/mmwr.mm6444a2
  17. Myers J. Cardiology patient pages. Exercise and cardiovascular health. Circulation 2003; 107(1):e2–e5. pmid:12515760
  18. Shiroma EJ, Lee IM. Physical activity and cardiovascular health: lessons learned from epidemiological studies across age, gender, and race/ethnicity. Circulation 2010; 122(7):743–752. doi:10.1161/CIRCULATIONAHA.109.914721
  19. Piercy KL, Troiano RP, Ballard RM, et al. The physical activity guidelines for Americans. JAMA 2018; 320(19):2020–2028. doi:10.1001/jama.2018.14854
  20. Omura JD, Carlson SA, Paul P, et al. Adults eligible for cardiovascular disease prevention counseling and participation in aerobic physical activity—United States, 2013. MMWR 2015; 64(37):1047–1051. doi:10.15585/mmwr.mm6437a4
  21. Dunstan DW, Barr EL, Healy GN, et al. Television viewing time and mortality: the Australian diabetes, obesity and lifestyle study (AusDiab). Circulation 2010; 121(3):384–391. doi:10.1161/CIRCULATIONAHA.109.894824
  22. Warren TY, Barry V, Hooker SP, Sui X, Church TS, Blair SN. Sedentary behaviors increase risk of cardiovascular disease mortality in men. Med Sci Sports Exerc 2010; 42(5):879–885. doi:10.1249/MSS.0b013e3181c3aa7e
  23. Byun W, Dowda M, Pate RR. Associations between screen-based sedentary behavior and cardiovascular disease risk factors in Korean youth. J Korean Med Sci 2012; 27(4):388–394. doi:10.3346/jkms.2012.27.4.388
  24. Getz GS, Reardon CA. Nutrition and cardiovascular disease. Arterioscler Thromb Vasc Biol 2007; 27(12):2499–2506. doi:10.1161/ATVBAHA.107.155853
  25. Eckel RH. Obesity and heart disease: a statement for healthcare professionals from the Nutrition Committee, American Heart Association. Circulation 1997; 96(9):3248–3250. pmid:9386201
  26. Rimm EB, Stampfer MJ, Giovannucci E, et al. Body size and fat distribution as predictors of coronary heart disease among middle-aged and older US men. Am J Epidemiol 1995; 141(12):1117–1127. pmid:7771450
  27. Duflou J, Virmani R, Rabin I, Burke A, Farb A, Smialek J. Sudden death as a result of heart disease in morbid obesity. Am Heart J 1995; 130(2):306–313. pmid:7631612
  28. Poirier P, Giles TD, Bray GA, et al; American Heart Association; Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation 2006; 113(6):898–918. doi:10.1161/CIRCULATIONAHA.106.171016
  29. Wang Y. Disparities in pediatric obesity in the United States. Adv Nutr 2011; 2(1):23–31. doi:10.3945/an.110.000083
  30. Taveras EM, Gillman MW, Kleinman KP, Rich-Edwards JW, Rifas-Shiman SL. Reducing racial/ethnic disparities in childhood obesity: the role of early life risk factors. JAMA Pediatr 2013; 167(8):731–738. doi:10.1001/jamapediatrics.2013.85
  31. Powell-Wiley TM, Ayers CR, Banks-Richard K, et al. Disparities in counseling for lifestyle modification among obese adults: insights from the Dallas heart study. Obesity (Silver Spring) 2012; 20(4):849–855. doi:10.1038/oby.2011.242
  32. Fuchs FD. Why do black Americans have higher prevalence of hypertension?: an enigma still unsolved. Hypertension 2011; 57(3):379–380. doi:10.1161/HYPERTENSIONAHA.110.163196
  33. Ferdinand KC, Armani AM. The management of hypertension in African Americans. Crit Pathw Cardiol 2007; 6(2):67–71. doi:10.1097/HPC.0b013e318053da59
  34. Voors AW, Webber LS, Berenson GS. Time course study of blood pressure in children over a three-year period. Bogalusa Heart Study. Hypertension 1980; 2(4 Pt 2):102–108. pmid:7399641
  35. Carson AP, Howard G, Burke GL, Shea S, Levitan EB, Muntner P. Ethnic differences in hypertension incidence among middle-aged and older adults: the multi-ethnic study of atherosclerosis. Hypertension 2011; 57(6):1101–1107. doi:10.1161/HYPERTENSIONAHA.110.168005
  36. Gu A, Yue Y, Desai RP, Argulian E. Racial and ethnic differences in antihypertensive medication use and blood pressure control among US adults with hypertension: the National Health and Nutrition Examination Survey, 2003 to 2012. Circ Cardiovasc Qual Outcomes 2017; 10(1). pii:e003166. doi:10.1161/CIRCOUTCOMES.116.003166
  37. Lackland DT. Racial differences in hypertension: implications for high blood pressure management. Am J Med Sci 2014; 348(2):135–138. doi:10.1097/MAJ.0000000000000308
  38. Bosworth HB, Dudley T, Olsen MK, et al. Racial differences in blood pressure control: potential explanatory factors. Am J Med 2006; 119(1):70.e9–e15. doi:10.1016/j.amjmed.2005.08.019
  39. Peck RN, Smart LR, Beier R, et al. Difference in blood pressure response to ACE-Inhibitor monotherapy between black and white adults with arterial hypertension: a meta-analysis of 13 clinical trials. BMC Nephrol 2013; 14:201. doi:10.1186/1471-2369-14-201
  40. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2018; 138(17):e484–e594. doi:10.1161/CIR.0000000000000596
  41. Peek ME, Cargill A, Huang ES. Diabetes health disparities: a systematic review of health care interventions. Med Care Res Rev 2007; 64(5 suppl):101S–156S. doi:10.1177/1077558707305409
  42. Narayan KM, Boyle JP, Geiss LS, Saaddine JB, Thompson TJ. Impact of recent increase in incidence on future diabetes burden: US, 2005–2050. Diabetes Care 2006; 29(9):2114–2116. doi:10.2337/dc06-1136
  43. Johnson PA, Lee TH, Cook EF, Rouan GW, Goldman L. Effect of race on the presentation and management of patients with acute chest pain. Ann Intern Med 1993; 118(8):593–601. pmid:8452325
  44. Hannan EL, van Ryn M, Burke J, et al. Access to coronary artery bypass surgery by race/ethnicity and gender among patients who are appropriate for surgery. Med Care 1999; 37(1):68–77. pmid:10413394
  45. Peterson ED, Shaw LK, DeLong ER, Pryor DB, Califf RM, Mark DB. Racial variation in the use of coronary-revascularization procedures. Are the differences real? Do they matter? N Engl J Med 1997; 336(7):480–486. doi:10.1056/NEJM199702133360706
  46. Wenneker MB, Epstein AM. Racial inequalities in the use of procedures for patients with ischemic heart disease in Massachusetts. JAMA 1989; 261(2):253–257. pmid:2521191
  47. Allison JJ, Kiefe CI, Centor RM, Box JB, Farmer RM. Racial differences in the medical treatment of elderly Medicare patients with acute myocardial infarction. J Gen Intern Med 1996; 11(12):736–743. pmid:9016420
  48. Mickelson JK, Blum CM, Geraci JM. Acute myocardial infarction: clinical characteristics, management and outcome in a metropolitan Veterans Affairs Medical Center teaching hospital. J Am Coll Cardiol 1997; 29(5):915–925. pmid:9120176
  49. Yarzebski J, Bujor CF, Lessard D, Gore JM, Goldberg RJ. Recent and temporal trends (1975 to 1999) in the treatment, hospital, and long-term outcomes of Hispanic and non-Hispanic white patients hospitalized with acute myocardial infarction: a population-based perspective. Am Heart J 2004; 147(4):690–697. doi:10.1016/j.ahj.2003.10.023
  50. Riley WJ. Health disparities: gaps in access, quality and affordability of medical care. Trans Am Clin Climatol Assoc 2012; 123:167–172. pmid:23303983
  51. Albert MA, Ayanian JZ, Silbaugh TS, et al. Early results of Massachusetts healthcare reform on racial, ethnic, and socioeconomic disparities in cardiovascular care. Circulation 2014; 129(24):2528–2538. doi:10.1161/CIRCULATIONAHA.113.005231
  52. Peterson ED, Wright SM, Daley J, Thibault GE. Racial variation in cardiac procedure use and survival following acute myocardial infarction in the Department of Veterans Affairs. JAMA 1994; 271(15):1175–1180. pmid:8151875
  53. Pearson-Stuttard J, Guzman-Castillo M, Penalvo JL, et al. Modeling future cardiovascular disease mortality in the United States: national trends and racial and ethnic disparities. Circulation 2016; 133(10):967–978. doi:10.1161/CIRCULATIONAHA.115.019904
  54. Heidenreich PA, Albert NM, Allen LA, et al; American Heart Association Advocacy Coordinating Committee; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Stroke Council. Forecasting the impact of heart failure in the United States: a policy statement from the American Heart Association. Circ Heart Fail 2013; 6(3):606–619. doi:10.1161/HHF.0b013e318291329a
  55. Bahrami H, Kronmal R, Bluemke DA, et al. Differences in the incidence of congestive heart failure by ethnicity: the multi-ethnic study of atherosclerosis. Arch Intern Med 2008; 168(19):2138–2145. doi:10.1001/archinte.168.19.2138
  56. Parmar KR, Xiu PY, Chowdhury MR, Patel E, Cohen M. In-hospital treatment and outcomes of heart failure in specialist and non-specialist services: a retrospective cohort study in the elderly. Open Heart 2015; 2(1):e000095. doi:10.1136/openhrt-2014-000095
  57. Avaldi VM, Lenzi J, Urbinati S, et al. Effect of cardiologist care on 6-month outcomes in patients discharged with heart failure: results from an observational study based on administrative data. BMJ Open 2017; 7(11):e018243. doi:10.1136/bmjopen-2017-018243
  58. Breathett K, Liu WG, Allen LA, et al. African Americans are less likely to receive care by a cardiologist during an intensive care unit admission for heart failure. JACC Heart Fail 2018; 6(5):413–420. doi:10.1016/j.jchf.2018.02.015
  59. Sridhar AR, Yarlagadda V, Parasa S, et al. Cardiac resynchronization therapy: US trends and disparities in utilization and outcomes. Circ Arrhythm Electrophysiol 2016; 9(3):e003108. doi:10.1161/CIRCEP.115.003108
  60. Abraham WT, Fisher WG, Smith AL, et al; MIRACLE Study Group. Multicenter insync randomized clinical evaluation. Cardiac resynchronization in chronic heart failure. N Engl J Med 2002; 346(24):1845–1853. doi:10.1056/NEJMoa013168
  61. Auricchio A, Stellbrink C, Sack S, et al; Pacing Therapies in Congestive Heart Failure (PATH-CHF) Study Group. Long-term clinical effect of hemodynamically optimized cardiac resynchronization therapy in patients with heart failure and ventricular conduction delay. J Am Coll Cardiol 2002; 39(12):2026–2033. pmid:12084604
  62. Cazeau S, Leclercq C, Lavergne T, et al; Multisite Stimulation in Cardiomyopathies (MUSTIC) Study Investigators. Effects of multisite biventricular pacing in patients with heart failure and intraventricular conduction delay. N Engl J Med 2001; 344(12):873–880. doi:10.1056/NEJM200103223441202
  63. Higgins SL, Hummel JD, Niazi IK, et al. Cardiac resynchronization therapy for the treatment of heart failure in patients with intraventricular conduction delay and malignant ventricular tachyarrhythmias. J Am Coll Cardiol 2003; 42(8):1454–1459. pmid:14563591
  64. Young JB, Abraham WT, Smith AL, et al; Multicenter InSync ICD Randomized Clinical Evaluation (MIRACLE ICD) Trial Investigators. Combined cardiac resynchronization and implantable cardioversion defibrillation in advanced chronic heart failure: the MIRACLE ICD Trial. JAMA 2003; 289(20):2685–2694. doi:10.1001/jama.289.20.2685
  65. Sutton MG, Plappert T, Hilpisch KE, Abraham WT, Hayes DL, Chinchoy E. Sustained reverse left ventricular structural remodeling with cardiac resynchronization at one year is a function of etiology: quantitative Doppler echocardiographic evidence from the Multicenter InSync Randomized Clinical Evaluation (MIRACLE). Circulation 2006; 113(2):266–272. doi:10.1161/CIRCULATIONAHA.104.520817
  66. Cleland JG, Daubert JC, Erdmann E, et al; Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The effect of cardiac resynchronization on morbidity and mortality in heart failure. N Engl J Med 2005; 352(15):1539–1549. doi:10.1056/NEJMoa050496
  67. Yancy CW, Jessup M, Bozkurt B, et al; American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation 2013; 128(16):e240–e327. doi:10.1161/CIR.0b013e31829e8776
  68. Farmer SA, Kirkpatrick JN, Heidenreich PA, Curtis JP, Wang Y, Groeneveld PW. Ethnic and racial disparities in cardiac resynchronization therapy. Heart Rhythm 2009; 6(3):325–331. doi:10.1016/j.hrthm.2008.12.018
  69. Rose EA, Gelijns AC, Moskowitz AJ, et al; Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH) Study Group. Long-term use of a left ventricular assist device for end-stage heart failure. N Engl J Med 2001; 345(20):1435–1443. doi:10.1056/NEJMoa012175
  70. Slaughter MS, Rogers JG, Milano CA, et al; HeartMate II Investigators. Advanced heart failure treated with continuous-flow left ventricular assist device. N Engl J Med 2009; 361(23):2241–2251. doi:10.1056/NEJMoa0909938
  71. Tsiouris A, Brewer RJ, Borgi J, Nemeh H, Paone G, Morgan JA. Continuous-flow left ventricular assist device implantation as a bridge to transplantation or destination therapy: racial disparities in outcomes. J Heart Lung Transplant 2013; 2(3):299–304. doi:10.1016/j.healun.2012.11.017
  72. Stulak JM, Deo S, Cowger J, et al. Do racial and sex disparities exist in clinical characteristics and outcomes for patients undergoing left ventricular assist device implantation? J Heart Lung Transplant 2013; 32(45):S279–S280.
  73. Joyce DL, Conte JV, Russell SD, Joyce LD, Chang DC. Disparities in access to left ventricular assist device therapy. J Surg Res 2009; 152(1):111–117. doi:10.1016/j.jss.2008.02.065
  74. Breathett K, Allen LA, Helmkamp L, et al. Temporal trends in contemporary use of ventricular assist devices by race and ethnicity. Circ Heart Fail 2018; 11(8):e005008. doi:10.1161/CIRCHEARTFAILURE.118.005008
  75. Kilic A, Higgins RS, Whitson BA, Kilic A. Racial disparities in outcomes of adult heart transplantation. Circulation 2015; 131(10):882–889. doi:10.1161/CIRCULATIONAHA.114.011676
  76. Liu V, Bhattacharya J, Weill D, Hlatky MA. Persistent racial disparities in survival after heart transplantation. Circulation 2011; 123(15):1642–1649. doi:10.1161/CIRCULATIONAHA.110.976811
  77. Mahle WT, Kanter KR, Vincent RN. Disparities in outcome for black patients after pediatric heart transplantation. J Pediatr 2005; 147(6):739–743. doi:10.1016/j.jpeds.2005.07.018
  78. Park MH, Tolman DE, Kimball PM. The impact of race and HLA matching on long-term survival following cardiac transplantation. Transplant Proc 1997; 29(1–2):1460–1463. pmid:9123381
  79. Higgins RS, Fishman JA. Disparities in solid organ transplantation for ethnic minorities: facts and solutions. Am J Transplant 2006; 6(11):2556–2562. doi:10.1111/j.1600-6143.2006.01514.x
  80. Taylor AL, Wright JT Jr. Should ethnicity serve as the basis for clinical trial design? Importance of race/ethnicity in clinical trials: lessons from the African-American Heart Failure Trial (A-HeFT), the African-American Study of Kidney Disease and Hypertension (AASK), and the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Circulation 2005; 112(23):3654–3660. doi:10.1161/CIRCULATIONAHA.105.540443
  81. Yancy CW. Heart failure in African Americans: a cardiovascular engima. J Card Fail 2000; 6(3):183–186. pmid:10997742
  82. Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report. JAMA 2003; 289(19):2560–2572. doi:10.1001/jama.289.19.2560
  83. Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration cooperative study. N Engl J Med 1986; 314(24):1547–1552. doi:10.1056/NEJM198606123142404
  84. Chen MS Jr, Lara PN, Dang JH, Paterniti DA, Kelly K. Twenty years post-NIH Revitalization Act: enhancing minority participation in clinical trials (EMPaCT): laying the groundwork for improving minority clinical trial accrual: renewing the case for enhancing minority participation in cancer clinical trials. Cancer 2014;120(suppl 7):1091–1096. doi:10.1002/cncr.28575
  85. Geller SE, Koch A, Pellettieri B, Carnes M. Inclusion, analysis, and reporting of sex and race/ethnicity in clinical trials: have we made progress? J Womens Health (Larchmt) 2011; 20(3):315–320. doi:10.1089/jwh.2010.2469
  86. Beta-Blocker Evaluation of Survival Trial Investigators; Eichhorn EJ, Domanski MJ, Krause-Steinrauf H, Bristow MR, Lavori PW. A trial of the beta-blocker bucindolol in patients with advanced chronic heart failure. N Engl J Med 2001; 344(22):1659–1667. doi:10.1056/NEJM200105313442202
  87. Taylor AL, Ziesche S, Yancy C, et al; African-American Heart Failure Trial Investigators. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med 2004; 351(20):2049–2057. doi:10.1056/NEJMoa042934
  88. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med 1999; 14(9):537–546. pmid:10491242
  89. Swanson GM, Ward AJ. Recruiting minorities into clinical trials: toward a participant-friendly system. J Natl Cancer Inst 1995; 87(23):1747–1759. doi:10.1093/jnci/87.23.1747
  90. Institute of Medicine (US) Committee on Understanding and Eliminating Racial and Ethnic Disparities in Health Care; Smedley BD, Stith AY, Nelson AR, eds. Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care. Washington, DC: National Academies Press (US); 2003. https://www.ncbi.nlm.nih.gov/books/NBK220358/. Accessed May 13, 2019.
  91. Fisher JA, Kalbaugh CA. Challenging assumptions about minority participation in US clinical research. Am J Public Health 2011; 101(12):2217–2222. doi:10.2105/AJPH.2011.300279
  92. Hall WJ, Chapman MV, Lee KM, et al. Implicit racial/ethnic bias among health care professionals and its influence on health care outcomes: a systematic review. Am J Public Health 2015; 105(12):e60–e76. doi:10.2105/AJPH.2015.302903
  93. Biernat M, Manis M. Shifting standards and stereotype-based judgments. J Pers Soc Psychol 1994; 66(1):5–20. pmid:8126651
  94. Chapman EN, Kaatz A, Carnes M. Physicians and implicit bias: how doctors may unwittingly perpetuate health care disparities. J Gen Intern Med 2013; 28(11):1504–1510. doi:10.1007/s11606-013-2441-1
  95. FitzGerald C, Hurst S. Implicit bias in healthcare professionals: a systematic review. BMC Med Ethics 2017; 18(1):19. doi:10.1186/s12910-017-0179-8
  96. van Ryn M, Burke J. The effect of patient race and socio-economic status on physicians’ perceptions of patients. Soc Sci Med 2000; 50(6):813–828. pmid:10695979
  97. Mayo RM, Sherrill WW, Sundareswaran P, Crew L. Attitudes and perceptions of Hispanic patients and health care providers in the treatment of Hispanic patients: a review of the literature. Hisp Health Care Int 2007; 5(2):64–72.
  98. Blair IV, Steiner JF, Havranek EP. Unconscious (implicit) bias and health disparities: where do we go from here? Perm J 2011; 15(2):71–78. pmid:21841929
  99. Green AR, Carney DR, Pallin DJ, et al. Implicit bias among physicians and its prediction of thrombolysis decisions for black and white patients. J Gen Intern Med 2007; 22(9):1231–1238. doi:10.1007/s11606-007-0258-5
  100. Sabin JA, Rivara FP, Greenwald AG. Physician implicit attitudes and stereotypes about race and quality of medical care. Med Care 2008; 46(7):678–685. doi:10.1097/MLR.0b013e3181653d58
  101. Smedley BD, Stith AY, Colburn L, et al; Institute of Medicine. The Right Thing to Do, The Smart Thing to Do: Enhancing Diversity in the Health Professions: Summary of the Symposium on Diversity in Health Professions in Honor of Herbert W. Nickens, MD. Washington, DC: National Academies Press; 2001. https://www.ncbi.nlm.nih.gov/books/NBK223633/. Accessed May 13, 2019.
  102. Chen J, Vargas-Bustamante A, Mortensen K, Ortega AN. Racial and ethnic disparities in health care access and utilization under the Affordable Care Act. Med Care 2016; 54(2):140–146. doi:10.1097/MLR.0000000000000467
  103. Saha S, Freeman M, Toure J, Tippens KM, Weeks C, Ibrahim S. Racial and ethnic disparities in the VA health care system: a systematic review. J Gen Intern Med 2008; 23(5):654–671. doi:10.1007/s11606-008-0521-4
  104. McCormick D, Sayah A, Lokko H, Woolhandler S, Nardin R. Access to care after Massachusetts’ health care reform: a safety net hospital patient survey. J Gen Intern Med 2012; 27(11):1548–1554. doi:10.1007/s11606-012-2173-7
  105. Burgos JL, Yee D, Csordas T, et al. Supporting the minority physician pipeline: providing global health experiences to undergraduate students in the United States-Mexico border region. Med Educ Online 2015; 20:27260. doi:10.3402/meo.v20.27260
  106. Traylor AH, Schmittdiel JA, Uratsu CS, Mangione CM, Subramanian U. The predictors of patient–physician race and ethnic concordance: a medical facility fixed-effects approach. Health Serv Res 2010; 45(3):792–805. doi:10.1111/j.1475-6773.2010.01086.x
  107. LaVeist TA, Nuru-Jeter A. Is doctor-patient race concordance associated with greater satisfaction with care? J Health Soc Behav 2002; 43(3):296–306. pmid:12467254
  108. Marrast LM, Zallman L, Woolhandler S, Bor DH, McCormick D. Minority physicians’ role in the care of underserved patients: diversifying the physician workforce may be key in addressing health disparities. JAMA Intern Med 2014; 174(2):289–291. doi:10.1001/jamainternmed.2013.12756
Article PDF
Author and Disclosure Information

Quentin R. Youmans, MD
Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Lindsey Hastings-Spaine, MD
Rutgers New Jersey Medical School, Department of Emergency Medicine, Newark, NJ

Oluseyi Princewill, MD, MPH
MedStar Health Cardiology Associates, Olney, MD

Titilayo Shobayo, BS
Morehouse School of Medicine, Atlanta, GA

Ike S. Okwuosa, MD
Assistant Professor of Medicine, Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Address: Ike S. Okwuosa, MD, Feinberg School of Medicine, Division of Cardiology, Northwestern University, 676 N St. Clair Street, Chicago, IL 60611; isokwuosa@gmail.com

Issue
Cleveland Clinic Journal of Medicine - 86(9)
Publications
Topics
Page Number
621-632
Legacy Keywords
disparities, cardiovascular care, heart care, racism, bias, race, African American, heart attack, stroke, hypertension, black, white, smoking, American Indian, Alaska Native, exercise, inactivity, sedentary lifestyle, nutrition, obesity, diabetes, coronary artery disease, heart failure, transplant, research study, minority physician, Tuskegee syphilis experiment, Quentin Youmans, Lindsey Hastings-Spaine, Oluseyi Princewill, Titilayo Shobayo, Ike Okwuosa
Sections
Author and Disclosure Information

Quentin R. Youmans, MD
Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Lindsey Hastings-Spaine, MD
Rutgers New Jersey Medical School, Department of Emergency Medicine, Newark, NJ

Oluseyi Princewill, MD, MPH
MedStar Health Cardiology Associates, Olney, MD

Titilayo Shobayo, BS
Morehouse School of Medicine, Atlanta, GA

Ike S. Okwuosa, MD
Assistant Professor of Medicine, Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Address: Ike S. Okwuosa, MD, Feinberg School of Medicine, Division of Cardiology, Northwestern University, 676 N St. Clair Street, Chicago, IL 60611; isokwuosa@gmail.com

Author and Disclosure Information

Quentin R. Youmans, MD
Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Lindsey Hastings-Spaine, MD
Rutgers New Jersey Medical School, Department of Emergency Medicine, Newark, NJ

Oluseyi Princewill, MD, MPH
MedStar Health Cardiology Associates, Olney, MD

Titilayo Shobayo, BS
Morehouse School of Medicine, Atlanta, GA

Ike S. Okwuosa, MD
Assistant Professor of Medicine, Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, IL

Address: Ike S. Okwuosa, MD, Feinberg School of Medicine, Division of Cardiology, Northwestern University, 676 N St. Clair Street, Chicago, IL 60611; isokwuosa@gmail.com

Article PDF
Article PDF
Related Articles

Cardiovascular disease became the leading cause of death in the United States in the early 20th century, and it accounts for nearly half of all deaths in industrialized nations.1 The mortality it inflicts was thought to be shared equally between both sexes and among all age groups and races.2 The cardiology community implemented innovative epidemiologic research, through which risk factors for cardiovascular disease were established.1 The development of coronary care units reduced in-hospital mortality from acute myocardial infarction from 30% to 15%.2–5 Further advances in pharmacology, revascularization, and imaging have aided in the detection and treatment of cardiovascular disease.6 Though cardiovascular disease remains the number-one cause of death worldwide, rates are on the decline.7

For several decades, health disparities have been recognized as a source of pathology in cardiovascular medicine, resulting in inequity of care administration among select populations. In this review, we examine whether the same forward thinking that has resulted in a decline in cardiovascular disease has had an impact on the pervasive disparities in cardiovascular medicine.

DISPARITIES DEFINED

Compared with whites, members of minority groups have a higher burden of chronic diseases, receive lower quality care, and have less access to medical care. Recognizing the potential public health ramifications, in 1999 the US Congress tasked the Institute of Medicine to study and assess the extent of healthcare disparities. This led to the landmark publication, Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care.8

The Institute of Medicine defines disparities in healthcare as racial or ethnic differences in the quality of healthcare that are not due to access-related factors, clinical needs, preferences, and appropriateness of intervention.8 Disparities can also exist according to socioeconomic status and sex.9

In an early study documenting the concept of disparities in cardiovascular disease, Stone and Vanzant10 concluded that heart disease was more common in African Americans than in whites, and that hypertension was the principal cause of cardiovascular disease mortality in African Americans.

youmans_disparitiesincardiovascularcare_f1.jpg
%3Cp%3EFigure%201.%20Avoidable%20deaths%20from%20heart%20disease%2C%20stroke%2C%20and%20hypertensive%20disease%2C%202001%20and%202010.%3C%2Fp%3E

Although avoidable deaths from heart disease, stroke, and hypertensive disease declined between 2001 and 2010, African Americans still have a higher mortality rate than other racial and ethnic groups (Figure 1).11

DISPARITIES AND CARDIOVASCULAR HEALTH

The concept of cardiovascular health was established by the American Heart Association (AHA) in efforts to achieve an additional 20% reduction in cardiovascular disease-related mortality by 2020.7 Cardiovascular health is defined as the absence of clinically manifest cardiovascular disease and is measured by 7 components:

  • Not smoking or abstaining from smoking for at least 1 year
  • A normal body weight, defined as a body mass index less than 25 kg/m2
  • Optimal physical activity, defined as 75 minutes of vigorous physical activity or 150 minutes of moderate-intensity physical activity per week
  • Regular consumption of a healthy diet
  • Total cholesterol below 200 mg/dL
  • Blood pressure less than 120/80 mm Hg
  • Fasting blood sugar below 100 mg/dL.

Nearly 70% of the US population can claim 2, 3, or 4 of these components, but differences exist according to race,12 and 60% of adult white Americans are limited to achieving no more than 3 of these healthy metrics, compared with 70% of adult African Americans and Hispanic Americans.

Smoking

Smoking is a major risk factor for cardiovascular disease.12–14

youmans_disparitiesincardiovascularcare_f2.jpg
%3Cp%3EFigure%202.%20Percentage%20of%20adults%20who%20are%20active%20smokers%2C%202005%20and%202014.%3C%2Fp%3E

During adolescence, white males are more likely to smoke than African American and Hispanic males,12 but this trend reverses in adulthood, when African American men have a higher prevalence of smoking than white men (21.4% vs 19%).7 Rates of lifetime use are highest among American Indian or Alaskan natives and whites (75.9%), followed by African Americans (58.4%), native Hawaiians (56.8%), and Hispanics (56.7%).15 Trends for current smoking are similar (Figure 2).16 Moreover, households with lower socioeconomic status have a higher prevalence of smoking.7

Physical activity

People with a sedentary lifestyle are more likely to die of cardiovascular disease. As many as 250,000 deaths annually in the United States are attributed to lack of regular physical activity.17

Recognizing the potential public health ramifications, the AHA and the 2018 Federal Guidelines on Physical Activity recommend that children engage in 60 minutes of daily physical activity and that adults participate in 150 minutes of moderate-intensity or 75 minutes of vigorous physical activity weekly.18,19

youmans_disparitiesincardiovascularcare_f3.jpg
%3Cp%3EFigure%203.%20Prevalence%20of%20inactivity%3Csup%3Ea%3C%2Fsup%3E%20in%20the%20United%20States%2C%202013.%3C%2Fp%3E%3Chr%3E%3Cp%3E%3Csup%3Ea%3C%2Fsup%3EPercentage%20of%20US%20adults%20eligible%20for%20intensive%20behavioral%20counseling%20for%20cardiovascular%20disease%20prevention%20and%20not%20meeting%20aerobic%20exercise%20guideline%3C%2Fp%3E

In the United States, 15.2% of adolescents reported being physically inactive, according to data published in 2016.7 Similar to most cardiovascular risk factors, minority populations and those of lower socioeconomic status had the worst profiles. The prevalence of physical inactivity was highest in African Americans and Hispanics (Figure 3).20

Studies have shown an association between screen-based sedentary behavior (computers, television, and video games) and cardiovascular disease.21–23 In the United States, 41% of adolescents used computers for activities other than homework for more than 3 hours per day on a school day.7 The pattern of use was highest in African American boys and African American girls, followed by Hispanic girls and Hispanic boys.18 Trends were similar with regard to watching television for more than 3 hours per day.

Sedentary behavior persists into adulthood, with rates of inactivity of 38.3% in African Americans, 40.1% in Hispanics, and 26.3% in white adults.7

 

 

Nutrition and obesity

Nutrition plays a major role in cardiovascular disease, specifically in the pathogenesis of atherosclerotic disease and hypertension.24 Most Americans do not meet dietary recommendations, with minority communities performing worse in specific metrics.7

Dietary patterns are reflected in the rate of obesity in this nation. Studies have shown a direct correlation between obesity and cardiovascular disease such as coronary artery disease, heart failure, and atrial fibrillation.25–28 According to data from the National Health and Nutrition Examination Survey (NHANES), 31% of children between the ages of 2 and 19 years are classified as obese or overweight. The highest rates of obesity are seen in Hispanic and African American boys and girls. The obesity epidemic is disproportionately rampant in children living in households with low income, low education, and high unemployment rates.7,29–31

Despite the risks associated with obesity, only 64.8% of obese adults report being informed by a doctor or health professional that they were overweight. The proportion of obese adults informed that they were overweight was significantly lower for African Americans and Hispanics compared with whites. Similar differences are seen based on socioeconomic status, as middle-income patients were less likely to be informed than those in the higher income strata (62.4% vs 70.6%).7,31

Blood pressure

Hypertension is a well-established risk factor for cardiovascular disease and stroke, and a blood pressure of 120/80 mm Hg or lower is identified as a component of ideal cardiovascular health.

In the United States the prevalence of hypertension in adults older than 20 is 32%.7 The prevalence of hypertension in African Americans is among the highest in the world.32,33 African Americans develop high blood pressure at earlier ages, and their average resting blood pressures are higher than in whites.34,35 For a 45-year-old without hypertension, the 40-year risk of developing hypertension is 92.7% for African Americans and 86% for whites.35 Hypertension is a major risk factor for stroke, and African Americans have a 1.8 times greater rate of fatal stroke than whites.7

In 2013 there were 71,942 deaths attributable to high blood pressure, and the 2011 death rate associated with hypertension was 18.9 per 100,000. By race, the death rate was 17.6 per 100,000 for white males and an alarming 47.1 per 100,000 for African American males; rates were 15.2 per 100,000 for white females and 35.1 per 100,000 for African American females.7

It is unclear what accounts for the racial difference in prevalence in hypertension. Studies have shown that African Americans are more likely than whites to have been told on more than 2 occasions that they have hypertension. And 85.7% of African Americans are aware that they have high blood pressure, compared with 82.7% of whites.14

African Americans and Hispanics have poorer hypertension control compared with whites.36,37 These observed differences cannot be attributed to access alone, as African Americans were more likely to be on higher-intensity blood pressure therapy, whereas Hispanics were more likely to be undertreated.36,38 In a meta-analysis of 13 trials, Peck et al39 showed that African Americans showed a lesser reduction in systolic and diastolic blood pressure when treated with angiotensin-converting enzyme (ACE) inhibitors.

The 2017 American College of Cardiology (ACC) and AHA guidelines for the prevention, detection, evaluation, and management of high blood pressure in adults40 identifies 4 drug classes as reducing cardiovascular disease morbidity and mortality: thiazide diuretics, ACE inhibitors, angiotensin II receptor blockers (ARBs), and calcium channel blockers. Of these 4 classes, thiazide diuretics and calcium channel blockers have been shown to lower blood pressure more effectively in African Americans than renin-angiotensin-aldosterone inhibition with ACE inhibitors or ARBs.

Glycemic control

Type 2 diabetes mellitus secondary to insulin resistance disproportionately affects minority groups, as the prevalence of diabetes mellitus in African Americans is almost twice as high as that in whites, and 35% higher in Hispanics compared with whites.7,41 Based on NHANES data between 1984 and 2004, the prevalence of diabetes mellitus is expected to increase by 99% in whites, 107% in African Americans, and 127% in Hispanics by 2050. Alarmingly, African Americans over age 75 are expected to experience a 606% increase by 2050.42

With regard to mortality, 21.7 deaths per 100,000 population were attributable to diabetes mellitus according to reports by the AHA in 2016. The death rate in white males was 24.3 per 100,000 compared with 44.9 per 100,000 for African Americans males. The associated mortality rate for white women was 16.2 per 100,000, and 35.8 per 100,000 for African American females.7

 

 

DISPARITIES AND CORONARY ARTERY DISEASE CARE

The management of coronary artery disease has evolved from prolonged bed rest to surgical, pharmacologic, and percutaneous revascularization.2,5 Coronary revascularization procedures are now relatively common: 950,000 percutaneous coronary interventions and 397,000 coronary artery bypass procedures were performed in 2010.7

Nevertheless, despite similar clinical presentations, African Americans with acute myocardial infarction were less likely to be referred for coronary artery bypass grafting than whites.43–46 They were also less likely to be given thrombolytics47 and less likely to undergo coronary angiography with percutaneous coronary intervention.48 Similar differences have been reported when comparing Hispanics with whites.49

Some suggest that healthcare access is a key mediator of health disparities.50 In 2009, Hispanics and African Americans accounted for more than 50% of those without health insurance.51 Improved access to healthcare might mitigate the disparity in revascularizations.

Massachusetts was one of the first states to mandate that all residents obtain health insurance. As a result, the uninsured rates declined in African Americans and Hispanics in Massachusetts, but a disparity in revascularization persisted. African Americans and Hispanics were 27% and 16% less likely to undergo revascularization procedures (coronary artery bypass grafting or percutaneous coronary intervention) than whites,51 suggesting that disparities in revascularization are not solely secondary to healthcare access.

These findings are consistent with a 2004 Veterans Administration study,52 in which healthcare access was equal among races. The study showed that African Americans received fewer cardiac procedures after an acute myocardial infarction compared with whites.

Have we made progress? The largest disparity between African Americans and whites in coronary artery disease mortality existed in 1990. The disparity persisted to 2012, and although decreased, it is projected to persist to 2030.53

DISPARITIES IN HEART FAILURE

An estimated 5.7 million Americans have heart failure, and 915,000 new cases are diagnosed annually.7 Unlike coronary artery disease, heart failure is expected to increase in prevalence by 46%, to 8 million Americans with heart failure by 2030.7,54

Our knowledge of disparities in the area of heart failure is derived primarily from epidemiologic studies. The Multi-Ethnic Study of Atherosclerosis55 showed that African Americans (4.6 per 1,000), followed by Hispanics (3.5 per 1,000) had a higher risk of developing heart failure compared with whites (2.4 per 1,000).The higher risk is in part due to disparities in socioeconomic status and prevalence of hypertension, as African Americans accounted for 75% of cases of nonischemic-related heart failure.55 African Americans also have a higher 5-year mortality rate than whites.55

Even though the 5-year mortality rate in heart failure is still 50%, the past 30 years have seen innovations in pharmacologic and device therapy and thus improved outcomes in heart failure patients. Still, significant gaps in the use of guideline-recommended therapies, quality of care, and clinical outcomes persist in contemporary practice for racial minorities with heart failure.

Disparities in inpatient care for heart failure

Patients admitted for heart failure and cared for by a cardiologist are more likely to be discharged on guideline-directed medical therapy, have fewer heart failure readmissions, and lower mortality.56,57 Breathett et al,58 in a study of 104,835 patients hospitalized in an intensive care unit for heart failure, found that primary intensive care by a cardiologist was associated with higher survival in both races. However, in the same study, white patients had a higher odds of receiving care from a cardiologist than African American patients.

Disparities and cardiac resynchronization therapy devices

In one-third of patients with heart failure, conduction delays result in dyssynchronous left ventricular contraction.59 Dyssynchrony leads to reduced cardiac performance, left ventricular remodeling, and increased mortality.56

Cardiac resynchronization therapy (CRT) was approved for clinical use in 2001, and studies have shown that it improves quality of life, exercise tolerance, cardiac performance, and morbidity and mortality rates.59–66 The 2013 ACC/AHA guidelines for the management of heart failure give a class IA recommendation (the highest) for its use in patients with a left ventricular ejection fraction of 35% or less, sinus rhythm, left bundle branch block and a QRS duration of 150 ms or greater, and New York Heart Association class II, III, or ambulatory IV symptoms while on guideline-directed medical therapy.67

Despite these recommendations, racial differences are observed. A study using the Nationwide Inpatient Sample database59 showed that between 2002 and 2010, a total of 374,202 CRT devices were implanted, averaging 41,578 annually. After adjusting for heart failure admissions, the study showed that CRT use was favored in men and in whites.

Another study, using the National Cardiovascular Data Registry,68 looked at patients who received implantable cardiac defibrillators (ICDs) and were eligible to receive CRT. It found that African Americans and Hispanics were less likely than whites to receive CRT, even though they were more likely to meet established criteria.

Disparities and left ventricular assist devices

The Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart failure (REMATCH) trial and Heart Mate II trial demonstrated that left ventricular assist devices (LVADs) were durable options for long-term support for patients with end-stage heart failure.69,70 Studies that examined the role of race and clinical outcomes after LVAD implantation have reported mixed findings.71,72 Few studies have looked at the role racial differences play in accessing LVAD therapy.

Joyce et al73 reviewed data from the Nationwide Inpatient Sample from 2002 to 2003 on patients admitted to the hospital with a primary diagnosis of heart failure or cardiogenic shock. A total of 297,866 patients were included in the study, of whom only 291 underwent LVAD implantation. A multivariate analysis found that factors such as age over 65, female sex, admission to a nonacademic center, geographic region, and African American race adversely influenced access to LVAD therapy.

Breathett et al74 evaluated racial differences in LVAD implantations from 2012 to 2015, a period that corresponds to increased health insurance expansion, and found LVAD implantations increased among African American patients with advanced heart failure, but no other racial or ethnic group.

 

 

Disparities and heart transplant

For patients with end-stage heart failure, orthotopic heart transplant is the most definitive and durable option for long-term survival. According to data from the United Network for Organ Sharing, 62,508 heart transplants were performed from January 1, 1988 to December 31, 2015. Compared with transplants of other solid organs, heart transplant occurs in significantly infrequent rates.

Barriers to transplant include lack of health insurance, considered a surrogate for low socioeconomic status. Hispanics and African Americans are less likely to have private health insurance than non-Hispanic whites, and this difference is magnified among the working poor.

Despite these perceived barriers, Kilic et al75 found that African Americans comprised 16.4% of heart transplant recipients, although they make up only approximately 13% of the US population. They also had significantly shorter wait-list times than whites. On the negative side, African Americans had a higher unadjusted mortality rate than whites (15% vs 12% P = .002). African Americans also tended to receive their transplants at centers with lower transplant volumes and higher transplant mortality rates.

Several other studies also showed that African Americans compared to whites have significantly worse outcomes after transplant.76–79 What accounts for this difference? Kilic et al75 showed that African Americans had the lowest proportion of blood type matching and lowest human leukocyte antigen matching, were younger (because African Americans develop more advanced heart failure at younger ages), had higher serum creatinine levels, and were more often bridged to transplant with an LVAD.

DISPARITIES IN CARDIOVASCULAR RESEARCH

Although the United States has the most sophisticated and robust medical system in the world, select groups have significant differences in delivery and healthcare outcomes. There are many explanations for these differences, but a contributing factor may be the paucity of research dedicated to understand racial and ethnic differences.80

Differences observed in epidemiologic studies may be secondary to pathophysiology, genetic differences, environment, and lifestyle choices. Historically, clinical trials were conducted in homogeneous populations with respect to age (middle-aged), sex (male), and race (white), and the results were generalized to heterogeneous populations.80

Disparities in research have implications in clinical practice. Overall, the primary cause of heart failure is ischemia; however, in African Americans, the primary cause is hypertensive heart disease.81 Studies in hypertension have shown that African Americans have less of a response to neurohormonal blockade with ACE inhibitors and beta-blockers than non-African Americans.82 Nevertheless, neurohormonal blockade has become the cornerstone of heart failure treatment.

Retrospective analysis of the Vasodilator-Heart Failure trials83 showed that treatment with isosorbide dinitrate plus hydralazine, compared with placebo, conferred a survival benefit for African Americans but not whites.80 No survival advantage was noted when isosorbide dinitrate/hydralazine was compared to enalapril in African Americans, although enalapril was superior to isosorbide dinitrate in whites.45 These observations were recognized 10 to 15 years after trial completion, and were only possible because the trials included sufficient numbers of African American patients to complete analysis.

In 1993, the US Congress passed the National Institutes of Health (NIH) Revitalization Act, which established guidelines requiring NIH grant applicants to include minorities in human subject research, as they were historically underrepresented in clinical research trials.84,85

In 2001, the Beta-Blocker Evaluation of Survival Trial86 reported its results investigating whether bucindolol, a nonselective beta-blocker, would reduce mortality in patients with advanced heart failure (New York Heart Association class III or IV). This was one of the first trials to prospectively investigate racial and ethnic differences in response to treatment. Though it showed no overall benefit in the use of bucindolol in the treatment of advanced heart failure, subgroup analysis showed that whites did enjoy a benefit in terms of lower mortality, whereas African Americans did not.

Results of the Vasodilator-Heart Failure trials led to further population-directed research, most notably the African American Heart Failure Trial,87 a double-blind, placebo-controlled, randomized trial in patients who identified as African American. Patients who were randomized to receive a fixed dose of hydralazine and isosorbide dinitrate had a 43% lower mortality rate, a 33% lower hospitalization rate for heart failure, and better quality of life than patients in the placebo group, leading to early termination of the trial. The outcomes suggested that the combination of isosorbide dinitrate and hydralazine treats heart failure in a manner independent of pure neurohormonal blockade.

CHALLENGES IN STUDY PARTICIPATION

Recruitment of minority participants in biomedical research is a challenging task for clinical investigators.88,89 Some of the factors thought to pose potential barriers for racial and ethnic minority participation in health research include poor access to primary medical care, failure of researchers to recruit minority populations actively, and language and cultural barriers.90

Further, it is widely claimed that African Americans are less willing than nonminority individuals to participate in clinical research trials due to general distrust of the medical community as a result of the Tuskegee Syphilis Experiment.91 That infamous study, conducted by the US Public Health Service between 1932 and 1972, sought to record the natural progression of untreated syphilis in poor African American men in Alabama. The participants were not informed of the true purpose of the study, and they were under the impression that they were simply receiving free healthcare from the US government. Further, they were denied appropriate treatment even after it became readily available, in order for researchers to observe the progression of the disease.

While the 1993 mandate did in fact increase pressure on researchers to develop strategies to overcome participation barriers, the issue of underrepresentation of racial minorities in clinical research, including cardiovascular research, has not been resolved and continues to be a problem today.

The overall goal of clinical research is to determine the best strategies to prevent and treat disease. But if the study population is not representative of the affected population at large, the results cannot be generalized to underrepresented subgroups. The implications of underrepresentation in research are far-reaching, and can further contribute to disparate care of minority patients such as African Americans, who have a higher prevalence of cardiovascular risk factors and greater burden of heart failure.

 

 

PROPOSING SOLUTIONS

Between 1986 and 2018, according to a PUBMED search, 10,462 articles highlighted the presence of a health-related disparity. Solutions to address and ultimately eradicate disparities will need to eliminate healthcare bias, increase patient access, and increase diversity and inclusion in the physician work force.

Eliminating bias

Implicit bias refers to attitudes, thoughts, and feelings that exist outside of the conscious awareness.92 These biases can be triggered by race, gender, or socioeconomic status. They have manifested in society as stereotypes that men are more competent than women, women are more verbal than men, and African Americans are more athletic than whites.93

The concept of implicit bias is important, in that the populations that experience the greatest health disparities also suffer from negative cultural stereotypes.94 Healthcare professionals are not inoculated against implicit bias.95 Studies have shown that most healthcare providers have implicit biases that reflect positive attitudes toward whites and negative attitudes toward people of color.92,94,96–98

The Implicit Association Test, introduced in 1998, is widely used to measure implicit bias. It measures response time of subjects to match particular social groups to particular attributes.99 Green et al,99 using this test, showed that although physicians reported no explicit preference for white vs African American patients or differences in perceived cooperativeness, the test revealed implicit preference favoring white Americans and implicit stereotypes of African Americans as less cooperative for medical procedures and in general. This also manifested in clinical decision-making, as white Americans were more likely, and African Americans less likely, to be treated with thrombolysis.99

Sabin et al100 showed that implicit bias was present among pediatricians, although less than in society as a whole and in other healthcare professionals.

But how does one change feelings that exist outside of the conscious awareness? Green et al99 showed that making physicians aware of their susceptibility to bias changed their behavior. A subset of physicians who were made aware that bias was a focus of the study were more likely to refer African Americans for thrombolysis even if they had a high degree of implicit pro-white bias.94,100 Perhaps mandating that all healthcare providers take a self-administered and confidentially reported Implicit Association Test will lead to awareness of implicit bias and minimize healthcare behaviors that contribute to the current state of disparities.

Improving access

Common indicators of access to healthcare include health insurance status, having a usual source of healthcare, and having a regular physician.101 Health insurance does offer protection from the costs associated with illness and health maintenance.101 It is also a major contributing factor in racial and ethnic disparities.

Chen et al102 examined the effects of the Affordable Care Act and found that it was associated with reduction in the probability of being uninsured, delaying necessary care, and forgoing necessary care, and increased probability of having a physician. However, earlier studies showed that access to health insurance by itself does not equate to equitable care.103,104

Diversifying the work force

African Americans comprise 4% of physicians and Hispanic Americans 5%, despite accounting for 13% and 16% of the US population.105 This underrepresentation has led to African American and Hispanic American patients being more likely than white patients to be treated by a physician from a dissimilar racial or ethnic background.106 Studies have shown that minority patients in a race- or ethnic-concordant relationship are more likely to use needed health services, less likely to postpone seeking care, and report greater satisfaction.106,107 Minority physicians often locate and practice in neighborhoods with high minority populations, and they disproportionately care for disadvantaged patients of lower socioeconomic status and poorer health.106,108

WE ARE STILL IN THE TUNNEL, BUT THERE IS LIGHT AT THE END

The cardiovascular community has faced tremendous challenges in the past and responded with innovative research that has led to imaging that aids in the diagnosis of subclinical cardiovascular disease and invasive and pharmacologic strategies that have improved cardiovascular outcomes. One may say that there is light at the end of the tunnel; however, the existence of disparate care reminds us that we are still in the tunnel.

Disparities in cardiovascular disease management present a unique challenge for the community. There is no drug, device, or invasive procedure to eliminate this pathology. However, by acknowledging the problem and implementing changes at the system, provider, and patient level, the cardiovascular community can achieve yet another momentous achievement: the end of cardiovascular health disparities. Cardiovascular disease makes no distinction in race, sex, age, or socioeconomic status, and neither should the medical community.

Cardiovascular disease became the leading cause of death in the United States in the early 20th century, and it accounts for nearly half of all deaths in industrialized nations.1 The mortality it inflicts was thought to be shared equally between both sexes and among all age groups and races.2 The cardiology community implemented innovative epidemiologic research, through which risk factors for cardiovascular disease were established.1 The development of coronary care units reduced in-hospital mortality from acute myocardial infarction from 30% to 15%.2–5 Further advances in pharmacology, revascularization, and imaging have aided in the detection and treatment of cardiovascular disease.6 Though cardiovascular disease remains the number-one cause of death worldwide, rates are on the decline.7

For several decades, health disparities have been recognized as a source of pathology in cardiovascular medicine, resulting in inequity of care administration among select populations. In this review, we examine whether the same forward thinking that has resulted in a decline in cardiovascular disease has had an impact on the pervasive disparities in cardiovascular medicine.

DISPARITIES DEFINED

Compared with whites, members of minority groups have a higher burden of chronic diseases, receive lower quality care, and have less access to medical care. Recognizing the potential public health ramifications, in 1999 the US Congress tasked the Institute of Medicine to study and assess the extent of healthcare disparities. This led to the landmark publication, Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care.8

The Institute of Medicine defines disparities in healthcare as racial or ethnic differences in the quality of healthcare that are not due to access-related factors, clinical needs, preferences, and appropriateness of intervention.8 Disparities can also exist according to socioeconomic status and sex.9

In an early study documenting the concept of disparities in cardiovascular disease, Stone and Vanzant10 concluded that heart disease was more common in African Americans than in whites, and that hypertension was the principal cause of cardiovascular disease mortality in African Americans.

youmans_disparitiesincardiovascularcare_f1.jpg
%3Cp%3EFigure%201.%20Avoidable%20deaths%20from%20heart%20disease%2C%20stroke%2C%20and%20hypertensive%20disease%2C%202001%20and%202010.%3C%2Fp%3E

Although avoidable deaths from heart disease, stroke, and hypertensive disease declined between 2001 and 2010, African Americans still have a higher mortality rate than other racial and ethnic groups (Figure 1).11

DISPARITIES AND CARDIOVASCULAR HEALTH

The concept of cardiovascular health was established by the American Heart Association (AHA) in efforts to achieve an additional 20% reduction in cardiovascular disease-related mortality by 2020.7 Cardiovascular health is defined as the absence of clinically manifest cardiovascular disease and is measured by 7 components:

  • Not smoking or abstaining from smoking for at least 1 year
  • A normal body weight, defined as a body mass index less than 25 kg/m2
  • Optimal physical activity, defined as 75 minutes of vigorous physical activity or 150 minutes of moderate-intensity physical activity per week
  • Regular consumption of a healthy diet
  • Total cholesterol below 200 mg/dL
  • Blood pressure less than 120/80 mm Hg
  • Fasting blood sugar below 100 mg/dL.

Nearly 70% of the US population can claim 2, 3, or 4 of these components, but differences exist according to race,12 and 60% of adult white Americans are limited to achieving no more than 3 of these healthy metrics, compared with 70% of adult African Americans and Hispanic Americans.

Smoking

Smoking is a major risk factor for cardiovascular disease.12–14

youmans_disparitiesincardiovascularcare_f2.jpg
%3Cp%3EFigure%202.%20Percentage%20of%20adults%20who%20are%20active%20smokers%2C%202005%20and%202014.%3C%2Fp%3E

During adolescence, white males are more likely to smoke than African American and Hispanic males,12 but this trend reverses in adulthood, when African American men have a higher prevalence of smoking than white men (21.4% vs 19%).7 Rates of lifetime use are highest among American Indian or Alaskan natives and whites (75.9%), followed by African Americans (58.4%), native Hawaiians (56.8%), and Hispanics (56.7%).15 Trends for current smoking are similar (Figure 2).16 Moreover, households with lower socioeconomic status have a higher prevalence of smoking.7

Physical activity

People with a sedentary lifestyle are more likely to die of cardiovascular disease. As many as 250,000 deaths annually in the United States are attributed to lack of regular physical activity.17

Recognizing the potential public health ramifications, the AHA and the 2018 Federal Guidelines on Physical Activity recommend that children engage in 60 minutes of daily physical activity and that adults participate in 150 minutes of moderate-intensity or 75 minutes of vigorous physical activity weekly.18,19

youmans_disparitiesincardiovascularcare_f3.jpg
%3Cp%3EFigure%203.%20Prevalence%20of%20inactivity%3Csup%3Ea%3C%2Fsup%3E%20in%20the%20United%20States%2C%202013.%3C%2Fp%3E%3Chr%3E%3Cp%3E%3Csup%3Ea%3C%2Fsup%3EPercentage%20of%20US%20adults%20eligible%20for%20intensive%20behavioral%20counseling%20for%20cardiovascular%20disease%20prevention%20and%20not%20meeting%20aerobic%20exercise%20guideline%3C%2Fp%3E

In the United States, 15.2% of adolescents reported being physically inactive, according to data published in 2016.7 Similar to most cardiovascular risk factors, minority populations and those of lower socioeconomic status had the worst profiles. The prevalence of physical inactivity was highest in African Americans and Hispanics (Figure 3).20

Studies have shown an association between screen-based sedentary behavior (computers, television, and video games) and cardiovascular disease.21–23 In the United States, 41% of adolescents used computers for activities other than homework for more than 3 hours per day on a school day.7 The pattern of use was highest in African American boys and African American girls, followed by Hispanic girls and Hispanic boys.18 Trends were similar with regard to watching television for more than 3 hours per day.

Sedentary behavior persists into adulthood, with rates of inactivity of 38.3% in African Americans, 40.1% in Hispanics, and 26.3% in white adults.7

 

 

Nutrition and obesity

Nutrition plays a major role in cardiovascular disease, specifically in the pathogenesis of atherosclerotic disease and hypertension.24 Most Americans do not meet dietary recommendations, with minority communities performing worse in specific metrics.7

Dietary patterns are reflected in the rate of obesity in this nation. Studies have shown a direct correlation between obesity and cardiovascular disease such as coronary artery disease, heart failure, and atrial fibrillation.25–28 According to data from the National Health and Nutrition Examination Survey (NHANES), 31% of children between the ages of 2 and 19 years are classified as obese or overweight. The highest rates of obesity are seen in Hispanic and African American boys and girls. The obesity epidemic is disproportionately rampant in children living in households with low income, low education, and high unemployment rates.7,29–31

Despite the risks associated with obesity, only 64.8% of obese adults report being informed by a doctor or health professional that they were overweight. The proportion of obese adults informed that they were overweight was significantly lower for African Americans and Hispanics compared with whites. Similar differences are seen based on socioeconomic status, as middle-income patients were less likely to be informed than those in the higher income strata (62.4% vs 70.6%).7,31

Blood pressure

Hypertension is a well-established risk factor for cardiovascular disease and stroke, and a blood pressure of 120/80 mm Hg or lower is identified as a component of ideal cardiovascular health.

In the United States the prevalence of hypertension in adults older than 20 is 32%.7 The prevalence of hypertension in African Americans is among the highest in the world.32,33 African Americans develop high blood pressure at earlier ages, and their average resting blood pressures are higher than in whites.34,35 For a 45-year-old without hypertension, the 40-year risk of developing hypertension is 92.7% for African Americans and 86% for whites.35 Hypertension is a major risk factor for stroke, and African Americans have a 1.8 times greater rate of fatal stroke than whites.7

In 2013 there were 71,942 deaths attributable to high blood pressure, and the 2011 death rate associated with hypertension was 18.9 per 100,000. By race, the death rate was 17.6 per 100,000 for white males and an alarming 47.1 per 100,000 for African American males; rates were 15.2 per 100,000 for white females and 35.1 per 100,000 for African American females.7

It is unclear what accounts for the racial difference in prevalence in hypertension. Studies have shown that African Americans are more likely than whites to have been told on more than 2 occasions that they have hypertension. And 85.7% of African Americans are aware that they have high blood pressure, compared with 82.7% of whites.14

African Americans and Hispanics have poorer hypertension control compared with whites.36,37 These observed differences cannot be attributed to access alone, as African Americans were more likely to be on higher-intensity blood pressure therapy, whereas Hispanics were more likely to be undertreated.36,38 In a meta-analysis of 13 trials, Peck et al39 showed that African Americans showed a lesser reduction in systolic and diastolic blood pressure when treated with angiotensin-converting enzyme (ACE) inhibitors.

The 2017 American College of Cardiology (ACC) and AHA guidelines for the prevention, detection, evaluation, and management of high blood pressure in adults40 identifies 4 drug classes as reducing cardiovascular disease morbidity and mortality: thiazide diuretics, ACE inhibitors, angiotensin II receptor blockers (ARBs), and calcium channel blockers. Of these 4 classes, thiazide diuretics and calcium channel blockers have been shown to lower blood pressure more effectively in African Americans than renin-angiotensin-aldosterone inhibition with ACE inhibitors or ARBs.

Glycemic control

Type 2 diabetes mellitus secondary to insulin resistance disproportionately affects minority groups, as the prevalence of diabetes mellitus in African Americans is almost twice as high as that in whites, and 35% higher in Hispanics compared with whites.7,41 Based on NHANES data between 1984 and 2004, the prevalence of diabetes mellitus is expected to increase by 99% in whites, 107% in African Americans, and 127% in Hispanics by 2050. Alarmingly, African Americans over age 75 are expected to experience a 606% increase by 2050.42

With regard to mortality, 21.7 deaths per 100,000 population were attributable to diabetes mellitus according to reports by the AHA in 2016. The death rate in white males was 24.3 per 100,000 compared with 44.9 per 100,000 for African Americans males. The associated mortality rate for white women was 16.2 per 100,000, and 35.8 per 100,000 for African American females.7

 

 

DISPARITIES AND CORONARY ARTERY DISEASE CARE

The management of coronary artery disease has evolved from prolonged bed rest to surgical, pharmacologic, and percutaneous revascularization.2,5 Coronary revascularization procedures are now relatively common: 950,000 percutaneous coronary interventions and 397,000 coronary artery bypass procedures were performed in 2010.7

Nevertheless, despite similar clinical presentations, African Americans with acute myocardial infarction were less likely to be referred for coronary artery bypass grafting than whites.43–46 They were also less likely to be given thrombolytics47 and less likely to undergo coronary angiography with percutaneous coronary intervention.48 Similar differences have been reported when comparing Hispanics with whites.49

Some suggest that healthcare access is a key mediator of health disparities.50 In 2009, Hispanics and African Americans accounted for more than 50% of those without health insurance.51 Improved access to healthcare might mitigate the disparity in revascularizations.

Massachusetts was one of the first states to mandate that all residents obtain health insurance. As a result, the uninsured rates declined in African Americans and Hispanics in Massachusetts, but a disparity in revascularization persisted. African Americans and Hispanics were 27% and 16% less likely to undergo revascularization procedures (coronary artery bypass grafting or percutaneous coronary intervention) than whites,51 suggesting that disparities in revascularization are not solely secondary to healthcare access.

These findings are consistent with a 2004 Veterans Administration study,52 in which healthcare access was equal among races. The study showed that African Americans received fewer cardiac procedures after an acute myocardial infarction compared with whites.

Have we made progress? The largest disparity between African Americans and whites in coronary artery disease mortality existed in 1990. The disparity persisted to 2012, and although decreased, it is projected to persist to 2030.53

DISPARITIES IN HEART FAILURE

An estimated 5.7 million Americans have heart failure, and 915,000 new cases are diagnosed annually.7 Unlike coronary artery disease, heart failure is expected to increase in prevalence by 46%, to 8 million Americans with heart failure by 2030.7,54

Our knowledge of disparities in the area of heart failure is derived primarily from epidemiologic studies. The Multi-Ethnic Study of Atherosclerosis55 showed that African Americans (4.6 per 1,000), followed by Hispanics (3.5 per 1,000) had a higher risk of developing heart failure compared with whites (2.4 per 1,000).The higher risk is in part due to disparities in socioeconomic status and prevalence of hypertension, as African Americans accounted for 75% of cases of nonischemic-related heart failure.55 African Americans also have a higher 5-year mortality rate than whites.55

Even though the 5-year mortality rate in heart failure is still 50%, the past 30 years have seen innovations in pharmacologic and device therapy and thus improved outcomes in heart failure patients. Still, significant gaps in the use of guideline-recommended therapies, quality of care, and clinical outcomes persist in contemporary practice for racial minorities with heart failure.

Disparities in inpatient care for heart failure

Patients admitted for heart failure and cared for by a cardiologist are more likely to be discharged on guideline-directed medical therapy, have fewer heart failure readmissions, and lower mortality.56,57 Breathett et al,58 in a study of 104,835 patients hospitalized in an intensive care unit for heart failure, found that primary intensive care by a cardiologist was associated with higher survival in both races. However, in the same study, white patients had a higher odds of receiving care from a cardiologist than African American patients.

Disparities and cardiac resynchronization therapy devices

In one-third of patients with heart failure, conduction delays result in dyssynchronous left ventricular contraction.59 Dyssynchrony leads to reduced cardiac performance, left ventricular remodeling, and increased mortality.56

Cardiac resynchronization therapy (CRT) was approved for clinical use in 2001, and studies have shown that it improves quality of life, exercise tolerance, cardiac performance, and morbidity and mortality rates.59–66 The 2013 ACC/AHA guidelines for the management of heart failure give a class IA recommendation (the highest) for its use in patients with a left ventricular ejection fraction of 35% or less, sinus rhythm, left bundle branch block and a QRS duration of 150 ms or greater, and New York Heart Association class II, III, or ambulatory IV symptoms while on guideline-directed medical therapy.67

Despite these recommendations, racial differences are observed. A study using the Nationwide Inpatient Sample database59 showed that between 2002 and 2010, a total of 374,202 CRT devices were implanted, averaging 41,578 annually. After adjusting for heart failure admissions, the study showed that CRT use was favored in men and in whites.

Another study, using the National Cardiovascular Data Registry,68 looked at patients who received implantable cardiac defibrillators (ICDs) and were eligible to receive CRT. It found that African Americans and Hispanics were less likely than whites to receive CRT, even though they were more likely to meet established criteria.

Disparities and left ventricular assist devices

The Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart failure (REMATCH) trial and Heart Mate II trial demonstrated that left ventricular assist devices (LVADs) were durable options for long-term support for patients with end-stage heart failure.69,70 Studies that examined the role of race and clinical outcomes after LVAD implantation have reported mixed findings.71,72 Few studies have looked at the role racial differences play in accessing LVAD therapy.

Joyce et al73 reviewed data from the Nationwide Inpatient Sample from 2002 to 2003 on patients admitted to the hospital with a primary diagnosis of heart failure or cardiogenic shock. A total of 297,866 patients were included in the study, of whom only 291 underwent LVAD implantation. A multivariate analysis found that factors such as age over 65, female sex, admission to a nonacademic center, geographic region, and African American race adversely influenced access to LVAD therapy.

Breathett et al74 evaluated racial differences in LVAD implantations from 2012 to 2015, a period that corresponds to increased health insurance expansion, and found LVAD implantations increased among African American patients with advanced heart failure, but no other racial or ethnic group.

 

 

Disparities and heart transplant

For patients with end-stage heart failure, orthotopic heart transplant is the most definitive and durable option for long-term survival. According to data from the United Network for Organ Sharing, 62,508 heart transplants were performed from January 1, 1988 to December 31, 2015. Compared with transplants of other solid organs, heart transplant occurs in significantly infrequent rates.

Barriers to transplant include lack of health insurance, considered a surrogate for low socioeconomic status. Hispanics and African Americans are less likely to have private health insurance than non-Hispanic whites, and this difference is magnified among the working poor.

Despite these perceived barriers, Kilic et al75 found that African Americans comprised 16.4% of heart transplant recipients, although they make up only approximately 13% of the US population. They also had significantly shorter wait-list times than whites. On the negative side, African Americans had a higher unadjusted mortality rate than whites (15% vs 12% P = .002). African Americans also tended to receive their transplants at centers with lower transplant volumes and higher transplant mortality rates.

Several other studies also showed that African Americans compared to whites have significantly worse outcomes after transplant.76–79 What accounts for this difference? Kilic et al75 showed that African Americans had the lowest proportion of blood type matching and lowest human leukocyte antigen matching, were younger (because African Americans develop more advanced heart failure at younger ages), had higher serum creatinine levels, and were more often bridged to transplant with an LVAD.

DISPARITIES IN CARDIOVASCULAR RESEARCH

Although the United States has the most sophisticated and robust medical system in the world, select groups have significant differences in delivery and healthcare outcomes. There are many explanations for these differences, but a contributing factor may be the paucity of research dedicated to understand racial and ethnic differences.80

Differences observed in epidemiologic studies may be secondary to pathophysiology, genetic differences, environment, and lifestyle choices. Historically, clinical trials were conducted in homogeneous populations with respect to age (middle-aged), sex (male), and race (white), and the results were generalized to heterogeneous populations.80

Disparities in research have implications in clinical practice. Overall, the primary cause of heart failure is ischemia; however, in African Americans, the primary cause is hypertensive heart disease.81 Studies in hypertension have shown that African Americans have less of a response to neurohormonal blockade with ACE inhibitors and beta-blockers than non-African Americans.82 Nevertheless, neurohormonal blockade has become the cornerstone of heart failure treatment.

Retrospective analysis of the Vasodilator-Heart Failure trials83 showed that treatment with isosorbide dinitrate plus hydralazine, compared with placebo, conferred a survival benefit for African Americans but not whites.80 No survival advantage was noted when isosorbide dinitrate/hydralazine was compared to enalapril in African Americans, although enalapril was superior to isosorbide dinitrate in whites.45 These observations were recognized 10 to 15 years after trial completion, and were only possible because the trials included sufficient numbers of African American patients to complete analysis.

In 1993, the US Congress passed the National Institutes of Health (NIH) Revitalization Act, which established guidelines requiring NIH grant applicants to include minorities in human subject research, as they were historically underrepresented in clinical research trials.84,85

In 2001, the Beta-Blocker Evaluation of Survival Trial86 reported its results investigating whether bucindolol, a nonselective beta-blocker, would reduce mortality in patients with advanced heart failure (New York Heart Association class III or IV). This was one of the first trials to prospectively investigate racial and ethnic differences in response to treatment. Though it showed no overall benefit in the use of bucindolol in the treatment of advanced heart failure, subgroup analysis showed that whites did enjoy a benefit in terms of lower mortality, whereas African Americans did not.

Results of the Vasodilator-Heart Failure trials led to further population-directed research, most notably the African American Heart Failure Trial,87 a double-blind, placebo-controlled, randomized trial in patients who identified as African American. Patients who were randomized to receive a fixed dose of hydralazine and isosorbide dinitrate had a 43% lower mortality rate, a 33% lower hospitalization rate for heart failure, and better quality of life than patients in the placebo group, leading to early termination of the trial. The outcomes suggested that the combination of isosorbide dinitrate and hydralazine treats heart failure in a manner independent of pure neurohormonal blockade.

CHALLENGES IN STUDY PARTICIPATION

Recruitment of minority participants in biomedical research is a challenging task for clinical investigators.88,89 Some of the factors thought to pose potential barriers for racial and ethnic minority participation in health research include poor access to primary medical care, failure of researchers to recruit minority populations actively, and language and cultural barriers.90

Further, it is widely claimed that African Americans are less willing than nonminority individuals to participate in clinical research trials due to general distrust of the medical community as a result of the Tuskegee Syphilis Experiment.91 That infamous study, conducted by the US Public Health Service between 1932 and 1972, sought to record the natural progression of untreated syphilis in poor African American men in Alabama. The participants were not informed of the true purpose of the study, and they were under the impression that they were simply receiving free healthcare from the US government. Further, they were denied appropriate treatment even after it became readily available, in order for researchers to observe the progression of the disease.

While the 1993 mandate did in fact increase pressure on researchers to develop strategies to overcome participation barriers, the issue of underrepresentation of racial minorities in clinical research, including cardiovascular research, has not been resolved and continues to be a problem today.

The overall goal of clinical research is to determine the best strategies to prevent and treat disease. But if the study population is not representative of the affected population at large, the results cannot be generalized to underrepresented subgroups. The implications of underrepresentation in research are far-reaching, and can further contribute to disparate care of minority patients such as African Americans, who have a higher prevalence of cardiovascular risk factors and greater burden of heart failure.

 

 

PROPOSING SOLUTIONS

Between 1986 and 2018, according to a PUBMED search, 10,462 articles highlighted the presence of a health-related disparity. Solutions to address and ultimately eradicate disparities will need to eliminate healthcare bias, increase patient access, and increase diversity and inclusion in the physician work force.

Eliminating bias

Implicit bias refers to attitudes, thoughts, and feelings that exist outside of the conscious awareness.92 These biases can be triggered by race, gender, or socioeconomic status. They have manifested in society as stereotypes that men are more competent than women, women are more verbal than men, and African Americans are more athletic than whites.93

The concept of implicit bias is important, in that the populations that experience the greatest health disparities also suffer from negative cultural stereotypes.94 Healthcare professionals are not inoculated against implicit bias.95 Studies have shown that most healthcare providers have implicit biases that reflect positive attitudes toward whites and negative attitudes toward people of color.92,94,96–98

The Implicit Association Test, introduced in 1998, is widely used to measure implicit bias. It measures response time of subjects to match particular social groups to particular attributes.99 Green et al,99 using this test, showed that although physicians reported no explicit preference for white vs African American patients or differences in perceived cooperativeness, the test revealed implicit preference favoring white Americans and implicit stereotypes of African Americans as less cooperative for medical procedures and in general. This also manifested in clinical decision-making, as white Americans were more likely, and African Americans less likely, to be treated with thrombolysis.99

Sabin et al100 showed that implicit bias was present among pediatricians, although less than in society as a whole and in other healthcare professionals.

But how does one change feelings that exist outside of the conscious awareness? Green et al99 showed that making physicians aware of their susceptibility to bias changed their behavior. A subset of physicians who were made aware that bias was a focus of the study were more likely to refer African Americans for thrombolysis even if they had a high degree of implicit pro-white bias.94,100 Perhaps mandating that all healthcare providers take a self-administered and confidentially reported Implicit Association Test will lead to awareness of implicit bias and minimize healthcare behaviors that contribute to the current state of disparities.

Improving access

Common indicators of access to healthcare include health insurance status, having a usual source of healthcare, and having a regular physician.101 Health insurance does offer protection from the costs associated with illness and health maintenance.101 It is also a major contributing factor in racial and ethnic disparities.

Chen et al102 examined the effects of the Affordable Care Act and found that it was associated with reduction in the probability of being uninsured, delaying necessary care, and forgoing necessary care, and increased probability of having a physician. However, earlier studies showed that access to health insurance by itself does not equate to equitable care.103,104

Diversifying the work force

African Americans comprise 4% of physicians and Hispanic Americans 5%, despite accounting for 13% and 16% of the US population.105 This underrepresentation has led to African American and Hispanic American patients being more likely than white patients to be treated by a physician from a dissimilar racial or ethnic background.106 Studies have shown that minority patients in a race- or ethnic-concordant relationship are more likely to use needed health services, less likely to postpone seeking care, and report greater satisfaction.106,107 Minority physicians often locate and practice in neighborhoods with high minority populations, and they disproportionately care for disadvantaged patients of lower socioeconomic status and poorer health.106,108

WE ARE STILL IN THE TUNNEL, BUT THERE IS LIGHT AT THE END

The cardiovascular community has faced tremendous challenges in the past and responded with innovative research that has led to imaging that aids in the diagnosis of subclinical cardiovascular disease and invasive and pharmacologic strategies that have improved cardiovascular outcomes. One may say that there is light at the end of the tunnel; however, the existence of disparate care reminds us that we are still in the tunnel.

Disparities in cardiovascular disease management present a unique challenge for the community. There is no drug, device, or invasive procedure to eliminate this pathology. However, by acknowledging the problem and implementing changes at the system, provider, and patient level, the cardiovascular community can achieve yet another momentous achievement: the end of cardiovascular health disparities. Cardiovascular disease makes no distinction in race, sex, age, or socioeconomic status, and neither should the medical community.

References
  1. Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial infarction. N Engl J Med 2012; 366(1):54–63. doi:10.1056/NEJMra1112570
  2. Braunwald E. Evolution of the management of acute myocardial infarction: a 20th century saga. Lancet 1998; 352(9142):1771–1774. doi:10.1016/S0140-6736(98)03212-7
  3. Julian DG. The history of coronary care units. Br Heart J 1987; 57(6):497–502. doi:10.1136/hrt.57.6.497
  4. Caswell JE. A brief history of coronary care units. Public Health Rep 1967; 82(12):1105–1111. pmid:19316519
  5. Day HW. History of coronary care units. Am J Cardiol 1972; 30(4):405–407. pmid:4560377
  6. Braunwald E. Shattuck lecture—cardiovascular medicine at the turn of the millennium: triumphs, concerns, and opportunities. N Engl J Med 1997; 337(19):1360–1369. doi:10.1056/NEJM199711063371906
  7. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee; Stroke Statistics Subcommittee. Heart disease and stroke statistics—2016 update: a report from the American Heart Association. Circulation 2016; 133(4):e38–e360. doi:10.1161/CIR.0000000000000350
  8. Nelson A. Unequal treatment: confronting racial and ethnic disparities in health care. J Natl Med Assoc 2002; 94(8):666–668. pmid:12152921
  9. McGuire TG, Alegria M, Cook BL, Wells KB, Zaslavsky AM. Implementing the Institute of Medicine definition of disparities: an application to mental health care. Health Serv Res 2006; 41(5):1979–2005. doi:10.1111/j.1475-6773.2006.00583.x
  10. Stone CT, Vanzant FR. Heart disease as seen in a southern clinic: a clinical and pathologic survey. JAMA 1927; 89(18):1473–1480. doi:10.1001/jama.1927.02690180005002
  11. Centers For Disease Control And Prevention (CDC). Vital signs: avoidable deaths from heart disease, stroke, and hypertensive disease—United States, 2001–2010. MMWR Morb Mortal Wkly Rep 2013; 62(35):727–727. pmid:PMC4585625
  12. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation 2015; 131(4):e29–e322. doi:10.1161/CIR.0000000000000152
  13. Ockene IS, Miller NH. Cigarette smoking, cardiovascular disease, and stroke: a statement for healthcare professionals from the American Heart Association. American Heart Association Task Force on Risk Reduction. Circulation 1997; 96(9):3243–3247. pmid:9386200
  14. Messner B, Bernhard D. Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis. Arterioscler Thromb Vasc Biol 2014; 34(3):509–515. doi:10.1161/ATVBAHA.113.300156
  15. Benjamin EJ, Virani SS, Callaway CW, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2018 update: a report from the American Heart Association. Circulation 2018; 137(12):e67–e492. doi:10.1161/CIR.0000000000000558
  16. Jamal A, Homa DM, O’Conner E, et al. Current cigarette smoking among adults—United States, 2005–2014. MMWR 2015; 64(44):1233–1240. doi:10.15585/mmwr.mm6444a2
  17. Myers J. Cardiology patient pages. Exercise and cardiovascular health. Circulation 2003; 107(1):e2–e5. pmid:12515760
  18. Shiroma EJ, Lee IM. Physical activity and cardiovascular health: lessons learned from epidemiological studies across age, gender, and race/ethnicity. Circulation 2010; 122(7):743–752. doi:10.1161/CIRCULATIONAHA.109.914721
  19. Piercy KL, Troiano RP, Ballard RM, et al. The physical activity guidelines for Americans. JAMA 2018; 320(19):2020–2028. doi:10.1001/jama.2018.14854
  20. Omura JD, Carlson SA, Paul P, et al. Adults eligible for cardiovascular disease prevention counseling and participation in aerobic physical activity—United States, 2013. MMWR 2015; 64(37):1047–1051. doi:10.15585/mmwr.mm6437a4
  21. Dunstan DW, Barr EL, Healy GN, et al. Television viewing time and mortality: the Australian diabetes, obesity and lifestyle study (AusDiab). Circulation 2010; 121(3):384–391. doi:10.1161/CIRCULATIONAHA.109.894824
  22. Warren TY, Barry V, Hooker SP, Sui X, Church TS, Blair SN. Sedentary behaviors increase risk of cardiovascular disease mortality in men. Med Sci Sports Exerc 2010; 42(5):879–885. doi:10.1249/MSS.0b013e3181c3aa7e
  23. Byun W, Dowda M, Pate RR. Associations between screen-based sedentary behavior and cardiovascular disease risk factors in Korean youth. J Korean Med Sci 2012; 27(4):388–394. doi:10.3346/jkms.2012.27.4.388
  24. Getz GS, Reardon CA. Nutrition and cardiovascular disease. Arterioscler Thromb Vasc Biol 2007; 27(12):2499–2506. doi:10.1161/ATVBAHA.107.155853
  25. Eckel RH. Obesity and heart disease: a statement for healthcare professionals from the Nutrition Committee, American Heart Association. Circulation 1997; 96(9):3248–3250. pmid:9386201
  26. Rimm EB, Stampfer MJ, Giovannucci E, et al. Body size and fat distribution as predictors of coronary heart disease among middle-aged and older US men. Am J Epidemiol 1995; 141(12):1117–1127. pmid:7771450
  27. Duflou J, Virmani R, Rabin I, Burke A, Farb A, Smialek J. Sudden death as a result of heart disease in morbid obesity. Am Heart J 1995; 130(2):306–313. pmid:7631612
  28. Poirier P, Giles TD, Bray GA, et al; American Heart Association; Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation 2006; 113(6):898–918. doi:10.1161/CIRCULATIONAHA.106.171016
  29. Wang Y. Disparities in pediatric obesity in the United States. Adv Nutr 2011; 2(1):23–31. doi:10.3945/an.110.000083
  30. Taveras EM, Gillman MW, Kleinman KP, Rich-Edwards JW, Rifas-Shiman SL. Reducing racial/ethnic disparities in childhood obesity: the role of early life risk factors. JAMA Pediatr 2013; 167(8):731–738. doi:10.1001/jamapediatrics.2013.85
  31. Powell-Wiley TM, Ayers CR, Banks-Richard K, et al. Disparities in counseling for lifestyle modification among obese adults: insights from the Dallas heart study. Obesity (Silver Spring) 2012; 20(4):849–855. doi:10.1038/oby.2011.242
  32. Fuchs FD. Why do black Americans have higher prevalence of hypertension?: an enigma still unsolved. Hypertension 2011; 57(3):379–380. doi:10.1161/HYPERTENSIONAHA.110.163196
  33. Ferdinand KC, Armani AM. The management of hypertension in African Americans. Crit Pathw Cardiol 2007; 6(2):67–71. doi:10.1097/HPC.0b013e318053da59
  34. Voors AW, Webber LS, Berenson GS. Time course study of blood pressure in children over a three-year period. Bogalusa Heart Study. Hypertension 1980; 2(4 Pt 2):102–108. pmid:7399641
  35. Carson AP, Howard G, Burke GL, Shea S, Levitan EB, Muntner P. Ethnic differences in hypertension incidence among middle-aged and older adults: the multi-ethnic study of atherosclerosis. Hypertension 2011; 57(6):1101–1107. doi:10.1161/HYPERTENSIONAHA.110.168005
  36. Gu A, Yue Y, Desai RP, Argulian E. Racial and ethnic differences in antihypertensive medication use and blood pressure control among US adults with hypertension: the National Health and Nutrition Examination Survey, 2003 to 2012. Circ Cardiovasc Qual Outcomes 2017; 10(1). pii:e003166. doi:10.1161/CIRCOUTCOMES.116.003166
  37. Lackland DT. Racial differences in hypertension: implications for high blood pressure management. Am J Med Sci 2014; 348(2):135–138. doi:10.1097/MAJ.0000000000000308
  38. Bosworth HB, Dudley T, Olsen MK, et al. Racial differences in blood pressure control: potential explanatory factors. Am J Med 2006; 119(1):70.e9–e15. doi:10.1016/j.amjmed.2005.08.019
  39. Peck RN, Smart LR, Beier R, et al. Difference in blood pressure response to ACE-Inhibitor monotherapy between black and white adults with arterial hypertension: a meta-analysis of 13 clinical trials. BMC Nephrol 2013; 14:201. doi:10.1186/1471-2369-14-201
  40. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2018; 138(17):e484–e594. doi:10.1161/CIR.0000000000000596
  41. Peek ME, Cargill A, Huang ES. Diabetes health disparities: a systematic review of health care interventions. Med Care Res Rev 2007; 64(5 suppl):101S–156S. doi:10.1177/1077558707305409
  42. Narayan KM, Boyle JP, Geiss LS, Saaddine JB, Thompson TJ. Impact of recent increase in incidence on future diabetes burden: US, 2005–2050. Diabetes Care 2006; 29(9):2114–2116. doi:10.2337/dc06-1136
  43. Johnson PA, Lee TH, Cook EF, Rouan GW, Goldman L. Effect of race on the presentation and management of patients with acute chest pain. Ann Intern Med 1993; 118(8):593–601. pmid:8452325
  44. Hannan EL, van Ryn M, Burke J, et al. Access to coronary artery bypass surgery by race/ethnicity and gender among patients who are appropriate for surgery. Med Care 1999; 37(1):68–77. pmid:10413394
  45. Peterson ED, Shaw LK, DeLong ER, Pryor DB, Califf RM, Mark DB. Racial variation in the use of coronary-revascularization procedures. Are the differences real? Do they matter? N Engl J Med 1997; 336(7):480–486. doi:10.1056/NEJM199702133360706
  46. Wenneker MB, Epstein AM. Racial inequalities in the use of procedures for patients with ischemic heart disease in Massachusetts. JAMA 1989; 261(2):253–257. pmid:2521191
  47. Allison JJ, Kiefe CI, Centor RM, Box JB, Farmer RM. Racial differences in the medical treatment of elderly Medicare patients with acute myocardial infarction. J Gen Intern Med 1996; 11(12):736–743. pmid:9016420
  48. Mickelson JK, Blum CM, Geraci JM. Acute myocardial infarction: clinical characteristics, management and outcome in a metropolitan Veterans Affairs Medical Center teaching hospital. J Am Coll Cardiol 1997; 29(5):915–925. pmid:9120176
  49. Yarzebski J, Bujor CF, Lessard D, Gore JM, Goldberg RJ. Recent and temporal trends (1975 to 1999) in the treatment, hospital, and long-term outcomes of Hispanic and non-Hispanic white patients hospitalized with acute myocardial infarction: a population-based perspective. Am Heart J 2004; 147(4):690–697. doi:10.1016/j.ahj.2003.10.023
  50. Riley WJ. Health disparities: gaps in access, quality and affordability of medical care. Trans Am Clin Climatol Assoc 2012; 123:167–172. pmid:23303983
  51. Albert MA, Ayanian JZ, Silbaugh TS, et al. Early results of Massachusetts healthcare reform on racial, ethnic, and socioeconomic disparities in cardiovascular care. Circulation 2014; 129(24):2528–2538. doi:10.1161/CIRCULATIONAHA.113.005231
  52. Peterson ED, Wright SM, Daley J, Thibault GE. Racial variation in cardiac procedure use and survival following acute myocardial infarction in the Department of Veterans Affairs. JAMA 1994; 271(15):1175–1180. pmid:8151875
  53. Pearson-Stuttard J, Guzman-Castillo M, Penalvo JL, et al. Modeling future cardiovascular disease mortality in the United States: national trends and racial and ethnic disparities. Circulation 2016; 133(10):967–978. doi:10.1161/CIRCULATIONAHA.115.019904
  54. Heidenreich PA, Albert NM, Allen LA, et al; American Heart Association Advocacy Coordinating Committee; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Stroke Council. Forecasting the impact of heart failure in the United States: a policy statement from the American Heart Association. Circ Heart Fail 2013; 6(3):606–619. doi:10.1161/HHF.0b013e318291329a
  55. Bahrami H, Kronmal R, Bluemke DA, et al. Differences in the incidence of congestive heart failure by ethnicity: the multi-ethnic study of atherosclerosis. Arch Intern Med 2008; 168(19):2138–2145. doi:10.1001/archinte.168.19.2138
  56. Parmar KR, Xiu PY, Chowdhury MR, Patel E, Cohen M. In-hospital treatment and outcomes of heart failure in specialist and non-specialist services: a retrospective cohort study in the elderly. Open Heart 2015; 2(1):e000095. doi:10.1136/openhrt-2014-000095
  57. Avaldi VM, Lenzi J, Urbinati S, et al. Effect of cardiologist care on 6-month outcomes in patients discharged with heart failure: results from an observational study based on administrative data. BMJ Open 2017; 7(11):e018243. doi:10.1136/bmjopen-2017-018243
  58. Breathett K, Liu WG, Allen LA, et al. African Americans are less likely to receive care by a cardiologist during an intensive care unit admission for heart failure. JACC Heart Fail 2018; 6(5):413–420. doi:10.1016/j.jchf.2018.02.015
  59. Sridhar AR, Yarlagadda V, Parasa S, et al. Cardiac resynchronization therapy: US trends and disparities in utilization and outcomes. Circ Arrhythm Electrophysiol 2016; 9(3):e003108. doi:10.1161/CIRCEP.115.003108
  60. Abraham WT, Fisher WG, Smith AL, et al; MIRACLE Study Group. Multicenter insync randomized clinical evaluation. Cardiac resynchronization in chronic heart failure. N Engl J Med 2002; 346(24):1845–1853. doi:10.1056/NEJMoa013168
  61. Auricchio A, Stellbrink C, Sack S, et al; Pacing Therapies in Congestive Heart Failure (PATH-CHF) Study Group. Long-term clinical effect of hemodynamically optimized cardiac resynchronization therapy in patients with heart failure and ventricular conduction delay. J Am Coll Cardiol 2002; 39(12):2026–2033. pmid:12084604
  62. Cazeau S, Leclercq C, Lavergne T, et al; Multisite Stimulation in Cardiomyopathies (MUSTIC) Study Investigators. Effects of multisite biventricular pacing in patients with heart failure and intraventricular conduction delay. N Engl J Med 2001; 344(12):873–880. doi:10.1056/NEJM200103223441202
  63. Higgins SL, Hummel JD, Niazi IK, et al. Cardiac resynchronization therapy for the treatment of heart failure in patients with intraventricular conduction delay and malignant ventricular tachyarrhythmias. J Am Coll Cardiol 2003; 42(8):1454–1459. pmid:14563591
  64. Young JB, Abraham WT, Smith AL, et al; Multicenter InSync ICD Randomized Clinical Evaluation (MIRACLE ICD) Trial Investigators. Combined cardiac resynchronization and implantable cardioversion defibrillation in advanced chronic heart failure: the MIRACLE ICD Trial. JAMA 2003; 289(20):2685–2694. doi:10.1001/jama.289.20.2685
  65. Sutton MG, Plappert T, Hilpisch KE, Abraham WT, Hayes DL, Chinchoy E. Sustained reverse left ventricular structural remodeling with cardiac resynchronization at one year is a function of etiology: quantitative Doppler echocardiographic evidence from the Multicenter InSync Randomized Clinical Evaluation (MIRACLE). Circulation 2006; 113(2):266–272. doi:10.1161/CIRCULATIONAHA.104.520817
  66. Cleland JG, Daubert JC, Erdmann E, et al; Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The effect of cardiac resynchronization on morbidity and mortality in heart failure. N Engl J Med 2005; 352(15):1539–1549. doi:10.1056/NEJMoa050496
  67. Yancy CW, Jessup M, Bozkurt B, et al; American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation 2013; 128(16):e240–e327. doi:10.1161/CIR.0b013e31829e8776
  68. Farmer SA, Kirkpatrick JN, Heidenreich PA, Curtis JP, Wang Y, Groeneveld PW. Ethnic and racial disparities in cardiac resynchronization therapy. Heart Rhythm 2009; 6(3):325–331. doi:10.1016/j.hrthm.2008.12.018
  69. Rose EA, Gelijns AC, Moskowitz AJ, et al; Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH) Study Group. Long-term use of a left ventricular assist device for end-stage heart failure. N Engl J Med 2001; 345(20):1435–1443. doi:10.1056/NEJMoa012175
  70. Slaughter MS, Rogers JG, Milano CA, et al; HeartMate II Investigators. Advanced heart failure treated with continuous-flow left ventricular assist device. N Engl J Med 2009; 361(23):2241–2251. doi:10.1056/NEJMoa0909938
  71. Tsiouris A, Brewer RJ, Borgi J, Nemeh H, Paone G, Morgan JA. Continuous-flow left ventricular assist device implantation as a bridge to transplantation or destination therapy: racial disparities in outcomes. J Heart Lung Transplant 2013; 2(3):299–304. doi:10.1016/j.healun.2012.11.017
  72. Stulak JM, Deo S, Cowger J, et al. Do racial and sex disparities exist in clinical characteristics and outcomes for patients undergoing left ventricular assist device implantation? J Heart Lung Transplant 2013; 32(45):S279–S280.
  73. Joyce DL, Conte JV, Russell SD, Joyce LD, Chang DC. Disparities in access to left ventricular assist device therapy. J Surg Res 2009; 152(1):111–117. doi:10.1016/j.jss.2008.02.065
  74. Breathett K, Allen LA, Helmkamp L, et al. Temporal trends in contemporary use of ventricular assist devices by race and ethnicity. Circ Heart Fail 2018; 11(8):e005008. doi:10.1161/CIRCHEARTFAILURE.118.005008
  75. Kilic A, Higgins RS, Whitson BA, Kilic A. Racial disparities in outcomes of adult heart transplantation. Circulation 2015; 131(10):882–889. doi:10.1161/CIRCULATIONAHA.114.011676
  76. Liu V, Bhattacharya J, Weill D, Hlatky MA. Persistent racial disparities in survival after heart transplantation. Circulation 2011; 123(15):1642–1649. doi:10.1161/CIRCULATIONAHA.110.976811
  77. Mahle WT, Kanter KR, Vincent RN. Disparities in outcome for black patients after pediatric heart transplantation. J Pediatr 2005; 147(6):739–743. doi:10.1016/j.jpeds.2005.07.018
  78. Park MH, Tolman DE, Kimball PM. The impact of race and HLA matching on long-term survival following cardiac transplantation. Transplant Proc 1997; 29(1–2):1460–1463. pmid:9123381
  79. Higgins RS, Fishman JA. Disparities in solid organ transplantation for ethnic minorities: facts and solutions. Am J Transplant 2006; 6(11):2556–2562. doi:10.1111/j.1600-6143.2006.01514.x
  80. Taylor AL, Wright JT Jr. Should ethnicity serve as the basis for clinical trial design? Importance of race/ethnicity in clinical trials: lessons from the African-American Heart Failure Trial (A-HeFT), the African-American Study of Kidney Disease and Hypertension (AASK), and the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Circulation 2005; 112(23):3654–3660. doi:10.1161/CIRCULATIONAHA.105.540443
  81. Yancy CW. Heart failure in African Americans: a cardiovascular engima. J Card Fail 2000; 6(3):183–186. pmid:10997742
  82. Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report. JAMA 2003; 289(19):2560–2572. doi:10.1001/jama.289.19.2560
  83. Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration cooperative study. N Engl J Med 1986; 314(24):1547–1552. doi:10.1056/NEJM198606123142404
  84. Chen MS Jr, Lara PN, Dang JH, Paterniti DA, Kelly K. Twenty years post-NIH Revitalization Act: enhancing minority participation in clinical trials (EMPaCT): laying the groundwork for improving minority clinical trial accrual: renewing the case for enhancing minority participation in cancer clinical trials. Cancer 2014;120(suppl 7):1091–1096. doi:10.1002/cncr.28575
  85. Geller SE, Koch A, Pellettieri B, Carnes M. Inclusion, analysis, and reporting of sex and race/ethnicity in clinical trials: have we made progress? J Womens Health (Larchmt) 2011; 20(3):315–320. doi:10.1089/jwh.2010.2469
  86. Beta-Blocker Evaluation of Survival Trial Investigators; Eichhorn EJ, Domanski MJ, Krause-Steinrauf H, Bristow MR, Lavori PW. A trial of the beta-blocker bucindolol in patients with advanced chronic heart failure. N Engl J Med 2001; 344(22):1659–1667. doi:10.1056/NEJM200105313442202
  87. Taylor AL, Ziesche S, Yancy C, et al; African-American Heart Failure Trial Investigators. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med 2004; 351(20):2049–2057. doi:10.1056/NEJMoa042934
  88. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med 1999; 14(9):537–546. pmid:10491242
  89. Swanson GM, Ward AJ. Recruiting minorities into clinical trials: toward a participant-friendly system. J Natl Cancer Inst 1995; 87(23):1747–1759. doi:10.1093/jnci/87.23.1747
  90. Institute of Medicine (US) Committee on Understanding and Eliminating Racial and Ethnic Disparities in Health Care; Smedley BD, Stith AY, Nelson AR, eds. Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care. Washington, DC: National Academies Press (US); 2003. https://www.ncbi.nlm.nih.gov/books/NBK220358/. Accessed May 13, 2019.
  91. Fisher JA, Kalbaugh CA. Challenging assumptions about minority participation in US clinical research. Am J Public Health 2011; 101(12):2217–2222. doi:10.2105/AJPH.2011.300279
  92. Hall WJ, Chapman MV, Lee KM, et al. Implicit racial/ethnic bias among health care professionals and its influence on health care outcomes: a systematic review. Am J Public Health 2015; 105(12):e60–e76. doi:10.2105/AJPH.2015.302903
  93. Biernat M, Manis M. Shifting standards and stereotype-based judgments. J Pers Soc Psychol 1994; 66(1):5–20. pmid:8126651
  94. Chapman EN, Kaatz A, Carnes M. Physicians and implicit bias: how doctors may unwittingly perpetuate health care disparities. J Gen Intern Med 2013; 28(11):1504–1510. doi:10.1007/s11606-013-2441-1
  95. FitzGerald C, Hurst S. Implicit bias in healthcare professionals: a systematic review. BMC Med Ethics 2017; 18(1):19. doi:10.1186/s12910-017-0179-8
  96. van Ryn M, Burke J. The effect of patient race and socio-economic status on physicians’ perceptions of patients. Soc Sci Med 2000; 50(6):813–828. pmid:10695979
  97. Mayo RM, Sherrill WW, Sundareswaran P, Crew L. Attitudes and perceptions of Hispanic patients and health care providers in the treatment of Hispanic patients: a review of the literature. Hisp Health Care Int 2007; 5(2):64–72.
  98. Blair IV, Steiner JF, Havranek EP. Unconscious (implicit) bias and health disparities: where do we go from here? Perm J 2011; 15(2):71–78. pmid:21841929
  99. Green AR, Carney DR, Pallin DJ, et al. Implicit bias among physicians and its prediction of thrombolysis decisions for black and white patients. J Gen Intern Med 2007; 22(9):1231–1238. doi:10.1007/s11606-007-0258-5
  100. Sabin JA, Rivara FP, Greenwald AG. Physician implicit attitudes and stereotypes about race and quality of medical care. Med Care 2008; 46(7):678–685. doi:10.1097/MLR.0b013e3181653d58
  101. Smedley BD, Stith AY, Colburn L, et al; Institute of Medicine. The Right Thing to Do, The Smart Thing to Do: Enhancing Diversity in the Health Professions: Summary of the Symposium on Diversity in Health Professions in Honor of Herbert W. Nickens, MD. Washington, DC: National Academies Press; 2001. https://www.ncbi.nlm.nih.gov/books/NBK223633/. Accessed May 13, 2019.
  102. Chen J, Vargas-Bustamante A, Mortensen K, Ortega AN. Racial and ethnic disparities in health care access and utilization under the Affordable Care Act. Med Care 2016; 54(2):140–146. doi:10.1097/MLR.0000000000000467
  103. Saha S, Freeman M, Toure J, Tippens KM, Weeks C, Ibrahim S. Racial and ethnic disparities in the VA health care system: a systematic review. J Gen Intern Med 2008; 23(5):654–671. doi:10.1007/s11606-008-0521-4
  104. McCormick D, Sayah A, Lokko H, Woolhandler S, Nardin R. Access to care after Massachusetts’ health care reform: a safety net hospital patient survey. J Gen Intern Med 2012; 27(11):1548–1554. doi:10.1007/s11606-012-2173-7
  105. Burgos JL, Yee D, Csordas T, et al. Supporting the minority physician pipeline: providing global health experiences to undergraduate students in the United States-Mexico border region. Med Educ Online 2015; 20:27260. doi:10.3402/meo.v20.27260
  106. Traylor AH, Schmittdiel JA, Uratsu CS, Mangione CM, Subramanian U. The predictors of patient–physician race and ethnic concordance: a medical facility fixed-effects approach. Health Serv Res 2010; 45(3):792–805. doi:10.1111/j.1475-6773.2010.01086.x
  107. LaVeist TA, Nuru-Jeter A. Is doctor-patient race concordance associated with greater satisfaction with care? J Health Soc Behav 2002; 43(3):296–306. pmid:12467254
  108. Marrast LM, Zallman L, Woolhandler S, Bor DH, McCormick D. Minority physicians’ role in the care of underserved patients: diversifying the physician workforce may be key in addressing health disparities. JAMA Intern Med 2014; 174(2):289–291. doi:10.1001/jamainternmed.2013.12756
References
  1. Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial infarction. N Engl J Med 2012; 366(1):54–63. doi:10.1056/NEJMra1112570
  2. Braunwald E. Evolution of the management of acute myocardial infarction: a 20th century saga. Lancet 1998; 352(9142):1771–1774. doi:10.1016/S0140-6736(98)03212-7
  3. Julian DG. The history of coronary care units. Br Heart J 1987; 57(6):497–502. doi:10.1136/hrt.57.6.497
  4. Caswell JE. A brief history of coronary care units. Public Health Rep 1967; 82(12):1105–1111. pmid:19316519
  5. Day HW. History of coronary care units. Am J Cardiol 1972; 30(4):405–407. pmid:4560377
  6. Braunwald E. Shattuck lecture—cardiovascular medicine at the turn of the millennium: triumphs, concerns, and opportunities. N Engl J Med 1997; 337(19):1360–1369. doi:10.1056/NEJM199711063371906
  7. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee; Stroke Statistics Subcommittee. Heart disease and stroke statistics—2016 update: a report from the American Heart Association. Circulation 2016; 133(4):e38–e360. doi:10.1161/CIR.0000000000000350
  8. Nelson A. Unequal treatment: confronting racial and ethnic disparities in health care. J Natl Med Assoc 2002; 94(8):666–668. pmid:12152921
  9. McGuire TG, Alegria M, Cook BL, Wells KB, Zaslavsky AM. Implementing the Institute of Medicine definition of disparities: an application to mental health care. Health Serv Res 2006; 41(5):1979–2005. doi:10.1111/j.1475-6773.2006.00583.x
  10. Stone CT, Vanzant FR. Heart disease as seen in a southern clinic: a clinical and pathologic survey. JAMA 1927; 89(18):1473–1480. doi:10.1001/jama.1927.02690180005002
  11. Centers For Disease Control And Prevention (CDC). Vital signs: avoidable deaths from heart disease, stroke, and hypertensive disease—United States, 2001–2010. MMWR Morb Mortal Wkly Rep 2013; 62(35):727–727. pmid:PMC4585625
  12. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation 2015; 131(4):e29–e322. doi:10.1161/CIR.0000000000000152
  13. Ockene IS, Miller NH. Cigarette smoking, cardiovascular disease, and stroke: a statement for healthcare professionals from the American Heart Association. American Heart Association Task Force on Risk Reduction. Circulation 1997; 96(9):3243–3247. pmid:9386200
  14. Messner B, Bernhard D. Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis. Arterioscler Thromb Vasc Biol 2014; 34(3):509–515. doi:10.1161/ATVBAHA.113.300156
  15. Benjamin EJ, Virani SS, Callaway CW, et al; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2018 update: a report from the American Heart Association. Circulation 2018; 137(12):e67–e492. doi:10.1161/CIR.0000000000000558
  16. Jamal A, Homa DM, O’Conner E, et al. Current cigarette smoking among adults—United States, 2005–2014. MMWR 2015; 64(44):1233–1240. doi:10.15585/mmwr.mm6444a2
  17. Myers J. Cardiology patient pages. Exercise and cardiovascular health. Circulation 2003; 107(1):e2–e5. pmid:12515760
  18. Shiroma EJ, Lee IM. Physical activity and cardiovascular health: lessons learned from epidemiological studies across age, gender, and race/ethnicity. Circulation 2010; 122(7):743–752. doi:10.1161/CIRCULATIONAHA.109.914721
  19. Piercy KL, Troiano RP, Ballard RM, et al. The physical activity guidelines for Americans. JAMA 2018; 320(19):2020–2028. doi:10.1001/jama.2018.14854
  20. Omura JD, Carlson SA, Paul P, et al. Adults eligible for cardiovascular disease prevention counseling and participation in aerobic physical activity—United States, 2013. MMWR 2015; 64(37):1047–1051. doi:10.15585/mmwr.mm6437a4
  21. Dunstan DW, Barr EL, Healy GN, et al. Television viewing time and mortality: the Australian diabetes, obesity and lifestyle study (AusDiab). Circulation 2010; 121(3):384–391. doi:10.1161/CIRCULATIONAHA.109.894824
  22. Warren TY, Barry V, Hooker SP, Sui X, Church TS, Blair SN. Sedentary behaviors increase risk of cardiovascular disease mortality in men. Med Sci Sports Exerc 2010; 42(5):879–885. doi:10.1249/MSS.0b013e3181c3aa7e
  23. Byun W, Dowda M, Pate RR. Associations between screen-based sedentary behavior and cardiovascular disease risk factors in Korean youth. J Korean Med Sci 2012; 27(4):388–394. doi:10.3346/jkms.2012.27.4.388
  24. Getz GS, Reardon CA. Nutrition and cardiovascular disease. Arterioscler Thromb Vasc Biol 2007; 27(12):2499–2506. doi:10.1161/ATVBAHA.107.155853
  25. Eckel RH. Obesity and heart disease: a statement for healthcare professionals from the Nutrition Committee, American Heart Association. Circulation 1997; 96(9):3248–3250. pmid:9386201
  26. Rimm EB, Stampfer MJ, Giovannucci E, et al. Body size and fat distribution as predictors of coronary heart disease among middle-aged and older US men. Am J Epidemiol 1995; 141(12):1117–1127. pmid:7771450
  27. Duflou J, Virmani R, Rabin I, Burke A, Farb A, Smialek J. Sudden death as a result of heart disease in morbid obesity. Am Heart J 1995; 130(2):306–313. pmid:7631612
  28. Poirier P, Giles TD, Bray GA, et al; American Heart Association; Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation 2006; 113(6):898–918. doi:10.1161/CIRCULATIONAHA.106.171016
  29. Wang Y. Disparities in pediatric obesity in the United States. Adv Nutr 2011; 2(1):23–31. doi:10.3945/an.110.000083
  30. Taveras EM, Gillman MW, Kleinman KP, Rich-Edwards JW, Rifas-Shiman SL. Reducing racial/ethnic disparities in childhood obesity: the role of early life risk factors. JAMA Pediatr 2013; 167(8):731–738. doi:10.1001/jamapediatrics.2013.85
  31. Powell-Wiley TM, Ayers CR, Banks-Richard K, et al. Disparities in counseling for lifestyle modification among obese adults: insights from the Dallas heart study. Obesity (Silver Spring) 2012; 20(4):849–855. doi:10.1038/oby.2011.242
  32. Fuchs FD. Why do black Americans have higher prevalence of hypertension?: an enigma still unsolved. Hypertension 2011; 57(3):379–380. doi:10.1161/HYPERTENSIONAHA.110.163196
  33. Ferdinand KC, Armani AM. The management of hypertension in African Americans. Crit Pathw Cardiol 2007; 6(2):67–71. doi:10.1097/HPC.0b013e318053da59
  34. Voors AW, Webber LS, Berenson GS. Time course study of blood pressure in children over a three-year period. Bogalusa Heart Study. Hypertension 1980; 2(4 Pt 2):102–108. pmid:7399641
  35. Carson AP, Howard G, Burke GL, Shea S, Levitan EB, Muntner P. Ethnic differences in hypertension incidence among middle-aged and older adults: the multi-ethnic study of atherosclerosis. Hypertension 2011; 57(6):1101–1107. doi:10.1161/HYPERTENSIONAHA.110.168005
  36. Gu A, Yue Y, Desai RP, Argulian E. Racial and ethnic differences in antihypertensive medication use and blood pressure control among US adults with hypertension: the National Health and Nutrition Examination Survey, 2003 to 2012. Circ Cardiovasc Qual Outcomes 2017; 10(1). pii:e003166. doi:10.1161/CIRCOUTCOMES.116.003166
  37. Lackland DT. Racial differences in hypertension: implications for high blood pressure management. Am J Med Sci 2014; 348(2):135–138. doi:10.1097/MAJ.0000000000000308
  38. Bosworth HB, Dudley T, Olsen MK, et al. Racial differences in blood pressure control: potential explanatory factors. Am J Med 2006; 119(1):70.e9–e15. doi:10.1016/j.amjmed.2005.08.019
  39. Peck RN, Smart LR, Beier R, et al. Difference in blood pressure response to ACE-Inhibitor monotherapy between black and white adults with arterial hypertension: a meta-analysis of 13 clinical trials. BMC Nephrol 2013; 14:201. doi:10.1186/1471-2369-14-201
  40. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2018; 138(17):e484–e594. doi:10.1161/CIR.0000000000000596
  41. Peek ME, Cargill A, Huang ES. Diabetes health disparities: a systematic review of health care interventions. Med Care Res Rev 2007; 64(5 suppl):101S–156S. doi:10.1177/1077558707305409
  42. Narayan KM, Boyle JP, Geiss LS, Saaddine JB, Thompson TJ. Impact of recent increase in incidence on future diabetes burden: US, 2005–2050. Diabetes Care 2006; 29(9):2114–2116. doi:10.2337/dc06-1136
  43. Johnson PA, Lee TH, Cook EF, Rouan GW, Goldman L. Effect of race on the presentation and management of patients with acute chest pain. Ann Intern Med 1993; 118(8):593–601. pmid:8452325
  44. Hannan EL, van Ryn M, Burke J, et al. Access to coronary artery bypass surgery by race/ethnicity and gender among patients who are appropriate for surgery. Med Care 1999; 37(1):68–77. pmid:10413394
  45. Peterson ED, Shaw LK, DeLong ER, Pryor DB, Califf RM, Mark DB. Racial variation in the use of coronary-revascularization procedures. Are the differences real? Do they matter? N Engl J Med 1997; 336(7):480–486. doi:10.1056/NEJM199702133360706
  46. Wenneker MB, Epstein AM. Racial inequalities in the use of procedures for patients with ischemic heart disease in Massachusetts. JAMA 1989; 261(2):253–257. pmid:2521191
  47. Allison JJ, Kiefe CI, Centor RM, Box JB, Farmer RM. Racial differences in the medical treatment of elderly Medicare patients with acute myocardial infarction. J Gen Intern Med 1996; 11(12):736–743. pmid:9016420
  48. Mickelson JK, Blum CM, Geraci JM. Acute myocardial infarction: clinical characteristics, management and outcome in a metropolitan Veterans Affairs Medical Center teaching hospital. J Am Coll Cardiol 1997; 29(5):915–925. pmid:9120176
  49. Yarzebski J, Bujor CF, Lessard D, Gore JM, Goldberg RJ. Recent and temporal trends (1975 to 1999) in the treatment, hospital, and long-term outcomes of Hispanic and non-Hispanic white patients hospitalized with acute myocardial infarction: a population-based perspective. Am Heart J 2004; 147(4):690–697. doi:10.1016/j.ahj.2003.10.023
  50. Riley WJ. Health disparities: gaps in access, quality and affordability of medical care. Trans Am Clin Climatol Assoc 2012; 123:167–172. pmid:23303983
  51. Albert MA, Ayanian JZ, Silbaugh TS, et al. Early results of Massachusetts healthcare reform on racial, ethnic, and socioeconomic disparities in cardiovascular care. Circulation 2014; 129(24):2528–2538. doi:10.1161/CIRCULATIONAHA.113.005231
  52. Peterson ED, Wright SM, Daley J, Thibault GE. Racial variation in cardiac procedure use and survival following acute myocardial infarction in the Department of Veterans Affairs. JAMA 1994; 271(15):1175–1180. pmid:8151875
  53. Pearson-Stuttard J, Guzman-Castillo M, Penalvo JL, et al. Modeling future cardiovascular disease mortality in the United States: national trends and racial and ethnic disparities. Circulation 2016; 133(10):967–978. doi:10.1161/CIRCULATIONAHA.115.019904
  54. Heidenreich PA, Albert NM, Allen LA, et al; American Heart Association Advocacy Coordinating Committee; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Stroke Council. Forecasting the impact of heart failure in the United States: a policy statement from the American Heart Association. Circ Heart Fail 2013; 6(3):606–619. doi:10.1161/HHF.0b013e318291329a
  55. Bahrami H, Kronmal R, Bluemke DA, et al. Differences in the incidence of congestive heart failure by ethnicity: the multi-ethnic study of atherosclerosis. Arch Intern Med 2008; 168(19):2138–2145. doi:10.1001/archinte.168.19.2138
  56. Parmar KR, Xiu PY, Chowdhury MR, Patel E, Cohen M. In-hospital treatment and outcomes of heart failure in specialist and non-specialist services: a retrospective cohort study in the elderly. Open Heart 2015; 2(1):e000095. doi:10.1136/openhrt-2014-000095
  57. Avaldi VM, Lenzi J, Urbinati S, et al. Effect of cardiologist care on 6-month outcomes in patients discharged with heart failure: results from an observational study based on administrative data. BMJ Open 2017; 7(11):e018243. doi:10.1136/bmjopen-2017-018243
  58. Breathett K, Liu WG, Allen LA, et al. African Americans are less likely to receive care by a cardiologist during an intensive care unit admission for heart failure. JACC Heart Fail 2018; 6(5):413–420. doi:10.1016/j.jchf.2018.02.015
  59. Sridhar AR, Yarlagadda V, Parasa S, et al. Cardiac resynchronization therapy: US trends and disparities in utilization and outcomes. Circ Arrhythm Electrophysiol 2016; 9(3):e003108. doi:10.1161/CIRCEP.115.003108
  60. Abraham WT, Fisher WG, Smith AL, et al; MIRACLE Study Group. Multicenter insync randomized clinical evaluation. Cardiac resynchronization in chronic heart failure. N Engl J Med 2002; 346(24):1845–1853. doi:10.1056/NEJMoa013168
  61. Auricchio A, Stellbrink C, Sack S, et al; Pacing Therapies in Congestive Heart Failure (PATH-CHF) Study Group. Long-term clinical effect of hemodynamically optimized cardiac resynchronization therapy in patients with heart failure and ventricular conduction delay. J Am Coll Cardiol 2002; 39(12):2026–2033. pmid:12084604
  62. Cazeau S, Leclercq C, Lavergne T, et al; Multisite Stimulation in Cardiomyopathies (MUSTIC) Study Investigators. Effects of multisite biventricular pacing in patients with heart failure and intraventricular conduction delay. N Engl J Med 2001; 344(12):873–880. doi:10.1056/NEJM200103223441202
  63. Higgins SL, Hummel JD, Niazi IK, et al. Cardiac resynchronization therapy for the treatment of heart failure in patients with intraventricular conduction delay and malignant ventricular tachyarrhythmias. J Am Coll Cardiol 2003; 42(8):1454–1459. pmid:14563591
  64. Young JB, Abraham WT, Smith AL, et al; Multicenter InSync ICD Randomized Clinical Evaluation (MIRACLE ICD) Trial Investigators. Combined cardiac resynchronization and implantable cardioversion defibrillation in advanced chronic heart failure: the MIRACLE ICD Trial. JAMA 2003; 289(20):2685–2694. doi:10.1001/jama.289.20.2685
  65. Sutton MG, Plappert T, Hilpisch KE, Abraham WT, Hayes DL, Chinchoy E. Sustained reverse left ventricular structural remodeling with cardiac resynchronization at one year is a function of etiology: quantitative Doppler echocardiographic evidence from the Multicenter InSync Randomized Clinical Evaluation (MIRACLE). Circulation 2006; 113(2):266–272. doi:10.1161/CIRCULATIONAHA.104.520817
  66. Cleland JG, Daubert JC, Erdmann E, et al; Cardiac Resynchronization-Heart Failure (CARE-HF) Study Investigators. The effect of cardiac resynchronization on morbidity and mortality in heart failure. N Engl J Med 2005; 352(15):1539–1549. doi:10.1056/NEJMoa050496
  67. Yancy CW, Jessup M, Bozkurt B, et al; American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation 2013; 128(16):e240–e327. doi:10.1161/CIR.0b013e31829e8776
  68. Farmer SA, Kirkpatrick JN, Heidenreich PA, Curtis JP, Wang Y, Groeneveld PW. Ethnic and racial disparities in cardiac resynchronization therapy. Heart Rhythm 2009; 6(3):325–331. doi:10.1016/j.hrthm.2008.12.018
  69. Rose EA, Gelijns AC, Moskowitz AJ, et al; Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH) Study Group. Long-term use of a left ventricular assist device for end-stage heart failure. N Engl J Med 2001; 345(20):1435–1443. doi:10.1056/NEJMoa012175
  70. Slaughter MS, Rogers JG, Milano CA, et al; HeartMate II Investigators. Advanced heart failure treated with continuous-flow left ventricular assist device. N Engl J Med 2009; 361(23):2241–2251. doi:10.1056/NEJMoa0909938
  71. Tsiouris A, Brewer RJ, Borgi J, Nemeh H, Paone G, Morgan JA. Continuous-flow left ventricular assist device implantation as a bridge to transplantation or destination therapy: racial disparities in outcomes. J Heart Lung Transplant 2013; 2(3):299–304. doi:10.1016/j.healun.2012.11.017
  72. Stulak JM, Deo S, Cowger J, et al. Do racial and sex disparities exist in clinical characteristics and outcomes for patients undergoing left ventricular assist device implantation? J Heart Lung Transplant 2013; 32(45):S279–S280.
  73. Joyce DL, Conte JV, Russell SD, Joyce LD, Chang DC. Disparities in access to left ventricular assist device therapy. J Surg Res 2009; 152(1):111–117. doi:10.1016/j.jss.2008.02.065
  74. Breathett K, Allen LA, Helmkamp L, et al. Temporal trends in contemporary use of ventricular assist devices by race and ethnicity. Circ Heart Fail 2018; 11(8):e005008. doi:10.1161/CIRCHEARTFAILURE.118.005008
  75. Kilic A, Higgins RS, Whitson BA, Kilic A. Racial disparities in outcomes of adult heart transplantation. Circulation 2015; 131(10):882–889. doi:10.1161/CIRCULATIONAHA.114.011676
  76. Liu V, Bhattacharya J, Weill D, Hlatky MA. Persistent racial disparities in survival after heart transplantation. Circulation 2011; 123(15):1642–1649. doi:10.1161/CIRCULATIONAHA.110.976811
  77. Mahle WT, Kanter KR, Vincent RN. Disparities in outcome for black patients after pediatric heart transplantation. J Pediatr 2005; 147(6):739–743. doi:10.1016/j.jpeds.2005.07.018
  78. Park MH, Tolman DE, Kimball PM. The impact of race and HLA matching on long-term survival following cardiac transplantation. Transplant Proc 1997; 29(1–2):1460–1463. pmid:9123381
  79. Higgins RS, Fishman JA. Disparities in solid organ transplantation for ethnic minorities: facts and solutions. Am J Transplant 2006; 6(11):2556–2562. doi:10.1111/j.1600-6143.2006.01514.x
  80. Taylor AL, Wright JT Jr. Should ethnicity serve as the basis for clinical trial design? Importance of race/ethnicity in clinical trials: lessons from the African-American Heart Failure Trial (A-HeFT), the African-American Study of Kidney Disease and Hypertension (AASK), and the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT). Circulation 2005; 112(23):3654–3660. doi:10.1161/CIRCULATIONAHA.105.540443
  81. Yancy CW. Heart failure in African Americans: a cardiovascular engima. J Card Fail 2000; 6(3):183–186. pmid:10997742
  82. Chobanian AV, Bakris GL, Black HR, et al; National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure; National High Blood Pressure Education Program Coordinating Committee. The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: the JNC 7 report. JAMA 2003; 289(19):2560–2572. doi:10.1001/jama.289.19.2560
  83. Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration cooperative study. N Engl J Med 1986; 314(24):1547–1552. doi:10.1056/NEJM198606123142404
  84. Chen MS Jr, Lara PN, Dang JH, Paterniti DA, Kelly K. Twenty years post-NIH Revitalization Act: enhancing minority participation in clinical trials (EMPaCT): laying the groundwork for improving minority clinical trial accrual: renewing the case for enhancing minority participation in cancer clinical trials. Cancer 2014;120(suppl 7):1091–1096. doi:10.1002/cncr.28575
  85. Geller SE, Koch A, Pellettieri B, Carnes M. Inclusion, analysis, and reporting of sex and race/ethnicity in clinical trials: have we made progress? J Womens Health (Larchmt) 2011; 20(3):315–320. doi:10.1089/jwh.2010.2469
  86. Beta-Blocker Evaluation of Survival Trial Investigators; Eichhorn EJ, Domanski MJ, Krause-Steinrauf H, Bristow MR, Lavori PW. A trial of the beta-blocker bucindolol in patients with advanced chronic heart failure. N Engl J Med 2001; 344(22):1659–1667. doi:10.1056/NEJM200105313442202
  87. Taylor AL, Ziesche S, Yancy C, et al; African-American Heart Failure Trial Investigators. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med 2004; 351(20):2049–2057. doi:10.1056/NEJMoa042934
  88. Corbie-Smith G, Thomas SB, Williams MV, Moody-Ayers S. Attitudes and beliefs of African Americans toward participation in medical research. J Gen Intern Med 1999; 14(9):537–546. pmid:10491242
  89. Swanson GM, Ward AJ. Recruiting minorities into clinical trials: toward a participant-friendly system. J Natl Cancer Inst 1995; 87(23):1747–1759. doi:10.1093/jnci/87.23.1747
  90. Institute of Medicine (US) Committee on Understanding and Eliminating Racial and Ethnic Disparities in Health Care; Smedley BD, Stith AY, Nelson AR, eds. Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care. Washington, DC: National Academies Press (US); 2003. https://www.ncbi.nlm.nih.gov/books/NBK220358/. Accessed May 13, 2019.
  91. Fisher JA, Kalbaugh CA. Challenging assumptions about minority participation in US clinical research. Am J Public Health 2011; 101(12):2217–2222. doi:10.2105/AJPH.2011.300279
  92. Hall WJ, Chapman MV, Lee KM, et al. Implicit racial/ethnic bias among health care professionals and its influence on health care outcomes: a systematic review. Am J Public Health 2015; 105(12):e60–e76. doi:10.2105/AJPH.2015.302903
  93. Biernat M, Manis M. Shifting standards and stereotype-based judgments. J Pers Soc Psychol 1994; 66(1):5–20. pmid:8126651
  94. Chapman EN, Kaatz A, Carnes M. Physicians and implicit bias: how doctors may unwittingly perpetuate health care disparities. J Gen Intern Med 2013; 28(11):1504–1510. doi:10.1007/s11606-013-2441-1
  95. FitzGerald C, Hurst S. Implicit bias in healthcare professionals: a systematic review. BMC Med Ethics 2017; 18(1):19. doi:10.1186/s12910-017-0179-8
  96. van Ryn M, Burke J. The effect of patient race and socio-economic status on physicians’ perceptions of patients. Soc Sci Med 2000; 50(6):813–828. pmid:10695979
  97. Mayo RM, Sherrill WW, Sundareswaran P, Crew L. Attitudes and perceptions of Hispanic patients and health care providers in the treatment of Hispanic patients: a review of the literature. Hisp Health Care Int 2007; 5(2):64–72.
  98. Blair IV, Steiner JF, Havranek EP. Unconscious (implicit) bias and health disparities: where do we go from here? Perm J 2011; 15(2):71–78. pmid:21841929
  99. Green AR, Carney DR, Pallin DJ, et al. Implicit bias among physicians and its prediction of thrombolysis decisions for black and white patients. J Gen Intern Med 2007; 22(9):1231–1238. doi:10.1007/s11606-007-0258-5
  100. Sabin JA, Rivara FP, Greenwald AG. Physician implicit attitudes and stereotypes about race and quality of medical care. Med Care 2008; 46(7):678–685. doi:10.1097/MLR.0b013e3181653d58
  101. Smedley BD, Stith AY, Colburn L, et al; Institute of Medicine. The Right Thing to Do, The Smart Thing to Do: Enhancing Diversity in the Health Professions: Summary of the Symposium on Diversity in Health Professions in Honor of Herbert W. Nickens, MD. Washington, DC: National Academies Press; 2001. https://www.ncbi.nlm.nih.gov/books/NBK223633/. Accessed May 13, 2019.
  102. Chen J, Vargas-Bustamante A, Mortensen K, Ortega AN. Racial and ethnic disparities in health care access and utilization under the Affordable Care Act. Med Care 2016; 54(2):140–146. doi:10.1097/MLR.0000000000000467
  103. Saha S, Freeman M, Toure J, Tippens KM, Weeks C, Ibrahim S. Racial and ethnic disparities in the VA health care system: a systematic review. J Gen Intern Med 2008; 23(5):654–671. doi:10.1007/s11606-008-0521-4
  104. McCormick D, Sayah A, Lokko H, Woolhandler S, Nardin R. Access to care after Massachusetts’ health care reform: a safety net hospital patient survey. J Gen Intern Med 2012; 27(11):1548–1554. doi:10.1007/s11606-012-2173-7
  105. Burgos JL, Yee D, Csordas T, et al. Supporting the minority physician pipeline: providing global health experiences to undergraduate students in the United States-Mexico border region. Med Educ Online 2015; 20:27260. doi:10.3402/meo.v20.27260
  106. Traylor AH, Schmittdiel JA, Uratsu CS, Mangione CM, Subramanian U. The predictors of patient–physician race and ethnic concordance: a medical facility fixed-effects approach. Health Serv Res 2010; 45(3):792–805. doi:10.1111/j.1475-6773.2010.01086.x
  107. LaVeist TA, Nuru-Jeter A. Is doctor-patient race concordance associated with greater satisfaction with care? J Health Soc Behav 2002; 43(3):296–306. pmid:12467254
  108. Marrast LM, Zallman L, Woolhandler S, Bor DH, McCormick D. Minority physicians’ role in the care of underserved patients: diversifying the physician workforce may be key in addressing health disparities. JAMA Intern Med 2014; 174(2):289–291. doi:10.1001/jamainternmed.2013.12756
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Page Number
621-632
Page Number
621-632
Publications
Publications
Topics
Article Type
Display Headline
Disparities in cardiovascular care: Past, present, and solutions
Display Headline
Disparities in cardiovascular care: Past, present, and solutions
Legacy Keywords
disparities, cardiovascular care, heart care, racism, bias, race, African American, heart attack, stroke, hypertension, black, white, smoking, American Indian, Alaska Native, exercise, inactivity, sedentary lifestyle, nutrition, obesity, diabetes, coronary artery disease, heart failure, transplant, research study, minority physician, Tuskegee syphilis experiment, Quentin Youmans, Lindsey Hastings-Spaine, Oluseyi Princewill, Titilayo Shobayo, Ike Okwuosa
Legacy Keywords
disparities, cardiovascular care, heart care, racism, bias, race, African American, heart attack, stroke, hypertension, black, white, smoking, American Indian, Alaska Native, exercise, inactivity, sedentary lifestyle, nutrition, obesity, diabetes, coronary artery disease, heart failure, transplant, research study, minority physician, Tuskegee syphilis experiment, Quentin Youmans, Lindsey Hastings-Spaine, Oluseyi Princewill, Titilayo Shobayo, Ike Okwuosa
Sections
Inside the Article

KEY POINTS

  • Although avoidable deaths from heart disease, stroke, and hypertensive disease have declined overall, African Americans still have a higher mortality rate than other racial and ethnic groups.
  • The prevalence of modifiable risk factors for cardiovascular disease is higher in African Americans than in the general US population.
  • Disparities in care exist and may persist even with equal access to care.
  • Since 1993, studies funded by the National Institutes of Health must include minorities that were historically underrepresented in clinical research trials.
  • Solutions to disparities will need to eliminate healthcare bias, increase patient access, and increase diversity and inclusion in the physician work force.
  • Cardiovascular disease makes no distinction in race, sex, age, or socioeconomic status, and neither should the medical community.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/22/2019 - 09:00
Un-Gate On Date
Thu, 08/22/2019 - 09:00
Use ProPublica
CFC Schedule Remove Status
Thu, 08/22/2019 - 09:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Hemorrhoids: A range of treatments

Article Type
Changed
Tue, 09/03/2019 - 10:59
Display Headline
Hemorrhoids: A range of treatments

Aspects of modern life that may promote hemorrhoids include increased consumption of processed foods, a sedentary lifestyle, and using cell phones while defecating, which translates to much more time spent on the toilet.

Hemorrhoids accounted for more than 3.5 million US outpatient visits in 2010, and they were the third leading cause of hospital admissions related to gastrointestinal disease.1

Here, we review the process for diagnosing and grading hemorrhoids, as well as for selecting the appropriate medical or surgical treatment based on the most recent clinical evidence.

DIAGNOSING AND CLASSIFYING HEMORRHOIDS

Hemorrhoids are the distal prolapse of the arteriovenous bundle, muscle fibers, and surrounding connective tissue as an envelope below the dentate line in the anal canal. They usually present with painless rectal bleeding.2

The diagnosis of hemorrhoids relies on the history and physical examination rather than on laboratory testing or imaging studies. Typically, the presenting symptom is painless rectal bleeding associated with bowel movements, usually appearing as bright red blood on the toilet paper or coating the stool. Severe itching and anal discomfort are also common, especially with chronic hemorrhoids.

Detailed patient history

A detailed patient history is important. It should include the extent, severity, and duration of symptoms, frequency of bowel movements, associated symptoms (eg, constipation, fecal incontinence), daily dietary habits, and details of bowel movements (eg, time spent during each bowel movement and concomitant cell phone use).3

Regarding bowel habits, some patients experience lifelong constipation or diarrhea. Therefore, what a patient considers a normal bowel habit may not be normal and should be investigated.4 Also, it is important to exclude external thrombosed hemorrhoids, anal fissure, anal abscess, and Crohn disease.5

Physical examination

A digital rectal examination is the second step. During the examination, look for skin tags, sphincter tone, perianal hygiene, and synchronous anal lesions.3 Of note, the Valsalva maneuver can be performed during the digital rectal examination.

Red flags for colorectal cancer on the digital rectal examination include a mass with or without presence of hemorrhoidal sacs and a bleeding source above the level of internal hemorrhoids.

cengiz_hemorrhoidtreatment_f1.jpg
%3Cp%3EFigure%201.%20Patient%20with%20Crohn%20disease.%20Note%20the%20fistula%20orifices%20and%20the%20skin%20tag.%3C%2Fp%3E
Patients with recurrent abscesses, fistulas, or skin tags (especially cauliflower-type skin tags) should be investigated for Crohn disease (Figure 1).

Endoscopy

Since rectal bleeding can be a sign of several diseases, including colorectal cancer, it is important to review any previous endoscopic results. Patients at high risk of colon cancer should undergo rigid proctoscopy, flexible sigmoidoscopy, or colonoscopy.3,4 In our practice, we recommend endoscopic evaluation for patients over age 40 with rectal bleeding, especially if they have a family history of colorectal cancer.

External or internal (grades I–IV)

Hemorrhoids can be categorized as either external or internal.

External hemorrhoids are distinguished by their outer covering with perianal skin and anoderm and their location inferior to the dentate line. They are painful if the hemorrhoidal sac is occluded by a thrombotic clot.

Internal hemorrhoids are above the dentate line and covered with rectal columnar and transitional mucosa. They are further graded on a 4-point scale3:

  • Grade I—Visible hemorrhoids that do not prolapse
  • Grade II—Hemorrhoids that prolapse during the Valsalva maneuver but spontaneously reduce
  • Grade III—Hemorrhoids that prolapse during the Valsalva maneuver and need manual reduction
  • Grade IV—Nonreducible hemorrhoids.

A RANGE OF TREATMENTS

In choosing the treatment for hemorrhoids, one should consider the disease grade and severity, its impact on the quality of life, the degree of pain it causes, the patient’s likelihood of adhering to treatment, and the patient’s personal preference.

Regardless of severity, treatment almost always starts with a high-fiber diet and other lifestyle modifications that include bowel movement behaviors. This requires practitioners to spend significant time on patient education regardless of the type or the severity of the disease.

Treatments can be grouped in 3 categories: conservative, office-based, and surgical. Practitioners should thoroughly discuss the options with the patient, emphasizing the pros and cons of each.

 

 

CONSERVATIVE MEASURES

Conservative measures are aimed at softening the stool, relieving pain, and correcting bad toileting habits. In most cases, the primary precipitating factor is lifestyle, and unless patients change it, they are more likely to have recurrent symptoms in the long term.

No phone in the bathroom

People take their phones into the bathroom, and this habit is blamed for increasing the time on the toilet and leading to increased pressure on the anal region and straining during defecation. Some research points to a direct correlation between the time spent on the toilet and hemorrhoidal disease, although the exact cause-and-effect relationship with cell phone use has not been determined. In general, spending excessive time on the commode, including reading, should be discouraged.

Less time in the bathroom

Johannsson et al6 reported that patients with hemorrhoids spent more time on the toilet and had to strain harder and more often than controls in the community and hospital.

Garg and Singh7 and Garg8 use the mnemonic “TONE” for appropriate defecation habits:

  • Three minutes during defecation
  • Once-daily defecation
  • No straining and no cell phone use during defecation
  • Enough fiber.

More fiber

Fiber draws water into the lumen of the colon, increasing the water content of the stool. Recommended daily fiber intake is about 28 g for women and 38 g for men.9 This high level of intake is hard to achieve without supplements for someone who consumes a classic American diet with a lot of fast food.

Fiber supplements are strongly recommended in the American Society of Colon and Rectal Surgeons (ASCRS) practice guidelines3 based on a Cochrane review.10 In this meta-analysis, with fiber supplements, the relative risk of persisting or nonimproving symptoms was 0.53 (95% confidence interval [CI] 0.38–0.73) and the relative risk of bleeding was 0.50 (95% CI 028–0.89). Psyllium husk is an inexpensive bulk-forming fiber supplement; the optimal daily dosage is not known.

We recommend at least 28 g of daily fiber intake for women and 38 g for men, for which psyllium husk can be used to complement the diet.

Laxatives for some

Laxatives such as docusate are used to change the stool consistency when there is an organic bowel problem rather than a dietary issue. They can be used as a complementary treatment to enhance the effect of the fiber treatment.

Other measures

Topical anesthesia (eg, 5% lidocaine) is commonly used to treat pain from low-grade lesions, but no reliable data have been published. As most cases of hemorrhoids tend to progress over time, one should not expect long-term improvement with topical anesthesia. Nevertheless, it can be used as an ancillary treatment in select cases when short-term improvement is the main goal, and we recommend it based on our own experience.

Hygiene. Bidet use or cleaning the perianal area with water is recommended.

Phlebotonics contain a variety of ingredients including natural plant extracts such as flavonoids and synthetic products. Even though the exact mechanism of action is not known, phlebotonics are thought to increase venous and lymphatic drainage, normalize capillary permeability, and decrease inflammation in the hemorrhoidal cushions.4,11–13

In a Cochrane review of 24 randomized controlled trials, Perera et al14 found that phlebotonics improved the outcomes of:

  • Bleeding (odds ratio [OR] 0.21, number needed to treat [NNT] 4.8, P = .0002)
  • Pruritus (OR 0.23, NNT 9.1, P = .02)
  • Discharge or leakage (OR 0.12, NNT 5, P = .0008)
  • Overall symptoms (OR 15.99, NNT 2.7, P < .00001). Overall symptoms were also improved in the subgroup of pregnant patients.

Although phlebotonics give better results than placebo in the short-term management of hemorrhoids, there is a paucity of long-term data. Thus, the ASCRS clinical practice guidelines gives the regular use of these agents only a weak recommendation.3

Flavonoids (diosmin, hesperidin, rutoside), in a meta-analysis vs placebo in 1,514 patients, showed a beneficial response in terms of bleeding (relative risk [RR] 0.33), pruritus (relative risk [RR] 0.65), and recurrences (RR 0.53).15

Although Preparation H is commonly used as an over-the-counter medication, there are no good data on it, and it is not considered a phlebotonic.

 

 

OFFICE-BASED TREATMENTS

Office-based treatments—rubber band ligation, infrared photocoagulation, and sclerotherapy—are commonly used for grade I, II, and III hemorrhoids that have not responded to conservative management. The primary goal of these treatments is to decrease blood flow into the hemorrhoidal sac.

Even though office-based treatments are highly effective and major complications are uncommon, recurrence rates can be high, requiring patients to undergo additional treatments. Moreover, septic complications can occur, so patients should be closely observed for fever and urinary problems. Pain is a common symptom after office-based treatments, and bleeding may also occur.

The ASCRS guidelines strongly recommend office-based treatments for patients with grade I and II hemorrhoids, and for some with grade III hemorrhoids.3

Rubber band ligation

cengiz_hemorrhoidtreatment_f2.jpg
%3Cp%3EFigure%202.%20In%20rubber%20band%20ligation%2C%20an%20internal%20hemorrhoid%20is%20grasped%20with%20a%20forceps%20(A)%20and%20drawn%20into%20the%20cylinder%20of%20the%20ligator%20(B).%20A%20band%20is%20deployed%20around%20the%20base%20of%20the%20hemorrhoid%20(C)%2C%20cutting%20off%20its%20blood%20supply%20and%20causing%20it%20to%20fall%20off%20within%20a%20few%20days.%3C%2Fp%3E
Ligating the apex of the hemorrhoidal cushion stops the arterial flow and causes the hemorrhoidal tissue to undergo necrosis (Figure 2). The ligation is performed above the dentate line, where the sensory nerve fibers differ from those found below the line; therefore, the operation causes less pain than one would expect. One or more hemorrhoidal cushions can be ligated at the same time, although increased pain, bleeding, and vasomotor reactions have been reported with multiple banding during a single procedure.16,17

Iyer et al18 reported that patients on warfarin therapy had up to a 9 times higher risk of postprocedural bleeding, and patients on aspirin had a risk up to 3 times higher. Therefore, whether patients on anticoagulant therapy should undergo this procedure is unclear.

A Cochrane database review19 found this technique effective for hemorrhoid grades I through III, although some patients with grade III hemorrhoids may benefit more from excisional hemorrhoidectomy, which is associated with a lower recurrence rate than rubber band ligation.

Brown et al20 performed a randomized controlled trial comparing hemorrhoidal artery ligation and rubber band ligation for symptomatic hemorrhoids in 372 patients with grade II and III hemorrhoids. Postprocedural pain scores on days 1 and 7 were significantly lower with rubber band ligation, but recurrences were more common (49% vs 30%, P = .0005, respectively).

Overall, rubber band ligation is an excellent option for grade II hemorrhoids, as it is easy to perform, is associated with low pain scores, and can be used to treat recurrences.

Infrared photocoagulation

In this procedure, an infrared probe produces heat to induce coagulation, fibrosis, and ultimately necrosis of the protruding tissue in the hemorrhoidal cushions.21 Even though its use was initially directed at grade I and II hemorrhoids, recent reports showed acceptable results for grades III and IV.22,23 A randomized controlled trial comparing infrared photocoagulation and rubber band ligation in 94 patients found that both procedures were well accepted and highly effective; however, patients had better pain scores with photocoagulation in the first 24 hours after the procedure (P < .05).24

Sclerotherapy

cengiz_hemorrhoidtreatment_f3.jpg
%3Cp%3EFigure%203.%20Sclerotherapy%20involves%20injecting%20an%20irritating%20solution%20into%20the%20hemorrhoid%2C%20reducing%20its%20blood%20supply%20and%20causing%20it%20to%20shrink.%3C%2Fp%3E
Sclerotherapy involves injection of a sclerotic agent into the submucosa of the hemorrhoidal sac (Figure 3), which causes an inflammatory reaction and eventually forms fibrotic tissue that stops the blood flow to the hemorrhoid. Many sclerotic agents are available, including 5% phenol in almond or vegetable oil, quinine, ethanolamine, and hypertonic saline.21

The injection can cause prostatic abscess and sepsis, although this is rare.25 Nevertheless, high fever and postprocedural pain should be carefully evaluated.

There have been few randomized trials of sclerotherapy, but success rates so far have been higher for grade I hemorrhoids than for grades II and III.26–28 It is the preferred method for patients who have bleeding abnormalities caused by medications or other diseases (eg, cirrhosis).

 

 

SURGERY

Although nonsurgical treatments have substantially improved, surgery is the most effective and strongly recommended treatment for patients with high-grade internal hemorrhoids (grades III and IV), external and mixed hemorrhoids, and recurrent hemorrhoids.

The most popular surgical options are open or closed hemorrhoidectomy, stapled hemorrhoidopexy, and Doppler-guided hemorrhoidal artery ligation. Each has different success rates and different complication profiles, which need to be discussed with the patient.

Overall, surgery is associated with more adverse effects than office-based treatments or medical management. Postoperative pain is the most common complaint, but anal stricture (rare) or incontinence may occur due to excessive tissue excision and damage to the sphincter muscles. These can be avoided by maintaining the normal anoderm between excisions, by not excising all hemorrhoid sacs at once if the patient has extensive lesions, and by performing a careful dissection in the submucosal plane.

Patients with profuse bleeding or an underlying bleeding abnormality are best managed with surgical approaches performed in an operating room.

Excisional surgical hemorrhoidectomy

Excision of the hemorrhoidal sac, the most conventional surgical technique, is generally reserved for prolapsing disease. The recurrence rate after excisional hemorrhoidectomy is significantly lower than with any other approach.29

Excisional hemorrhoidectomy can be performed using either an open approach, in which the edges of the mucosal defect are not reapproximated, or a closed approach, in which they are. In a systematic review, Bhatti et al30 compared open vs closed techniques and found that the closed technique resulted in less postoperative pain, better wound healing, and less bleeding. Rates of recurrence, postoperative complications, and surgical site infection and lengths of stay were comparable with either procedure.

Overall, excisional hemorrhoidectomy is associated with higher pain scores than any other surgical method.29 Recently, the use of electrodiathermy energy devices, also described as electrosurgical vessel-sealing devices, have further improved overall patient satisfaction.31

Multiple painful hemorrhoidal sacs require a careful surgical approach, as extensive resection may cause widespread fibrosis and stricture. As with anal stricture, fecal incontinence can be prevented by careful dissection. However, already existing incontinence is not a contraindication for the surgery.

Doppler-guided hemorrhoidal artery ligation

Doppler-guided hemorrhoidal artery ligation involves using a Doppler probe to find and ligate individual hemorrhoidal arteries. Additionally, mucopexy (transanal rectoanal repair) is performed to relocate the prolapsing tissue. Avital et al32 reported that at 1 year after this procedure, recurrence rates were 5.3% for grade II hemorrhoids and 13% for grade III hemorrhoids. At 5 years, recurrence rates were 12% for grade II and 31% for grade III.

To date, this procedure appears to be suitable for grade I, II, and III hemorrhoids, especially for grade II, but more studies are needed to prove its efficacy and recurrence rates for more advanced lesions. Although this technique has a high morbidity rate (18%), primarily pain or tenesmus, it causes less postoperative pain than other surgical methods.33 Overall, it has the potential to become a favored treatment.

Stapled hemorrhoidopexy

cengiz_hemorrhoidtreatment_f4.jpg
%3Cp%3EFigure%204.%20In%20stapled%20hemorrhoidopexy%2C%20a%20special%20tool%20is%20inserted%20(A).%20Excess%20tissue%20is%20excised%20(B)%2C%20and%20the%20remaining%20tissue%20is%20drawn%20up%20to%20its%20normal%20position%20and%20fastened%20in%20place%2C%20yielding%20the%20result%20pictured%20in%20panel%20C.%3C%2Fp%3E
In this procedure, the prolapsing part of the internal hemorrhoidal cushion is moved upward by stapling the rectal mucosa just above the hemorrhoid (Figure 4). This is not an option for patients with thrombosed internal hemorrhoids or with external hemorrhoids.

Although pain scores are lower with stapled hemorrhoidopexy than with excisional hemorrhoidectomy, this procedure is not superior in terms of recurrences.34,35 Also, practitioners should be careful about specific complications of stapled hemorrhoidopexy, such as rectovaginal fistula, anal stenosis, or sphincter injuries. These specific complications should be clearly explained to patients, and necessary information should be given to patients upon discharge. The primary care physician should also be careful about fistulas and stenoses in this particular patient population.

 

 

NO ‘BEST’ TREATMENT

There is no best treatment for hemorrhoids. Every patient is different, and the physician and patient need to understand each other’s expectations, weigh the risks and benefits, and arrive at a mutual decision. A good patient-doctor relationship is essential.

cengiz_hemorrhoidtreatment_f5.jpg
%3Cp%3EFigure%205.%20Algorithm%20for%20hemorrhoid%20management.%3C%2Fp%3E
A thorough history and physical examination will enable the practitioner to understand the patient’s problem (Figure 5).

Given the variety of available treatments, head-to-head comparisons are difficult. Moreover, the efficacy and applicability of each technique changes with the grade of the lesion or lesions and the skill of the practitioner. Lacking comprehensive studies comparing conservative, office-based, and surgical management, no decisive statements can be made based on current evidence.

Patients with compounding conditions

Pregnant patients often develop hemorrhoids as intra-abdominal pressure increases, particularly during the third trimester.36 Also, acute episodes of pain and bleeding are common in pregnant women with preexisting hemorrhoids.

Conservative treatment is the main approach in pregnant patients because most hemorrhoids regress after childbirth. This includes increased dietary fiber, stool softeners, and sitz baths, which are safe to use for external hemorrhoids. Any office-based or surgical intervention should be postponed until after childbirth, if possible. Kegel exercises and lying on the left side are also recommended to relieve symptoms. In cases of severe bleeding, anal packing appears to be useful.

Immunosuppressed patients and those on anticoagulant therapy are more prone to serious complications such as sepsis and profuse bleeding. Thus, conservative management should be used in these patients as well. Injection sclerotherapy may be beneficial, as it has been shown to decrease bleeding. Of note, patients on immunosuppressive agents should stop taking them and start taking an antibiotic, and patients on anticoagulant or antiplatelet medications should be instructed to stop them 1 week before any intervention.

Crohn disease. Some patients with Crohn disease may have hemorrhoids, though this is rare. Eglinton et al,37 in a series of 715 patients with Crohn disease, reported that 190 (26.6%) had symptomatic perianal disease. Of these, only 3 (1.6%) had hemorrhoids. Treatment is always conservative and directed at the Crohn disease rather than the hemorrhoids.

Patients with portal hypertension (eg, due to cirrhosis) are prone to have anorectal varices that may resemble hemorrhoids. Anorectal varices can be treated with vascular ligation, whereas sclerotherapy is the preferred method for hemorrhoids in this group, in whom coagulopathy is common.

TAKE-HOME MESSAGES

Hemorrhoidal disease is common in the United States, and with our diet and lifestyle, the incidence is likely to increase. (A national survey found that overall dietary quality improved modestly in children and adolescents in the United States from 1999 to 2012 but remained far below optimal.38) Practitioners need to carefully assess hemorrhoidal symptoms and complete any necessary screening tests before establishing a diagnosis. This helps to avoid missing any underlying disease.

Fiber supplements along with dietary and lifestyle changes constitute the baseline of the management regardless of the disease grade. Office-based interventions are beneficial for grade I and II hemorrhoids and for some grade III hemorrhoids. Repeated interventions can increase the success rate. In patients with high-grade, symptomatic hemorrhoids, surgical hemorrhoidectomy is the most effective modality with the lowest recurrence rates, although it causes more pain than conservative methods.

References
  1. Peery AF, Crockett SD, Barritt AS, et al. Burden of gastrointestinal, liver, and pancreatic diseases in the United States. Gastroenterology 2015; 149(7):1731–1741.e3. doi:10.1053/j.gastro.2015.08.045
  2. Thomson WH. The nature and cause of haemorrhoids. Proc R Soc Med 1975; 68(9):574–575. pmid:1197343
  3. Davis BR, Lee-Kong SA, Migaly J, Feingold DL, Steele SR. The American Society of Colon and Rectal Surgeons clinical practice guidelines for the management of hemorrhoids. Dis Colon Rectum 2018; 61(3):284–292. doi:10.1097/DCR.0000000000001030
  4. Lohsiriwat V. Treatment of hemorrhoids: a coloproctologist’s view. World J Gastroenterol 2015; 21(31):9245–9252. doi:10.3748/wjg.v21.i31.9245
  5. Wolf AMD, Fontham ETH, Church TR, et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin 2018; 68(4):250–281. doi:10.3322/caac.21457
  6. Johannsson HO, Graf W, Påhlman L. Bowel habits in hemorrhoid patients and normal subjects. Am J Gastroenterol 2005; 100(2):401–406. doi:10.1111/j.1572-0241.2005.40195.x
  7. Garg P, Singh P. Adequate dietary fiber supplement and TONE can help avoid surgery in most patients with advanced hemorrhoids. Minerva Gastroenterol Dietol 2017; 63(2):92–96. doi:10.23736/S1121-421X.17.02364-9
  8. Garg P. Conservative treatment of hemorrhoids deserves more attention in guidelines and clinical practice [letter]. Dis Colon Rectum 2018; 61(7):e348. doi:10.1097/DCR.0000000000001127
  9. Rakinic J, Poola VP. Hemorrhoids and fistulas: new solutions to old problems. Curr Probl Surg 2014; 51(3):98–137. doi:10.1067/j.cpsurg.2013.11.002
  10. Alonso-Coello P, Guyatt G, Heels-Ansdell D, et al. Laxatives for the treatment of hemorrhoids. Cochrane Database Syst Rev 2005; (4):CD004649. doi:10.1002/14651858.CD004649.pub2
  11. Struckmann JR. Clinical efficacy of micronized purified flavonoid fraction: an overview. J Vasc Res 1999; 36(suppl 1):37–41. doi:10.1159/000054072
  12. Shoab SS, Porter J, Scurr JH, Coleridge-Smith PD. Endothelial activation response to oral micronised flavonoid therapy in patients with chronic venous disease—a prospective study. Eur J Vasc Endovasc Surg 1999; 17(4):313–318. doi:10.1053/ejvs.1998.0751
  13. Meyer OC. Safety and security of Daflon 500 mg in venous insufficiency and in hemorrhoidal disease. Angiology 1994; 45(6 pt 2):579–584. pmid:8203791
  14. Perera N, Liolitsa D, Iype S, et al. Phlebotonics for haemorrhoids. Cochrane Database Syst Rev 2012;(8):CD004322. doi:10.1002/14651858.CD004322.pub3
  15. Alonso-Coello P, Zhou Q, Martinez-Zapata MJ, et al. Meta-analysis of flavonoids for the treatment of haemorrhoids. Br J Surg 2006; 93(8):909–920. doi:10.1002/bjs.5378
  16. Lee HH, Spencer RJ, Beart RW Jr. Multiple hemorrhoidal bandings in a single session. Dis Colon Rectum 1994; 37(1):37–41. pmid:8287745
  17. Law WL, Chu KW. Triple rubber band ligation for hemorrhoids: prospective, randomized trial of use of local anesthetic injection. Dis Colon Rectum 1999; 42(3):363–366. pmid:10223757
  18. Iyer VS, Shrier I, Gordon PH. Long-term outcome of rubber band ligation for symptomatic primary and recurrent internal hemorrhoids. Dis Colon Rectum 2004; 47(8):1364–1370. pmid:15484351
  19. Shanmugam V, Thaha MA, Rabindranath KS, Campbell KL, Steele RJ, Loudon MA. Rubber band ligation versus excisional haemorrhoidectomy for haemorrhoids. Cochrane Database Syst Rev 2005; (3):CD005034. doi:10.1002/14651858.CD005034.pub2
  20. Brown SR, Tiernan JP, Watson AJM, et al; HubBLe Study team. Haemorrhoidal artery ligation versus rubber band ligation for the management of symptomatic second-degree and third-degree haemorrhoids (HubBLe): a multicentre, open-label, randomised controlled trial. Lancet 2016; 388(10042):356–364. doi:10.1016/S0140-6736(16)30584-0
  21. ASGE Technology Committee; Siddiqui UD, Barth BA, Banerjee S, et al. Devices for the endoscopic treatment of hemorrhoids. Gastrointest Endosc 2014; 79(1):8–14. doi:10.1016/j.gie.2013.07.021
  22. Ahmad A, Kant R, Gupta A. Comparative analysis of Doppler guided hemorrhoidal artery ligation (DG-HAL) & infrared coagulation (IRC) in management of hemorrhoids. Indian J Surg 2013; 75(4):274–277. doi:10.1007/s12262-012-0444-5
  23. Poen AC, Felt-Bersma RJ, Cuesta MA, Devillé W, Meuwissen SG. A randomized controlled trial of rubber band ligation versus infra-red coagulation in the treatment of internal haemorrhoids. Eur J Gastroenterol Hepatol 2000; 12(5):535–539. pmid:10833097
  24. Marques CF, Nahas SC, Nahas CS, Sobrado CW Jr, Habr-Gama A, Kiss DR. Early results of the treatment of internal hemorrhoid disease by infrared coagulation and elastic banding: a prospective randomized cross-over trial. Tech Coloproctol 2006; 10(4):312–317. doi:10.1007/s10151-006-0299-5
  25. Madoff RD, Fleshman JW; Clinical Practice Committee, American Gastroenterological Association. American Gastroenterological Association technical review on the diagnosis and treatment of hemorrhoids. Gastroenterology 2004; 126(5):1463–1473. pmid:15131807
  26. Yano T, Yano K. Comparison of injection sclerotherapy between 5% phenol in almond oil and aluminum potassium sulfate and tannic acid for grade 3 hemorrhoids. Ann Coloproctol 2015; 31(3):103–105. doi:10.3393/ac.2015.31.3.103
  27. Kanellos I, Goulimaris I, Vakalis I, Dadoukis I. Long-term evaluation of sclerotherapy for haemorrhoids. A prospective study. Int J Surg Investig 2000; 2(4):295–298. pmid:12678531
  28. Moser KH, Mosch C, Walgenbach M, et al. Efficacy and safety of sclerotherapy with polidocanol foam in comparison with fluid sclerosant in the treatment of first-grade haemorrhoidal disease: a randomised, controlled, single-blind, multicentre trial. Int J Colorectal Dis 2013; 28(10):1439–1447. doi:10.1007/s00384-013-1729-2
  29. MacRae HM, McLeod RS. Comparison of hemorrhoidal treatments: a meta-analysis. Can J Surg 1997; 40(1):14–7. pmid:9030078
  30. Bhatti MI, Sajid MS, Baig MK. Milligan-Morgan (open) versus Ferguson haemorrhoidectomy (closed): a systematic review and meta-analysis of published randomized, controlled trials. World J Surg 2016; 40(6):1509–1519. doi:10.1007/s00268-016-3419-z
  31. Nienhuijs S, de Hingh I. Conventional versus LigaSure hemorrhoidectomy for patients with symptomatic hemorrhoids. Cochrane Database Syst Rev 2009; (1):CD006761. doi:10.1002/14651858.CD006761.pub2
  32. Avital S, Inbar R, Karin E, Greenberg R. Five-year follow-up of Doppler-guided hemorrhoidal artery ligation. Tech Coloproctol 2012; 16(1):61–65. doi:10.1007/s10151-011-0801-6
  33. Ratto C, Parello A, Veronese E, et al. Doppler-guided transanal haemorrhoidal dearterialization for haemorrhoids: results from a multicentre trial. Colorectal Dis 2015; 17(1):010–019. doi:10.1111/codi.12779
  34. Senagore AJ, Singer M, Abcarian H, et al; Procedure for Prolapse and Hemmorrhoids (PPH) Multicenter Study Group. A prospective, randomized, controlled multicenter trial comparing stapled hemorrhoidopexy and Ferguson hemorrhoidectomy: perioperative and one-year results. Dis Colon Rectum 2004; 47(11):1824–1836. pmid:15622574
  35. Jayaraman S, Colquhoun PH, Malthaner RA. Stapled versus conventional surgery for hemorrhoids. Cochrane Database Syst Rev 2006; (4):CD005393.
  36. Poskus T, Buzinskiene D, Drasutiene G, et al. Haemorrhoids and anal fissures during pregnancy and after childbirth: a prospective cohort study. BJOG 2014; 121(13):1666–1671. doi:10.1111/1471-0528.12838
  37. Eglinton TW, Barclay ML, Gearry RB, Frizelle FA. The spectrum of perianal Crohn’s disease in a population-based cohort. Dis Colon Rectum 2012; 55(7):773–777. doi:10.1097/DCR.0b013e31825228b0
  38. Gu X, Tucker KL. Dietary quality of the US child and adolescent population: trends from 1999 to 2012 and associations with the use of federal nutrition assistance programs. Am J Clin Nutr 2017; 105(1):194–202. doi:10.3945/ajcn.116.135095
Article PDF
Author and Disclosure Information

Turgut Bora Cengiz, MD
Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic

Emre Gorgun, MD, FACS, FASCRS
Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Emre Gorgun, MD, FACS, FASCRS, Department of Colorectal Surgery, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; gorgune@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 86(9)
Publications
Topics
Page Number
612-620
Legacy Keywords
hemorrhoids, rectal bleeding, fiber, bowel movement, Crohn disease, colorectal cancer, endoscopy, sigmoidoscopy, colonoscopy, laxative, psyllium, phlebotonic, rubber band ligation, sclerotherapy, infrared photocoagulation, excisional surgical hemorrhoidectomy, hemorrhoidal artery ligation, stapled hemorrhoidopexy, portal hypertension, Turgut Bora Cengiz, Emre Gorgun
Sections
Author and Disclosure Information

Turgut Bora Cengiz, MD
Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic

Emre Gorgun, MD, FACS, FASCRS
Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Emre Gorgun, MD, FACS, FASCRS, Department of Colorectal Surgery, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; gorgune@ccf.org

Author and Disclosure Information

Turgut Bora Cengiz, MD
Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic

Emre Gorgun, MD, FACS, FASCRS
Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Emre Gorgun, MD, FACS, FASCRS, Department of Colorectal Surgery, A30, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; gorgune@ccf.org

Article PDF
Article PDF
Related Articles

Aspects of modern life that may promote hemorrhoids include increased consumption of processed foods, a sedentary lifestyle, and using cell phones while defecating, which translates to much more time spent on the toilet.

Hemorrhoids accounted for more than 3.5 million US outpatient visits in 2010, and they were the third leading cause of hospital admissions related to gastrointestinal disease.1

Here, we review the process for diagnosing and grading hemorrhoids, as well as for selecting the appropriate medical or surgical treatment based on the most recent clinical evidence.

DIAGNOSING AND CLASSIFYING HEMORRHOIDS

Hemorrhoids are the distal prolapse of the arteriovenous bundle, muscle fibers, and surrounding connective tissue as an envelope below the dentate line in the anal canal. They usually present with painless rectal bleeding.2

The diagnosis of hemorrhoids relies on the history and physical examination rather than on laboratory testing or imaging studies. Typically, the presenting symptom is painless rectal bleeding associated with bowel movements, usually appearing as bright red blood on the toilet paper or coating the stool. Severe itching and anal discomfort are also common, especially with chronic hemorrhoids.

Detailed patient history

A detailed patient history is important. It should include the extent, severity, and duration of symptoms, frequency of bowel movements, associated symptoms (eg, constipation, fecal incontinence), daily dietary habits, and details of bowel movements (eg, time spent during each bowel movement and concomitant cell phone use).3

Regarding bowel habits, some patients experience lifelong constipation or diarrhea. Therefore, what a patient considers a normal bowel habit may not be normal and should be investigated.4 Also, it is important to exclude external thrombosed hemorrhoids, anal fissure, anal abscess, and Crohn disease.5

Physical examination

A digital rectal examination is the second step. During the examination, look for skin tags, sphincter tone, perianal hygiene, and synchronous anal lesions.3 Of note, the Valsalva maneuver can be performed during the digital rectal examination.

Red flags for colorectal cancer on the digital rectal examination include a mass with or without presence of hemorrhoidal sacs and a bleeding source above the level of internal hemorrhoids.

cengiz_hemorrhoidtreatment_f1.jpg
%3Cp%3EFigure%201.%20Patient%20with%20Crohn%20disease.%20Note%20the%20fistula%20orifices%20and%20the%20skin%20tag.%3C%2Fp%3E
Patients with recurrent abscesses, fistulas, or skin tags (especially cauliflower-type skin tags) should be investigated for Crohn disease (Figure 1).

Endoscopy

Since rectal bleeding can be a sign of several diseases, including colorectal cancer, it is important to review any previous endoscopic results. Patients at high risk of colon cancer should undergo rigid proctoscopy, flexible sigmoidoscopy, or colonoscopy.3,4 In our practice, we recommend endoscopic evaluation for patients over age 40 with rectal bleeding, especially if they have a family history of colorectal cancer.

External or internal (grades I–IV)

Hemorrhoids can be categorized as either external or internal.

External hemorrhoids are distinguished by their outer covering with perianal skin and anoderm and their location inferior to the dentate line. They are painful if the hemorrhoidal sac is occluded by a thrombotic clot.

Internal hemorrhoids are above the dentate line and covered with rectal columnar and transitional mucosa. They are further graded on a 4-point scale3:

  • Grade I—Visible hemorrhoids that do not prolapse
  • Grade II—Hemorrhoids that prolapse during the Valsalva maneuver but spontaneously reduce
  • Grade III—Hemorrhoids that prolapse during the Valsalva maneuver and need manual reduction
  • Grade IV—Nonreducible hemorrhoids.

A RANGE OF TREATMENTS

In choosing the treatment for hemorrhoids, one should consider the disease grade and severity, its impact on the quality of life, the degree of pain it causes, the patient’s likelihood of adhering to treatment, and the patient’s personal preference.

Regardless of severity, treatment almost always starts with a high-fiber diet and other lifestyle modifications that include bowel movement behaviors. This requires practitioners to spend significant time on patient education regardless of the type or the severity of the disease.

Treatments can be grouped in 3 categories: conservative, office-based, and surgical. Practitioners should thoroughly discuss the options with the patient, emphasizing the pros and cons of each.

 

 

CONSERVATIVE MEASURES

Conservative measures are aimed at softening the stool, relieving pain, and correcting bad toileting habits. In most cases, the primary precipitating factor is lifestyle, and unless patients change it, they are more likely to have recurrent symptoms in the long term.

No phone in the bathroom

People take their phones into the bathroom, and this habit is blamed for increasing the time on the toilet and leading to increased pressure on the anal region and straining during defecation. Some research points to a direct correlation between the time spent on the toilet and hemorrhoidal disease, although the exact cause-and-effect relationship with cell phone use has not been determined. In general, spending excessive time on the commode, including reading, should be discouraged.

Less time in the bathroom

Johannsson et al6 reported that patients with hemorrhoids spent more time on the toilet and had to strain harder and more often than controls in the community and hospital.

Garg and Singh7 and Garg8 use the mnemonic “TONE” for appropriate defecation habits:

  • Three minutes during defecation
  • Once-daily defecation
  • No straining and no cell phone use during defecation
  • Enough fiber.

More fiber

Fiber draws water into the lumen of the colon, increasing the water content of the stool. Recommended daily fiber intake is about 28 g for women and 38 g for men.9 This high level of intake is hard to achieve without supplements for someone who consumes a classic American diet with a lot of fast food.

Fiber supplements are strongly recommended in the American Society of Colon and Rectal Surgeons (ASCRS) practice guidelines3 based on a Cochrane review.10 In this meta-analysis, with fiber supplements, the relative risk of persisting or nonimproving symptoms was 0.53 (95% confidence interval [CI] 0.38–0.73) and the relative risk of bleeding was 0.50 (95% CI 028–0.89). Psyllium husk is an inexpensive bulk-forming fiber supplement; the optimal daily dosage is not known.

We recommend at least 28 g of daily fiber intake for women and 38 g for men, for which psyllium husk can be used to complement the diet.

Laxatives for some

Laxatives such as docusate are used to change the stool consistency when there is an organic bowel problem rather than a dietary issue. They can be used as a complementary treatment to enhance the effect of the fiber treatment.

Other measures

Topical anesthesia (eg, 5% lidocaine) is commonly used to treat pain from low-grade lesions, but no reliable data have been published. As most cases of hemorrhoids tend to progress over time, one should not expect long-term improvement with topical anesthesia. Nevertheless, it can be used as an ancillary treatment in select cases when short-term improvement is the main goal, and we recommend it based on our own experience.

Hygiene. Bidet use or cleaning the perianal area with water is recommended.

Phlebotonics contain a variety of ingredients including natural plant extracts such as flavonoids and synthetic products. Even though the exact mechanism of action is not known, phlebotonics are thought to increase venous and lymphatic drainage, normalize capillary permeability, and decrease inflammation in the hemorrhoidal cushions.4,11–13

In a Cochrane review of 24 randomized controlled trials, Perera et al14 found that phlebotonics improved the outcomes of:

  • Bleeding (odds ratio [OR] 0.21, number needed to treat [NNT] 4.8, P = .0002)
  • Pruritus (OR 0.23, NNT 9.1, P = .02)
  • Discharge or leakage (OR 0.12, NNT 5, P = .0008)
  • Overall symptoms (OR 15.99, NNT 2.7, P < .00001). Overall symptoms were also improved in the subgroup of pregnant patients.

Although phlebotonics give better results than placebo in the short-term management of hemorrhoids, there is a paucity of long-term data. Thus, the ASCRS clinical practice guidelines gives the regular use of these agents only a weak recommendation.3

Flavonoids (diosmin, hesperidin, rutoside), in a meta-analysis vs placebo in 1,514 patients, showed a beneficial response in terms of bleeding (relative risk [RR] 0.33), pruritus (relative risk [RR] 0.65), and recurrences (RR 0.53).15

Although Preparation H is commonly used as an over-the-counter medication, there are no good data on it, and it is not considered a phlebotonic.

 

 

OFFICE-BASED TREATMENTS

Office-based treatments—rubber band ligation, infrared photocoagulation, and sclerotherapy—are commonly used for grade I, II, and III hemorrhoids that have not responded to conservative management. The primary goal of these treatments is to decrease blood flow into the hemorrhoidal sac.

Even though office-based treatments are highly effective and major complications are uncommon, recurrence rates can be high, requiring patients to undergo additional treatments. Moreover, septic complications can occur, so patients should be closely observed for fever and urinary problems. Pain is a common symptom after office-based treatments, and bleeding may also occur.

The ASCRS guidelines strongly recommend office-based treatments for patients with grade I and II hemorrhoids, and for some with grade III hemorrhoids.3

Rubber band ligation

cengiz_hemorrhoidtreatment_f2.jpg
%3Cp%3EFigure%202.%20In%20rubber%20band%20ligation%2C%20an%20internal%20hemorrhoid%20is%20grasped%20with%20a%20forceps%20(A)%20and%20drawn%20into%20the%20cylinder%20of%20the%20ligator%20(B).%20A%20band%20is%20deployed%20around%20the%20base%20of%20the%20hemorrhoid%20(C)%2C%20cutting%20off%20its%20blood%20supply%20and%20causing%20it%20to%20fall%20off%20within%20a%20few%20days.%3C%2Fp%3E
Ligating the apex of the hemorrhoidal cushion stops the arterial flow and causes the hemorrhoidal tissue to undergo necrosis (Figure 2). The ligation is performed above the dentate line, where the sensory nerve fibers differ from those found below the line; therefore, the operation causes less pain than one would expect. One or more hemorrhoidal cushions can be ligated at the same time, although increased pain, bleeding, and vasomotor reactions have been reported with multiple banding during a single procedure.16,17

Iyer et al18 reported that patients on warfarin therapy had up to a 9 times higher risk of postprocedural bleeding, and patients on aspirin had a risk up to 3 times higher. Therefore, whether patients on anticoagulant therapy should undergo this procedure is unclear.

A Cochrane database review19 found this technique effective for hemorrhoid grades I through III, although some patients with grade III hemorrhoids may benefit more from excisional hemorrhoidectomy, which is associated with a lower recurrence rate than rubber band ligation.

Brown et al20 performed a randomized controlled trial comparing hemorrhoidal artery ligation and rubber band ligation for symptomatic hemorrhoids in 372 patients with grade II and III hemorrhoids. Postprocedural pain scores on days 1 and 7 were significantly lower with rubber band ligation, but recurrences were more common (49% vs 30%, P = .0005, respectively).

Overall, rubber band ligation is an excellent option for grade II hemorrhoids, as it is easy to perform, is associated with low pain scores, and can be used to treat recurrences.

Infrared photocoagulation

In this procedure, an infrared probe produces heat to induce coagulation, fibrosis, and ultimately necrosis of the protruding tissue in the hemorrhoidal cushions.21 Even though its use was initially directed at grade I and II hemorrhoids, recent reports showed acceptable results for grades III and IV.22,23 A randomized controlled trial comparing infrared photocoagulation and rubber band ligation in 94 patients found that both procedures were well accepted and highly effective; however, patients had better pain scores with photocoagulation in the first 24 hours after the procedure (P < .05).24

Sclerotherapy

cengiz_hemorrhoidtreatment_f3.jpg
%3Cp%3EFigure%203.%20Sclerotherapy%20involves%20injecting%20an%20irritating%20solution%20into%20the%20hemorrhoid%2C%20reducing%20its%20blood%20supply%20and%20causing%20it%20to%20shrink.%3C%2Fp%3E
Sclerotherapy involves injection of a sclerotic agent into the submucosa of the hemorrhoidal sac (Figure 3), which causes an inflammatory reaction and eventually forms fibrotic tissue that stops the blood flow to the hemorrhoid. Many sclerotic agents are available, including 5% phenol in almond or vegetable oil, quinine, ethanolamine, and hypertonic saline.21

The injection can cause prostatic abscess and sepsis, although this is rare.25 Nevertheless, high fever and postprocedural pain should be carefully evaluated.

There have been few randomized trials of sclerotherapy, but success rates so far have been higher for grade I hemorrhoids than for grades II and III.26–28 It is the preferred method for patients who have bleeding abnormalities caused by medications or other diseases (eg, cirrhosis).

 

 

SURGERY

Although nonsurgical treatments have substantially improved, surgery is the most effective and strongly recommended treatment for patients with high-grade internal hemorrhoids (grades III and IV), external and mixed hemorrhoids, and recurrent hemorrhoids.

The most popular surgical options are open or closed hemorrhoidectomy, stapled hemorrhoidopexy, and Doppler-guided hemorrhoidal artery ligation. Each has different success rates and different complication profiles, which need to be discussed with the patient.

Overall, surgery is associated with more adverse effects than office-based treatments or medical management. Postoperative pain is the most common complaint, but anal stricture (rare) or incontinence may occur due to excessive tissue excision and damage to the sphincter muscles. These can be avoided by maintaining the normal anoderm between excisions, by not excising all hemorrhoid sacs at once if the patient has extensive lesions, and by performing a careful dissection in the submucosal plane.

Patients with profuse bleeding or an underlying bleeding abnormality are best managed with surgical approaches performed in an operating room.

Excisional surgical hemorrhoidectomy

Excision of the hemorrhoidal sac, the most conventional surgical technique, is generally reserved for prolapsing disease. The recurrence rate after excisional hemorrhoidectomy is significantly lower than with any other approach.29

Excisional hemorrhoidectomy can be performed using either an open approach, in which the edges of the mucosal defect are not reapproximated, or a closed approach, in which they are. In a systematic review, Bhatti et al30 compared open vs closed techniques and found that the closed technique resulted in less postoperative pain, better wound healing, and less bleeding. Rates of recurrence, postoperative complications, and surgical site infection and lengths of stay were comparable with either procedure.

Overall, excisional hemorrhoidectomy is associated with higher pain scores than any other surgical method.29 Recently, the use of electrodiathermy energy devices, also described as electrosurgical vessel-sealing devices, have further improved overall patient satisfaction.31

Multiple painful hemorrhoidal sacs require a careful surgical approach, as extensive resection may cause widespread fibrosis and stricture. As with anal stricture, fecal incontinence can be prevented by careful dissection. However, already existing incontinence is not a contraindication for the surgery.

Doppler-guided hemorrhoidal artery ligation

Doppler-guided hemorrhoidal artery ligation involves using a Doppler probe to find and ligate individual hemorrhoidal arteries. Additionally, mucopexy (transanal rectoanal repair) is performed to relocate the prolapsing tissue. Avital et al32 reported that at 1 year after this procedure, recurrence rates were 5.3% for grade II hemorrhoids and 13% for grade III hemorrhoids. At 5 years, recurrence rates were 12% for grade II and 31% for grade III.

To date, this procedure appears to be suitable for grade I, II, and III hemorrhoids, especially for grade II, but more studies are needed to prove its efficacy and recurrence rates for more advanced lesions. Although this technique has a high morbidity rate (18%), primarily pain or tenesmus, it causes less postoperative pain than other surgical methods.33 Overall, it has the potential to become a favored treatment.

Stapled hemorrhoidopexy

cengiz_hemorrhoidtreatment_f4.jpg
%3Cp%3EFigure%204.%20In%20stapled%20hemorrhoidopexy%2C%20a%20special%20tool%20is%20inserted%20(A).%20Excess%20tissue%20is%20excised%20(B)%2C%20and%20the%20remaining%20tissue%20is%20drawn%20up%20to%20its%20normal%20position%20and%20fastened%20in%20place%2C%20yielding%20the%20result%20pictured%20in%20panel%20C.%3C%2Fp%3E
In this procedure, the prolapsing part of the internal hemorrhoidal cushion is moved upward by stapling the rectal mucosa just above the hemorrhoid (Figure 4). This is not an option for patients with thrombosed internal hemorrhoids or with external hemorrhoids.

Although pain scores are lower with stapled hemorrhoidopexy than with excisional hemorrhoidectomy, this procedure is not superior in terms of recurrences.34,35 Also, practitioners should be careful about specific complications of stapled hemorrhoidopexy, such as rectovaginal fistula, anal stenosis, or sphincter injuries. These specific complications should be clearly explained to patients, and necessary information should be given to patients upon discharge. The primary care physician should also be careful about fistulas and stenoses in this particular patient population.

 

 

NO ‘BEST’ TREATMENT

There is no best treatment for hemorrhoids. Every patient is different, and the physician and patient need to understand each other’s expectations, weigh the risks and benefits, and arrive at a mutual decision. A good patient-doctor relationship is essential.

cengiz_hemorrhoidtreatment_f5.jpg
%3Cp%3EFigure%205.%20Algorithm%20for%20hemorrhoid%20management.%3C%2Fp%3E
A thorough history and physical examination will enable the practitioner to understand the patient’s problem (Figure 5).

Given the variety of available treatments, head-to-head comparisons are difficult. Moreover, the efficacy and applicability of each technique changes with the grade of the lesion or lesions and the skill of the practitioner. Lacking comprehensive studies comparing conservative, office-based, and surgical management, no decisive statements can be made based on current evidence.

Patients with compounding conditions

Pregnant patients often develop hemorrhoids as intra-abdominal pressure increases, particularly during the third trimester.36 Also, acute episodes of pain and bleeding are common in pregnant women with preexisting hemorrhoids.

Conservative treatment is the main approach in pregnant patients because most hemorrhoids regress after childbirth. This includes increased dietary fiber, stool softeners, and sitz baths, which are safe to use for external hemorrhoids. Any office-based or surgical intervention should be postponed until after childbirth, if possible. Kegel exercises and lying on the left side are also recommended to relieve symptoms. In cases of severe bleeding, anal packing appears to be useful.

Immunosuppressed patients and those on anticoagulant therapy are more prone to serious complications such as sepsis and profuse bleeding. Thus, conservative management should be used in these patients as well. Injection sclerotherapy may be beneficial, as it has been shown to decrease bleeding. Of note, patients on immunosuppressive agents should stop taking them and start taking an antibiotic, and patients on anticoagulant or antiplatelet medications should be instructed to stop them 1 week before any intervention.

Crohn disease. Some patients with Crohn disease may have hemorrhoids, though this is rare. Eglinton et al,37 in a series of 715 patients with Crohn disease, reported that 190 (26.6%) had symptomatic perianal disease. Of these, only 3 (1.6%) had hemorrhoids. Treatment is always conservative and directed at the Crohn disease rather than the hemorrhoids.

Patients with portal hypertension (eg, due to cirrhosis) are prone to have anorectal varices that may resemble hemorrhoids. Anorectal varices can be treated with vascular ligation, whereas sclerotherapy is the preferred method for hemorrhoids in this group, in whom coagulopathy is common.

TAKE-HOME MESSAGES

Hemorrhoidal disease is common in the United States, and with our diet and lifestyle, the incidence is likely to increase. (A national survey found that overall dietary quality improved modestly in children and adolescents in the United States from 1999 to 2012 but remained far below optimal.38) Practitioners need to carefully assess hemorrhoidal symptoms and complete any necessary screening tests before establishing a diagnosis. This helps to avoid missing any underlying disease.

Fiber supplements along with dietary and lifestyle changes constitute the baseline of the management regardless of the disease grade. Office-based interventions are beneficial for grade I and II hemorrhoids and for some grade III hemorrhoids. Repeated interventions can increase the success rate. In patients with high-grade, symptomatic hemorrhoids, surgical hemorrhoidectomy is the most effective modality with the lowest recurrence rates, although it causes more pain than conservative methods.

Aspects of modern life that may promote hemorrhoids include increased consumption of processed foods, a sedentary lifestyle, and using cell phones while defecating, which translates to much more time spent on the toilet.

Hemorrhoids accounted for more than 3.5 million US outpatient visits in 2010, and they were the third leading cause of hospital admissions related to gastrointestinal disease.1

Here, we review the process for diagnosing and grading hemorrhoids, as well as for selecting the appropriate medical or surgical treatment based on the most recent clinical evidence.

DIAGNOSING AND CLASSIFYING HEMORRHOIDS

Hemorrhoids are the distal prolapse of the arteriovenous bundle, muscle fibers, and surrounding connective tissue as an envelope below the dentate line in the anal canal. They usually present with painless rectal bleeding.2

The diagnosis of hemorrhoids relies on the history and physical examination rather than on laboratory testing or imaging studies. Typically, the presenting symptom is painless rectal bleeding associated with bowel movements, usually appearing as bright red blood on the toilet paper or coating the stool. Severe itching and anal discomfort are also common, especially with chronic hemorrhoids.

Detailed patient history

A detailed patient history is important. It should include the extent, severity, and duration of symptoms, frequency of bowel movements, associated symptoms (eg, constipation, fecal incontinence), daily dietary habits, and details of bowel movements (eg, time spent during each bowel movement and concomitant cell phone use).3

Regarding bowel habits, some patients experience lifelong constipation or diarrhea. Therefore, what a patient considers a normal bowel habit may not be normal and should be investigated.4 Also, it is important to exclude external thrombosed hemorrhoids, anal fissure, anal abscess, and Crohn disease.5

Physical examination

A digital rectal examination is the second step. During the examination, look for skin tags, sphincter tone, perianal hygiene, and synchronous anal lesions.3 Of note, the Valsalva maneuver can be performed during the digital rectal examination.

Red flags for colorectal cancer on the digital rectal examination include a mass with or without presence of hemorrhoidal sacs and a bleeding source above the level of internal hemorrhoids.

cengiz_hemorrhoidtreatment_f1.jpg
%3Cp%3EFigure%201.%20Patient%20with%20Crohn%20disease.%20Note%20the%20fistula%20orifices%20and%20the%20skin%20tag.%3C%2Fp%3E
Patients with recurrent abscesses, fistulas, or skin tags (especially cauliflower-type skin tags) should be investigated for Crohn disease (Figure 1).

Endoscopy

Since rectal bleeding can be a sign of several diseases, including colorectal cancer, it is important to review any previous endoscopic results. Patients at high risk of colon cancer should undergo rigid proctoscopy, flexible sigmoidoscopy, or colonoscopy.3,4 In our practice, we recommend endoscopic evaluation for patients over age 40 with rectal bleeding, especially if they have a family history of colorectal cancer.

External or internal (grades I–IV)

Hemorrhoids can be categorized as either external or internal.

External hemorrhoids are distinguished by their outer covering with perianal skin and anoderm and their location inferior to the dentate line. They are painful if the hemorrhoidal sac is occluded by a thrombotic clot.

Internal hemorrhoids are above the dentate line and covered with rectal columnar and transitional mucosa. They are further graded on a 4-point scale3:

  • Grade I—Visible hemorrhoids that do not prolapse
  • Grade II—Hemorrhoids that prolapse during the Valsalva maneuver but spontaneously reduce
  • Grade III—Hemorrhoids that prolapse during the Valsalva maneuver and need manual reduction
  • Grade IV—Nonreducible hemorrhoids.

A RANGE OF TREATMENTS

In choosing the treatment for hemorrhoids, one should consider the disease grade and severity, its impact on the quality of life, the degree of pain it causes, the patient’s likelihood of adhering to treatment, and the patient’s personal preference.

Regardless of severity, treatment almost always starts with a high-fiber diet and other lifestyle modifications that include bowel movement behaviors. This requires practitioners to spend significant time on patient education regardless of the type or the severity of the disease.

Treatments can be grouped in 3 categories: conservative, office-based, and surgical. Practitioners should thoroughly discuss the options with the patient, emphasizing the pros and cons of each.

 

 

CONSERVATIVE MEASURES

Conservative measures are aimed at softening the stool, relieving pain, and correcting bad toileting habits. In most cases, the primary precipitating factor is lifestyle, and unless patients change it, they are more likely to have recurrent symptoms in the long term.

No phone in the bathroom

People take their phones into the bathroom, and this habit is blamed for increasing the time on the toilet and leading to increased pressure on the anal region and straining during defecation. Some research points to a direct correlation between the time spent on the toilet and hemorrhoidal disease, although the exact cause-and-effect relationship with cell phone use has not been determined. In general, spending excessive time on the commode, including reading, should be discouraged.

Less time in the bathroom

Johannsson et al6 reported that patients with hemorrhoids spent more time on the toilet and had to strain harder and more often than controls in the community and hospital.

Garg and Singh7 and Garg8 use the mnemonic “TONE” for appropriate defecation habits:

  • Three minutes during defecation
  • Once-daily defecation
  • No straining and no cell phone use during defecation
  • Enough fiber.

More fiber

Fiber draws water into the lumen of the colon, increasing the water content of the stool. Recommended daily fiber intake is about 28 g for women and 38 g for men.9 This high level of intake is hard to achieve without supplements for someone who consumes a classic American diet with a lot of fast food.

Fiber supplements are strongly recommended in the American Society of Colon and Rectal Surgeons (ASCRS) practice guidelines3 based on a Cochrane review.10 In this meta-analysis, with fiber supplements, the relative risk of persisting or nonimproving symptoms was 0.53 (95% confidence interval [CI] 0.38–0.73) and the relative risk of bleeding was 0.50 (95% CI 028–0.89). Psyllium husk is an inexpensive bulk-forming fiber supplement; the optimal daily dosage is not known.

We recommend at least 28 g of daily fiber intake for women and 38 g for men, for which psyllium husk can be used to complement the diet.

Laxatives for some

Laxatives such as docusate are used to change the stool consistency when there is an organic bowel problem rather than a dietary issue. They can be used as a complementary treatment to enhance the effect of the fiber treatment.

Other measures

Topical anesthesia (eg, 5% lidocaine) is commonly used to treat pain from low-grade lesions, but no reliable data have been published. As most cases of hemorrhoids tend to progress over time, one should not expect long-term improvement with topical anesthesia. Nevertheless, it can be used as an ancillary treatment in select cases when short-term improvement is the main goal, and we recommend it based on our own experience.

Hygiene. Bidet use or cleaning the perianal area with water is recommended.

Phlebotonics contain a variety of ingredients including natural plant extracts such as flavonoids and synthetic products. Even though the exact mechanism of action is not known, phlebotonics are thought to increase venous and lymphatic drainage, normalize capillary permeability, and decrease inflammation in the hemorrhoidal cushions.4,11–13

In a Cochrane review of 24 randomized controlled trials, Perera et al14 found that phlebotonics improved the outcomes of:

  • Bleeding (odds ratio [OR] 0.21, number needed to treat [NNT] 4.8, P = .0002)
  • Pruritus (OR 0.23, NNT 9.1, P = .02)
  • Discharge or leakage (OR 0.12, NNT 5, P = .0008)
  • Overall symptoms (OR 15.99, NNT 2.7, P < .00001). Overall symptoms were also improved in the subgroup of pregnant patients.

Although phlebotonics give better results than placebo in the short-term management of hemorrhoids, there is a paucity of long-term data. Thus, the ASCRS clinical practice guidelines gives the regular use of these agents only a weak recommendation.3

Flavonoids (diosmin, hesperidin, rutoside), in a meta-analysis vs placebo in 1,514 patients, showed a beneficial response in terms of bleeding (relative risk [RR] 0.33), pruritus (relative risk [RR] 0.65), and recurrences (RR 0.53).15

Although Preparation H is commonly used as an over-the-counter medication, there are no good data on it, and it is not considered a phlebotonic.

 

 

OFFICE-BASED TREATMENTS

Office-based treatments—rubber band ligation, infrared photocoagulation, and sclerotherapy—are commonly used for grade I, II, and III hemorrhoids that have not responded to conservative management. The primary goal of these treatments is to decrease blood flow into the hemorrhoidal sac.

Even though office-based treatments are highly effective and major complications are uncommon, recurrence rates can be high, requiring patients to undergo additional treatments. Moreover, septic complications can occur, so patients should be closely observed for fever and urinary problems. Pain is a common symptom after office-based treatments, and bleeding may also occur.

The ASCRS guidelines strongly recommend office-based treatments for patients with grade I and II hemorrhoids, and for some with grade III hemorrhoids.3

Rubber band ligation

cengiz_hemorrhoidtreatment_f2.jpg
%3Cp%3EFigure%202.%20In%20rubber%20band%20ligation%2C%20an%20internal%20hemorrhoid%20is%20grasped%20with%20a%20forceps%20(A)%20and%20drawn%20into%20the%20cylinder%20of%20the%20ligator%20(B).%20A%20band%20is%20deployed%20around%20the%20base%20of%20the%20hemorrhoid%20(C)%2C%20cutting%20off%20its%20blood%20supply%20and%20causing%20it%20to%20fall%20off%20within%20a%20few%20days.%3C%2Fp%3E
Ligating the apex of the hemorrhoidal cushion stops the arterial flow and causes the hemorrhoidal tissue to undergo necrosis (Figure 2). The ligation is performed above the dentate line, where the sensory nerve fibers differ from those found below the line; therefore, the operation causes less pain than one would expect. One or more hemorrhoidal cushions can be ligated at the same time, although increased pain, bleeding, and vasomotor reactions have been reported with multiple banding during a single procedure.16,17

Iyer et al18 reported that patients on warfarin therapy had up to a 9 times higher risk of postprocedural bleeding, and patients on aspirin had a risk up to 3 times higher. Therefore, whether patients on anticoagulant therapy should undergo this procedure is unclear.

A Cochrane database review19 found this technique effective for hemorrhoid grades I through III, although some patients with grade III hemorrhoids may benefit more from excisional hemorrhoidectomy, which is associated with a lower recurrence rate than rubber band ligation.

Brown et al20 performed a randomized controlled trial comparing hemorrhoidal artery ligation and rubber band ligation for symptomatic hemorrhoids in 372 patients with grade II and III hemorrhoids. Postprocedural pain scores on days 1 and 7 were significantly lower with rubber band ligation, but recurrences were more common (49% vs 30%, P = .0005, respectively).

Overall, rubber band ligation is an excellent option for grade II hemorrhoids, as it is easy to perform, is associated with low pain scores, and can be used to treat recurrences.

Infrared photocoagulation

In this procedure, an infrared probe produces heat to induce coagulation, fibrosis, and ultimately necrosis of the protruding tissue in the hemorrhoidal cushions.21 Even though its use was initially directed at grade I and II hemorrhoids, recent reports showed acceptable results for grades III and IV.22,23 A randomized controlled trial comparing infrared photocoagulation and rubber band ligation in 94 patients found that both procedures were well accepted and highly effective; however, patients had better pain scores with photocoagulation in the first 24 hours after the procedure (P < .05).24

Sclerotherapy

cengiz_hemorrhoidtreatment_f3.jpg
%3Cp%3EFigure%203.%20Sclerotherapy%20involves%20injecting%20an%20irritating%20solution%20into%20the%20hemorrhoid%2C%20reducing%20its%20blood%20supply%20and%20causing%20it%20to%20shrink.%3C%2Fp%3E
Sclerotherapy involves injection of a sclerotic agent into the submucosa of the hemorrhoidal sac (Figure 3), which causes an inflammatory reaction and eventually forms fibrotic tissue that stops the blood flow to the hemorrhoid. Many sclerotic agents are available, including 5% phenol in almond or vegetable oil, quinine, ethanolamine, and hypertonic saline.21

The injection can cause prostatic abscess and sepsis, although this is rare.25 Nevertheless, high fever and postprocedural pain should be carefully evaluated.

There have been few randomized trials of sclerotherapy, but success rates so far have been higher for grade I hemorrhoids than for grades II and III.26–28 It is the preferred method for patients who have bleeding abnormalities caused by medications or other diseases (eg, cirrhosis).

 

 

SURGERY

Although nonsurgical treatments have substantially improved, surgery is the most effective and strongly recommended treatment for patients with high-grade internal hemorrhoids (grades III and IV), external and mixed hemorrhoids, and recurrent hemorrhoids.

The most popular surgical options are open or closed hemorrhoidectomy, stapled hemorrhoidopexy, and Doppler-guided hemorrhoidal artery ligation. Each has different success rates and different complication profiles, which need to be discussed with the patient.

Overall, surgery is associated with more adverse effects than office-based treatments or medical management. Postoperative pain is the most common complaint, but anal stricture (rare) or incontinence may occur due to excessive tissue excision and damage to the sphincter muscles. These can be avoided by maintaining the normal anoderm between excisions, by not excising all hemorrhoid sacs at once if the patient has extensive lesions, and by performing a careful dissection in the submucosal plane.

Patients with profuse bleeding or an underlying bleeding abnormality are best managed with surgical approaches performed in an operating room.

Excisional surgical hemorrhoidectomy

Excision of the hemorrhoidal sac, the most conventional surgical technique, is generally reserved for prolapsing disease. The recurrence rate after excisional hemorrhoidectomy is significantly lower than with any other approach.29

Excisional hemorrhoidectomy can be performed using either an open approach, in which the edges of the mucosal defect are not reapproximated, or a closed approach, in which they are. In a systematic review, Bhatti et al30 compared open vs closed techniques and found that the closed technique resulted in less postoperative pain, better wound healing, and less bleeding. Rates of recurrence, postoperative complications, and surgical site infection and lengths of stay were comparable with either procedure.

Overall, excisional hemorrhoidectomy is associated with higher pain scores than any other surgical method.29 Recently, the use of electrodiathermy energy devices, also described as electrosurgical vessel-sealing devices, have further improved overall patient satisfaction.31

Multiple painful hemorrhoidal sacs require a careful surgical approach, as extensive resection may cause widespread fibrosis and stricture. As with anal stricture, fecal incontinence can be prevented by careful dissection. However, already existing incontinence is not a contraindication for the surgery.

Doppler-guided hemorrhoidal artery ligation

Doppler-guided hemorrhoidal artery ligation involves using a Doppler probe to find and ligate individual hemorrhoidal arteries. Additionally, mucopexy (transanal rectoanal repair) is performed to relocate the prolapsing tissue. Avital et al32 reported that at 1 year after this procedure, recurrence rates were 5.3% for grade II hemorrhoids and 13% for grade III hemorrhoids. At 5 years, recurrence rates were 12% for grade II and 31% for grade III.

To date, this procedure appears to be suitable for grade I, II, and III hemorrhoids, especially for grade II, but more studies are needed to prove its efficacy and recurrence rates for more advanced lesions. Although this technique has a high morbidity rate (18%), primarily pain or tenesmus, it causes less postoperative pain than other surgical methods.33 Overall, it has the potential to become a favored treatment.

Stapled hemorrhoidopexy

cengiz_hemorrhoidtreatment_f4.jpg
%3Cp%3EFigure%204.%20In%20stapled%20hemorrhoidopexy%2C%20a%20special%20tool%20is%20inserted%20(A).%20Excess%20tissue%20is%20excised%20(B)%2C%20and%20the%20remaining%20tissue%20is%20drawn%20up%20to%20its%20normal%20position%20and%20fastened%20in%20place%2C%20yielding%20the%20result%20pictured%20in%20panel%20C.%3C%2Fp%3E
In this procedure, the prolapsing part of the internal hemorrhoidal cushion is moved upward by stapling the rectal mucosa just above the hemorrhoid (Figure 4). This is not an option for patients with thrombosed internal hemorrhoids or with external hemorrhoids.

Although pain scores are lower with stapled hemorrhoidopexy than with excisional hemorrhoidectomy, this procedure is not superior in terms of recurrences.34,35 Also, practitioners should be careful about specific complications of stapled hemorrhoidopexy, such as rectovaginal fistula, anal stenosis, or sphincter injuries. These specific complications should be clearly explained to patients, and necessary information should be given to patients upon discharge. The primary care physician should also be careful about fistulas and stenoses in this particular patient population.

 

 

NO ‘BEST’ TREATMENT

There is no best treatment for hemorrhoids. Every patient is different, and the physician and patient need to understand each other’s expectations, weigh the risks and benefits, and arrive at a mutual decision. A good patient-doctor relationship is essential.

cengiz_hemorrhoidtreatment_f5.jpg
%3Cp%3EFigure%205.%20Algorithm%20for%20hemorrhoid%20management.%3C%2Fp%3E
A thorough history and physical examination will enable the practitioner to understand the patient’s problem (Figure 5).

Given the variety of available treatments, head-to-head comparisons are difficult. Moreover, the efficacy and applicability of each technique changes with the grade of the lesion or lesions and the skill of the practitioner. Lacking comprehensive studies comparing conservative, office-based, and surgical management, no decisive statements can be made based on current evidence.

Patients with compounding conditions

Pregnant patients often develop hemorrhoids as intra-abdominal pressure increases, particularly during the third trimester.36 Also, acute episodes of pain and bleeding are common in pregnant women with preexisting hemorrhoids.

Conservative treatment is the main approach in pregnant patients because most hemorrhoids regress after childbirth. This includes increased dietary fiber, stool softeners, and sitz baths, which are safe to use for external hemorrhoids. Any office-based or surgical intervention should be postponed until after childbirth, if possible. Kegel exercises and lying on the left side are also recommended to relieve symptoms. In cases of severe bleeding, anal packing appears to be useful.

Immunosuppressed patients and those on anticoagulant therapy are more prone to serious complications such as sepsis and profuse bleeding. Thus, conservative management should be used in these patients as well. Injection sclerotherapy may be beneficial, as it has been shown to decrease bleeding. Of note, patients on immunosuppressive agents should stop taking them and start taking an antibiotic, and patients on anticoagulant or antiplatelet medications should be instructed to stop them 1 week before any intervention.

Crohn disease. Some patients with Crohn disease may have hemorrhoids, though this is rare. Eglinton et al,37 in a series of 715 patients with Crohn disease, reported that 190 (26.6%) had symptomatic perianal disease. Of these, only 3 (1.6%) had hemorrhoids. Treatment is always conservative and directed at the Crohn disease rather than the hemorrhoids.

Patients with portal hypertension (eg, due to cirrhosis) are prone to have anorectal varices that may resemble hemorrhoids. Anorectal varices can be treated with vascular ligation, whereas sclerotherapy is the preferred method for hemorrhoids in this group, in whom coagulopathy is common.

TAKE-HOME MESSAGES

Hemorrhoidal disease is common in the United States, and with our diet and lifestyle, the incidence is likely to increase. (A national survey found that overall dietary quality improved modestly in children and adolescents in the United States from 1999 to 2012 but remained far below optimal.38) Practitioners need to carefully assess hemorrhoidal symptoms and complete any necessary screening tests before establishing a diagnosis. This helps to avoid missing any underlying disease.

Fiber supplements along with dietary and lifestyle changes constitute the baseline of the management regardless of the disease grade. Office-based interventions are beneficial for grade I and II hemorrhoids and for some grade III hemorrhoids. Repeated interventions can increase the success rate. In patients with high-grade, symptomatic hemorrhoids, surgical hemorrhoidectomy is the most effective modality with the lowest recurrence rates, although it causes more pain than conservative methods.

References
  1. Peery AF, Crockett SD, Barritt AS, et al. Burden of gastrointestinal, liver, and pancreatic diseases in the United States. Gastroenterology 2015; 149(7):1731–1741.e3. doi:10.1053/j.gastro.2015.08.045
  2. Thomson WH. The nature and cause of haemorrhoids. Proc R Soc Med 1975; 68(9):574–575. pmid:1197343
  3. Davis BR, Lee-Kong SA, Migaly J, Feingold DL, Steele SR. The American Society of Colon and Rectal Surgeons clinical practice guidelines for the management of hemorrhoids. Dis Colon Rectum 2018; 61(3):284–292. doi:10.1097/DCR.0000000000001030
  4. Lohsiriwat V. Treatment of hemorrhoids: a coloproctologist’s view. World J Gastroenterol 2015; 21(31):9245–9252. doi:10.3748/wjg.v21.i31.9245
  5. Wolf AMD, Fontham ETH, Church TR, et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin 2018; 68(4):250–281. doi:10.3322/caac.21457
  6. Johannsson HO, Graf W, Påhlman L. Bowel habits in hemorrhoid patients and normal subjects. Am J Gastroenterol 2005; 100(2):401–406. doi:10.1111/j.1572-0241.2005.40195.x
  7. Garg P, Singh P. Adequate dietary fiber supplement and TONE can help avoid surgery in most patients with advanced hemorrhoids. Minerva Gastroenterol Dietol 2017; 63(2):92–96. doi:10.23736/S1121-421X.17.02364-9
  8. Garg P. Conservative treatment of hemorrhoids deserves more attention in guidelines and clinical practice [letter]. Dis Colon Rectum 2018; 61(7):e348. doi:10.1097/DCR.0000000000001127
  9. Rakinic J, Poola VP. Hemorrhoids and fistulas: new solutions to old problems. Curr Probl Surg 2014; 51(3):98–137. doi:10.1067/j.cpsurg.2013.11.002
  10. Alonso-Coello P, Guyatt G, Heels-Ansdell D, et al. Laxatives for the treatment of hemorrhoids. Cochrane Database Syst Rev 2005; (4):CD004649. doi:10.1002/14651858.CD004649.pub2
  11. Struckmann JR. Clinical efficacy of micronized purified flavonoid fraction: an overview. J Vasc Res 1999; 36(suppl 1):37–41. doi:10.1159/000054072
  12. Shoab SS, Porter J, Scurr JH, Coleridge-Smith PD. Endothelial activation response to oral micronised flavonoid therapy in patients with chronic venous disease—a prospective study. Eur J Vasc Endovasc Surg 1999; 17(4):313–318. doi:10.1053/ejvs.1998.0751
  13. Meyer OC. Safety and security of Daflon 500 mg in venous insufficiency and in hemorrhoidal disease. Angiology 1994; 45(6 pt 2):579–584. pmid:8203791
  14. Perera N, Liolitsa D, Iype S, et al. Phlebotonics for haemorrhoids. Cochrane Database Syst Rev 2012;(8):CD004322. doi:10.1002/14651858.CD004322.pub3
  15. Alonso-Coello P, Zhou Q, Martinez-Zapata MJ, et al. Meta-analysis of flavonoids for the treatment of haemorrhoids. Br J Surg 2006; 93(8):909–920. doi:10.1002/bjs.5378
  16. Lee HH, Spencer RJ, Beart RW Jr. Multiple hemorrhoidal bandings in a single session. Dis Colon Rectum 1994; 37(1):37–41. pmid:8287745
  17. Law WL, Chu KW. Triple rubber band ligation for hemorrhoids: prospective, randomized trial of use of local anesthetic injection. Dis Colon Rectum 1999; 42(3):363–366. pmid:10223757
  18. Iyer VS, Shrier I, Gordon PH. Long-term outcome of rubber band ligation for symptomatic primary and recurrent internal hemorrhoids. Dis Colon Rectum 2004; 47(8):1364–1370. pmid:15484351
  19. Shanmugam V, Thaha MA, Rabindranath KS, Campbell KL, Steele RJ, Loudon MA. Rubber band ligation versus excisional haemorrhoidectomy for haemorrhoids. Cochrane Database Syst Rev 2005; (3):CD005034. doi:10.1002/14651858.CD005034.pub2
  20. Brown SR, Tiernan JP, Watson AJM, et al; HubBLe Study team. Haemorrhoidal artery ligation versus rubber band ligation for the management of symptomatic second-degree and third-degree haemorrhoids (HubBLe): a multicentre, open-label, randomised controlled trial. Lancet 2016; 388(10042):356–364. doi:10.1016/S0140-6736(16)30584-0
  21. ASGE Technology Committee; Siddiqui UD, Barth BA, Banerjee S, et al. Devices for the endoscopic treatment of hemorrhoids. Gastrointest Endosc 2014; 79(1):8–14. doi:10.1016/j.gie.2013.07.021
  22. Ahmad A, Kant R, Gupta A. Comparative analysis of Doppler guided hemorrhoidal artery ligation (DG-HAL) & infrared coagulation (IRC) in management of hemorrhoids. Indian J Surg 2013; 75(4):274–277. doi:10.1007/s12262-012-0444-5
  23. Poen AC, Felt-Bersma RJ, Cuesta MA, Devillé W, Meuwissen SG. A randomized controlled trial of rubber band ligation versus infra-red coagulation in the treatment of internal haemorrhoids. Eur J Gastroenterol Hepatol 2000; 12(5):535–539. pmid:10833097
  24. Marques CF, Nahas SC, Nahas CS, Sobrado CW Jr, Habr-Gama A, Kiss DR. Early results of the treatment of internal hemorrhoid disease by infrared coagulation and elastic banding: a prospective randomized cross-over trial. Tech Coloproctol 2006; 10(4):312–317. doi:10.1007/s10151-006-0299-5
  25. Madoff RD, Fleshman JW; Clinical Practice Committee, American Gastroenterological Association. American Gastroenterological Association technical review on the diagnosis and treatment of hemorrhoids. Gastroenterology 2004; 126(5):1463–1473. pmid:15131807
  26. Yano T, Yano K. Comparison of injection sclerotherapy between 5% phenol in almond oil and aluminum potassium sulfate and tannic acid for grade 3 hemorrhoids. Ann Coloproctol 2015; 31(3):103–105. doi:10.3393/ac.2015.31.3.103
  27. Kanellos I, Goulimaris I, Vakalis I, Dadoukis I. Long-term evaluation of sclerotherapy for haemorrhoids. A prospective study. Int J Surg Investig 2000; 2(4):295–298. pmid:12678531
  28. Moser KH, Mosch C, Walgenbach M, et al. Efficacy and safety of sclerotherapy with polidocanol foam in comparison with fluid sclerosant in the treatment of first-grade haemorrhoidal disease: a randomised, controlled, single-blind, multicentre trial. Int J Colorectal Dis 2013; 28(10):1439–1447. doi:10.1007/s00384-013-1729-2
  29. MacRae HM, McLeod RS. Comparison of hemorrhoidal treatments: a meta-analysis. Can J Surg 1997; 40(1):14–7. pmid:9030078
  30. Bhatti MI, Sajid MS, Baig MK. Milligan-Morgan (open) versus Ferguson haemorrhoidectomy (closed): a systematic review and meta-analysis of published randomized, controlled trials. World J Surg 2016; 40(6):1509–1519. doi:10.1007/s00268-016-3419-z
  31. Nienhuijs S, de Hingh I. Conventional versus LigaSure hemorrhoidectomy for patients with symptomatic hemorrhoids. Cochrane Database Syst Rev 2009; (1):CD006761. doi:10.1002/14651858.CD006761.pub2
  32. Avital S, Inbar R, Karin E, Greenberg R. Five-year follow-up of Doppler-guided hemorrhoidal artery ligation. Tech Coloproctol 2012; 16(1):61–65. doi:10.1007/s10151-011-0801-6
  33. Ratto C, Parello A, Veronese E, et al. Doppler-guided transanal haemorrhoidal dearterialization for haemorrhoids: results from a multicentre trial. Colorectal Dis 2015; 17(1):010–019. doi:10.1111/codi.12779
  34. Senagore AJ, Singer M, Abcarian H, et al; Procedure for Prolapse and Hemmorrhoids (PPH) Multicenter Study Group. A prospective, randomized, controlled multicenter trial comparing stapled hemorrhoidopexy and Ferguson hemorrhoidectomy: perioperative and one-year results. Dis Colon Rectum 2004; 47(11):1824–1836. pmid:15622574
  35. Jayaraman S, Colquhoun PH, Malthaner RA. Stapled versus conventional surgery for hemorrhoids. Cochrane Database Syst Rev 2006; (4):CD005393.
  36. Poskus T, Buzinskiene D, Drasutiene G, et al. Haemorrhoids and anal fissures during pregnancy and after childbirth: a prospective cohort study. BJOG 2014; 121(13):1666–1671. doi:10.1111/1471-0528.12838
  37. Eglinton TW, Barclay ML, Gearry RB, Frizelle FA. The spectrum of perianal Crohn’s disease in a population-based cohort. Dis Colon Rectum 2012; 55(7):773–777. doi:10.1097/DCR.0b013e31825228b0
  38. Gu X, Tucker KL. Dietary quality of the US child and adolescent population: trends from 1999 to 2012 and associations with the use of federal nutrition assistance programs. Am J Clin Nutr 2017; 105(1):194–202. doi:10.3945/ajcn.116.135095
References
  1. Peery AF, Crockett SD, Barritt AS, et al. Burden of gastrointestinal, liver, and pancreatic diseases in the United States. Gastroenterology 2015; 149(7):1731–1741.e3. doi:10.1053/j.gastro.2015.08.045
  2. Thomson WH. The nature and cause of haemorrhoids. Proc R Soc Med 1975; 68(9):574–575. pmid:1197343
  3. Davis BR, Lee-Kong SA, Migaly J, Feingold DL, Steele SR. The American Society of Colon and Rectal Surgeons clinical practice guidelines for the management of hemorrhoids. Dis Colon Rectum 2018; 61(3):284–292. doi:10.1097/DCR.0000000000001030
  4. Lohsiriwat V. Treatment of hemorrhoids: a coloproctologist’s view. World J Gastroenterol 2015; 21(31):9245–9252. doi:10.3748/wjg.v21.i31.9245
  5. Wolf AMD, Fontham ETH, Church TR, et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin 2018; 68(4):250–281. doi:10.3322/caac.21457
  6. Johannsson HO, Graf W, Påhlman L. Bowel habits in hemorrhoid patients and normal subjects. Am J Gastroenterol 2005; 100(2):401–406. doi:10.1111/j.1572-0241.2005.40195.x
  7. Garg P, Singh P. Adequate dietary fiber supplement and TONE can help avoid surgery in most patients with advanced hemorrhoids. Minerva Gastroenterol Dietol 2017; 63(2):92–96. doi:10.23736/S1121-421X.17.02364-9
  8. Garg P. Conservative treatment of hemorrhoids deserves more attention in guidelines and clinical practice [letter]. Dis Colon Rectum 2018; 61(7):e348. doi:10.1097/DCR.0000000000001127
  9. Rakinic J, Poola VP. Hemorrhoids and fistulas: new solutions to old problems. Curr Probl Surg 2014; 51(3):98–137. doi:10.1067/j.cpsurg.2013.11.002
  10. Alonso-Coello P, Guyatt G, Heels-Ansdell D, et al. Laxatives for the treatment of hemorrhoids. Cochrane Database Syst Rev 2005; (4):CD004649. doi:10.1002/14651858.CD004649.pub2
  11. Struckmann JR. Clinical efficacy of micronized purified flavonoid fraction: an overview. J Vasc Res 1999; 36(suppl 1):37–41. doi:10.1159/000054072
  12. Shoab SS, Porter J, Scurr JH, Coleridge-Smith PD. Endothelial activation response to oral micronised flavonoid therapy in patients with chronic venous disease—a prospective study. Eur J Vasc Endovasc Surg 1999; 17(4):313–318. doi:10.1053/ejvs.1998.0751
  13. Meyer OC. Safety and security of Daflon 500 mg in venous insufficiency and in hemorrhoidal disease. Angiology 1994; 45(6 pt 2):579–584. pmid:8203791
  14. Perera N, Liolitsa D, Iype S, et al. Phlebotonics for haemorrhoids. Cochrane Database Syst Rev 2012;(8):CD004322. doi:10.1002/14651858.CD004322.pub3
  15. Alonso-Coello P, Zhou Q, Martinez-Zapata MJ, et al. Meta-analysis of flavonoids for the treatment of haemorrhoids. Br J Surg 2006; 93(8):909–920. doi:10.1002/bjs.5378
  16. Lee HH, Spencer RJ, Beart RW Jr. Multiple hemorrhoidal bandings in a single session. Dis Colon Rectum 1994; 37(1):37–41. pmid:8287745
  17. Law WL, Chu KW. Triple rubber band ligation for hemorrhoids: prospective, randomized trial of use of local anesthetic injection. Dis Colon Rectum 1999; 42(3):363–366. pmid:10223757
  18. Iyer VS, Shrier I, Gordon PH. Long-term outcome of rubber band ligation for symptomatic primary and recurrent internal hemorrhoids. Dis Colon Rectum 2004; 47(8):1364–1370. pmid:15484351
  19. Shanmugam V, Thaha MA, Rabindranath KS, Campbell KL, Steele RJ, Loudon MA. Rubber band ligation versus excisional haemorrhoidectomy for haemorrhoids. Cochrane Database Syst Rev 2005; (3):CD005034. doi:10.1002/14651858.CD005034.pub2
  20. Brown SR, Tiernan JP, Watson AJM, et al; HubBLe Study team. Haemorrhoidal artery ligation versus rubber band ligation for the management of symptomatic second-degree and third-degree haemorrhoids (HubBLe): a multicentre, open-label, randomised controlled trial. Lancet 2016; 388(10042):356–364. doi:10.1016/S0140-6736(16)30584-0
  21. ASGE Technology Committee; Siddiqui UD, Barth BA, Banerjee S, et al. Devices for the endoscopic treatment of hemorrhoids. Gastrointest Endosc 2014; 79(1):8–14. doi:10.1016/j.gie.2013.07.021
  22. Ahmad A, Kant R, Gupta A. Comparative analysis of Doppler guided hemorrhoidal artery ligation (DG-HAL) & infrared coagulation (IRC) in management of hemorrhoids. Indian J Surg 2013; 75(4):274–277. doi:10.1007/s12262-012-0444-5
  23. Poen AC, Felt-Bersma RJ, Cuesta MA, Devillé W, Meuwissen SG. A randomized controlled trial of rubber band ligation versus infra-red coagulation in the treatment of internal haemorrhoids. Eur J Gastroenterol Hepatol 2000; 12(5):535–539. pmid:10833097
  24. Marques CF, Nahas SC, Nahas CS, Sobrado CW Jr, Habr-Gama A, Kiss DR. Early results of the treatment of internal hemorrhoid disease by infrared coagulation and elastic banding: a prospective randomized cross-over trial. Tech Coloproctol 2006; 10(4):312–317. doi:10.1007/s10151-006-0299-5
  25. Madoff RD, Fleshman JW; Clinical Practice Committee, American Gastroenterological Association. American Gastroenterological Association technical review on the diagnosis and treatment of hemorrhoids. Gastroenterology 2004; 126(5):1463–1473. pmid:15131807
  26. Yano T, Yano K. Comparison of injection sclerotherapy between 5% phenol in almond oil and aluminum potassium sulfate and tannic acid for grade 3 hemorrhoids. Ann Coloproctol 2015; 31(3):103–105. doi:10.3393/ac.2015.31.3.103
  27. Kanellos I, Goulimaris I, Vakalis I, Dadoukis I. Long-term evaluation of sclerotherapy for haemorrhoids. A prospective study. Int J Surg Investig 2000; 2(4):295–298. pmid:12678531
  28. Moser KH, Mosch C, Walgenbach M, et al. Efficacy and safety of sclerotherapy with polidocanol foam in comparison with fluid sclerosant in the treatment of first-grade haemorrhoidal disease: a randomised, controlled, single-blind, multicentre trial. Int J Colorectal Dis 2013; 28(10):1439–1447. doi:10.1007/s00384-013-1729-2
  29. MacRae HM, McLeod RS. Comparison of hemorrhoidal treatments: a meta-analysis. Can J Surg 1997; 40(1):14–7. pmid:9030078
  30. Bhatti MI, Sajid MS, Baig MK. Milligan-Morgan (open) versus Ferguson haemorrhoidectomy (closed): a systematic review and meta-analysis of published randomized, controlled trials. World J Surg 2016; 40(6):1509–1519. doi:10.1007/s00268-016-3419-z
  31. Nienhuijs S, de Hingh I. Conventional versus LigaSure hemorrhoidectomy for patients with symptomatic hemorrhoids. Cochrane Database Syst Rev 2009; (1):CD006761. doi:10.1002/14651858.CD006761.pub2
  32. Avital S, Inbar R, Karin E, Greenberg R. Five-year follow-up of Doppler-guided hemorrhoidal artery ligation. Tech Coloproctol 2012; 16(1):61–65. doi:10.1007/s10151-011-0801-6
  33. Ratto C, Parello A, Veronese E, et al. Doppler-guided transanal haemorrhoidal dearterialization for haemorrhoids: results from a multicentre trial. Colorectal Dis 2015; 17(1):010–019. doi:10.1111/codi.12779
  34. Senagore AJ, Singer M, Abcarian H, et al; Procedure for Prolapse and Hemmorrhoids (PPH) Multicenter Study Group. A prospective, randomized, controlled multicenter trial comparing stapled hemorrhoidopexy and Ferguson hemorrhoidectomy: perioperative and one-year results. Dis Colon Rectum 2004; 47(11):1824–1836. pmid:15622574
  35. Jayaraman S, Colquhoun PH, Malthaner RA. Stapled versus conventional surgery for hemorrhoids. Cochrane Database Syst Rev 2006; (4):CD005393.
  36. Poskus T, Buzinskiene D, Drasutiene G, et al. Haemorrhoids and anal fissures during pregnancy and after childbirth: a prospective cohort study. BJOG 2014; 121(13):1666–1671. doi:10.1111/1471-0528.12838
  37. Eglinton TW, Barclay ML, Gearry RB, Frizelle FA. The spectrum of perianal Crohn’s disease in a population-based cohort. Dis Colon Rectum 2012; 55(7):773–777. doi:10.1097/DCR.0b013e31825228b0
  38. Gu X, Tucker KL. Dietary quality of the US child and adolescent population: trends from 1999 to 2012 and associations with the use of federal nutrition assistance programs. Am J Clin Nutr 2017; 105(1):194–202. doi:10.3945/ajcn.116.135095
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Page Number
612-620
Page Number
612-620
Publications
Publications
Topics
Article Type
Display Headline
Hemorrhoids: A range of treatments
Display Headline
Hemorrhoids: A range of treatments
Legacy Keywords
hemorrhoids, rectal bleeding, fiber, bowel movement, Crohn disease, colorectal cancer, endoscopy, sigmoidoscopy, colonoscopy, laxative, psyllium, phlebotonic, rubber band ligation, sclerotherapy, infrared photocoagulation, excisional surgical hemorrhoidectomy, hemorrhoidal artery ligation, stapled hemorrhoidopexy, portal hypertension, Turgut Bora Cengiz, Emre Gorgun
Legacy Keywords
hemorrhoids, rectal bleeding, fiber, bowel movement, Crohn disease, colorectal cancer, endoscopy, sigmoidoscopy, colonoscopy, laxative, psyllium, phlebotonic, rubber band ligation, sclerotherapy, infrared photocoagulation, excisional surgical hemorrhoidectomy, hemorrhoidal artery ligation, stapled hemorrhoidopexy, portal hypertension, Turgut Bora Cengiz, Emre Gorgun
Sections
Inside the Article

KEY POINTS

  • Hemorrhoids account for more than 3.5 million office visits annually.
  • Most patients present with painless rectal bleeding, but this can also be a sign of colorectal cancer, which needs to be ruled out.
  • Fiber supplements along with dietary and lifestyle changes are recommended for all patients with hemorrhoids regardless of symptom severity.
  • Hemorrhoids are graded on a scale of I (least severe) through IV (most severe). Office-based treatments are effective for grades I, II, and some grade III hemorrhoids. Surgical excision is the standard for high-grade hemorrhoids.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/22/2019 - 09:00
Un-Gate On Date
Thu, 08/22/2019 - 09:00
Use ProPublica
CFC Schedule Remove Status
Thu, 08/22/2019 - 09:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

A young man with acute chest pain

Article Type
Changed
Tue, 09/03/2019 - 08:23
Display Headline
A young man with acute chest pain

An 18-year-old man without any significant medical history was transferred from another hospital for higher-level care after presenting with unremitting chest pain. He had been in his usual state of good health until 7 days before presentation, when he developed mild rhinorrhea and a sore throat, but not a cough. He went to an outpatient clinic, where a rapid test for group A streptococci was done; the result was negative, and he was sent home on supportive measures.

On the day of admission, he awoke with severe, pressure-like, midsternal, nonradiating pain, which he rated 10 on a scale of 10. The pain intensified in the supine position and improved with sitting. A complete review of systems was otherwise negative. He denied having had similar symptoms in the past, as well as sick contacts, recent travel, toxin exposure, illicit substance abuse, pets at home, or tick bites. His family history was negative for cardiac arrhythmias, premature coronary artery disease, thoracic aneurysms or dissection, and infiltrative disorders. His surgical and social histories were unremarkable. He said he had no drug allergies.

farid_myocarditis_f1.jpg
%3Cp%3EFigure%201.%20The%20patient%E2%80%99s%20electrocardiogram%20on%20presentation%20shows%20ST-segment%20elevation%20(arrows)%20over%20the%20lateral%20and%20inferior%20distribution%20(V%3Csub%3E4%3C%2Fsub%3E%E2%80%93V%3Csub%3E6%3C%2Fsub%3E%2C%20II%2C%20III%2C%20and%20aVF).%3C%2Fp%3E
An electrocardiogram was obtained (Figure 1). His troponin I level was 7.0 ng/mL (reference range < 0.04 ng/mL).

On examination, his temperature was 38.1°C (100.6°F), heart rate 101 beats per minute, blood pressure 142/78 mm Hg, respiratory rate 16 breaths per minute, and oxygen saturation 98% on room air. He appeared anxious but was in no acute distress. Neck examination showed no elevation in jugular venous pulsation, bruits, thyromegaly, or lymphadenopathy. Cardiac examination revealed tachycardia without murmurs, rubs, or gallops. Lungs were clear to auscultation. Examination of all 4 extremities found 2+ pulses (on a scale of 0 to 4+) throughout and no cyanosis, clubbing, or edema. Abdominal, neurologic, and dermatologic examinations were unremarkable.

Further blood testing revealed the following:

  • Troponin I (3 hours after the first level) 15.5 ng/mL
  • B-type natriuretic peptide 200 mg/dL (reference range 0–100 mg/dL)
  • C-reactive protein 0.9 mg/dL (reference range 0.0–0.8 mg/dL)
  • Erythrocyte sedimentation rate 10 mm/h (reference range < 15 mm/h).

Metabolic and hematologic assessments were unremarkable. A toxicology screen for drugs of abuse was negative. Viral serologic testing was not done.

A chest radiograph showed no acute cardiopulmonary processes.

Given his presenting symptoms, persistent tachycardia, rapidly rising troponin I level, and electrocardiogram showing diffuse ST elevation, he was taken for urgent cardiac catheterization. Coronary angiography revealed no evidence of atherosclerotic disease, acute thrombosis, dissection, or aneurysm. Echocardiography 2 hours after the procedure showed a normal ejection fraction and no regional wall-motion abnormalities or valvular heart disease.

 

 

FURTHER TESTING

1. Which test should be done next to further evaluate this patient’s chest pain?

  • Serum viral serologic testing
  • Serum free light chain assay
  • Nuclear myocardial perfusion study
  • Cardiac magnetic resonance imaging (MRI)
  • Endomyocardial biopsy

In this patient without ischemic coronary disease or valvular heart disease, the recent upper respiratory tract prodrome, active positional chest pain, and diffuse electrocardiographic changes raise the possibility of myocarditis with pericardial involvement.

Viral serologic tests

Viral serologic tests are often obtained in the workup of myocarditis as a noninvasive means of detecting an infectious cause.

However, this approach has several problems. First, a positive serologic result is a signal of the peripheral immune response to a pathogen but does not necessarily indicate active myocardial inflammation. Additionally, circulating immunoglobulin G against cardiotropic viruses is commonly found, even in the absence of myocarditis.1 This is often the result of a high prevalence and exposure to these viruses in the general population. Further, trials have shown no correlation between serologic results and organisms identified by endomyocardial biopsy.2

Thus, serologic testing seems to be of limited utility, reserved for testing for infection with Borrelia burgdorferi (Lyme disease) in endemic areas, hepatitis C virus, human immunodeficiency virus in patients at high risk, Rickettsia conorii, and Rickettsia rickettsii.3

Serum free light chain testing for amyloidosis

Serum free light chain testing is replacing serum and urine protein electrophoresis in the workup of cardiac amyloidosis,4 as electrophoresis has poor sensitivity.4,5

Cardiac amyloidosis often affects older persons, although in rare cases it can affect young patients who carry mutations in the transthyretin gene (ATTR amyloidosis).6 This diagnosis is unlikely in our patient, as he has no other affected organ systems (amyloidosis often affects the renal and neurologic systems), normal QRS voltages on electrocardiography (which are often but not always low in amyloidosis), and no left ventricular hypertrophy or diastolic dysfunction on echocardiography (which are often seen in amyloidosis).4

Nuclear perfusion imaging for sarcoidosis

Nuclear imaging has a limited role in evaluating myocarditis,3 but positron-emission tomography with fluorine-18 fluorodeoxyglucose has a diagnostic role in sarcoidosis, an immune-mediated cause of myocarditis.7

Based on the acuity of the patient’s presentation, preceded by upper respiratory tract symptoms, sarcoidosis is less likely. Sarcoidosis is difficult to diagnose, although when it is the cause of myocarditis, some clues exist, as patients usually present with heart failure symptoms, a second- or third-degree atrioventricular block, or a dilated left ventricle on echocardiography.3 All of these were absent in our patient.

Cardiac MRI

Cardiac MRI has undergone many advances, making it an extremely useful noninvasive test. It has excellent utility as a stand-alone test in diagnosing myocarditis and has synergistic value when combined with endomyocardial biopsy.8 It is indicated in hemodynamically stable patients with a clinical suspicion of myocarditis, persistent symptoms, absence of heart failure, and when imaging findings will change management. It is particularly useful to help elucidate a cause and guide tailored therapy.9 Therefore, it is a reasonable next step in the diagnostic pathway for this patient.10

Cardiac MRI also allows for concurrent assessment of scar. In myocardial infarction, the late gadolinium enhancement is subendocardial or transmural. In myocarditis, the pattern differs, being found in the subepicardial lateral free wall (in most patients with parvovirus B19) and mid-myocardial septum (in most patients with herpesvirus 6).9,11 Cardiac MRI also confers prognostic information for patients with suspected myocarditis.12

The Lake Louise criteria9 for the diagnosis of myocarditis require 2 of the following:

  • Evidence of myocardial edema
  • Increased ratio of early gadolinium enhancement between myocardium and skeletal muscle (indicates hyperemia)
  • At least 1 focal lesion with nonischemic late gadolinium enhancement (indicates cardiac myocyte injury or scarring).

The Lake Louise criteria may be replaced by T1 and T2 mapping, which was found to be considerably better for diagnosing myocarditis when the 2 were compared.9,13,14

Endomyocardial biopsy

Endomyocardial biopsy should not be delayed while waiting for cardiac MRI in patients who are hemodynamically unstable or present with life-threatening features (ventricular arrhythmia, left ventricular failure, or resuscitation after sudden cardiac death).3,10

The indications for endomyocardial biopsy have been highly debated. The 2013 guidelines from the European Society of Cardiology (ESC) recommending endomyocardial biopsy  in all clinically suspected cases of myocarditis have only heightened the controversy.3 The American Heart Association (AHA) guidelines reserve biopsy for patients with suspected myocarditis who have acute or subacute heart failure symptoms or who do not respond to standard medical therapy.15 Other reasonable indications may include the following: myocarditis with life-threatening ventricular arrhythmias, suspicion of giant cell myocarditis, necrotizing eosinophilic myocarditis, or cardiac sarcoidosis.16

Endomyocardial biopsy is the only way to make a definitive diagnosis of myocarditis.3 However, given the patchy distribution of myocardial involvement, a negative result does not rule out myocarditis. The diagnostic utility can be improved by increasing the number of samples taken (at least 3 but up to 10), obtaining samples from both ventricles, and using cardiac MRI data to determine which sites to biopsy.3,13,17,18

Noninvasive testing such as cardiac MRI does not distinguish cell type or etiology (viral vs nonviral).3 Further, endomyocardial biopsy must be performed before immunosuppressive therapy can be safely started.3,16 At experienced centers, the complication rate is 0% to 0.8%.3 The addition of immunohistochemical testing and viral genomic detection by polymerase chain reaction testing have increased the sensitivity of this technique.19 Finally, endomyocardial biopsy can help rule out some of the other possibilities in the differential diagnosis for myocarditis, including infiltrative and storage diseases, and possibly cardiac tumors.3

Of additional note, the diffuse ST-segment elevation seen on the patient’s electrocardiogram (Figure 1) is indicative of subepicardial inflammation. Since the distribution involves more than one epicardial coronary territory, this helps to differentiate the changes from those that occur with myocardial infarction.20

 

 

CASE CONTINUED

farid_myocarditis_f2.jpg
%3Cp%3EFigure%202.%20Cardiac%20magnetic%20resonance%20imaging%20shows%20areas%20of%20patchy%20subepicardial%20late%20gadolinium%20enhancement%20(arrows).%3C%2Fp%3E
The patient underwent cardiac MRI, which showed myocardial edema and patchy areas of late gadolinium enhancement, raising suspicion for myocarditis (Figure 2).

Causes of myocarditis are numerous (Table 1),3,21,22 but viral and postinfectious etiologies remain the most common causes of acute myocarditis.23

farid_myocarditis_t1.jpg
2. What is the most likely causative infectious agent?

  • Parvovirus B19
  • Coxsackievirus B
  • Adenovirus species
  • Human herpesvirus 6
  • Staphylococcus aureus
  • Corynebacterium diphtheria
  • Trypanosoma cruzi
  • Influenza H1/N1

INFECTIOUS CAUSES OF MYOCARDITIS

Coxsackievirus B was the agent most often linked to this condition from the 1950s through the 1990s. However, in the last 2 decades, adenovirus species and human herpesvirus 6 have been increasingly encountered, and recently, parvovirus B19 has been credited as the most common culprit,11,23 at least in the Western world. In developing nations, T cruzi and C diphtheria are the most common offenders.21

S aureus is a common cause of endocarditis, but it rarely plays a role in myocarditis. When it does, the myocarditis is often the sequela of profound bacteremia. This was much more common before antibiotics were invented.24,25

Influenza H1/N1 is not among the most common causes of viral myocarditis, but it should be considered during flu season, given its ability to result in fulminant myocarditis.3,26

TREATMENT FOR MYOCARDITIS

3. Which treatment is the most appropriate at this time?

  • Intravenous immunoglobulin
  • Interferon beta
  • Acyclovir
  • Prednisone
  • Colchicine

Treatment for myocarditis depends on the cause but always includes supportive care to address the constellation of presenting symptoms. Standard therapies for tachy- or bradyarrhythmias, heart failure, and hemodynamic derangement should be started.

Supportive care

In patients with severe left ventricular dysfunction, an implantable cardiac electronic device, left ventricular assist device, or heart transplant may ultimately be needed. However, if possible these should be deferred for several months to determine response to treatment, since the myocardium can possibly recover.16

Diuretics, beta-blockers, angiotensin II receptor blockers, angiotensin-converting enzyme inhibitors, and aldosterone antagonists should be given as part of guideline-directed medical therapy for patients with heart failure and reduced ejection fraction.3,27 However, whether and how the patient should be weaned from these agents after disease recovery are unknown.3

Intravenous immunoglobulin

Intravenous immunoglobulin in high doses has had mixed results. Its efficacy is well documented in children,21 but limited supportive data are available in adults.3 As such, recent ESC guidelines do not provide recommendations regarding its use in adults.3

Interferon beta

Interferon beta has shown promise in improving New York Heart Association class and left ventricular ejection fraction.3 This is attributed to its effects on eliminating adenoviral species and enteroviruses. Treatment of enteroviral organisms in particular has been associated with improved 10-year prognosis.3 Interferon beta also has in vitro data showing efficacy at diminishing apoptosis from parvovirus B19.28

Nucleoside analogues

Empiric treatment with nucleoside analogues (acyclovir, ganciclovir, and valacyclovir) has been tried for patients in whom human herpesvirus is suspected as the causative organism, although with unconfirmed effects.3 Consultation with an infectious disease specialist is recommended before starting these agents, and biopsy is often needed beforehand.3

Immunosuppressive agents

Immunosuppressive agents such as prednisone, azathioprine, and cyclosporine can be used in cases of biopsy-proven disease with manifestations of severe heart failure, especially if biopsy results reveal sarcoidosis, giant cell myocarditis, or necrotizing eosinophilic myocarditis. Although the results were neutral in the Myocarditis Treatment Trial,29 the cause of myocarditis in this trial was unknown. Therapy with such agents should be initiated after active infection is ruled out, which also would require a biopsy.

Colchicine

Mechanisms of chest pain in myocarditis include associated pericarditis and coronary artery vasospasm.3,23 Our patient’s chest pain changed when he changed position, possibly indicating associated pericarditis. In myocarditis with accompanying pericarditis symptoms, colchicine (1–2 mg as an initial dose and then 0.6 mg daily for up to 3 months) can be helpful in alleviating symptoms.21,30 Thus, starting this agent in a patient who presents with myocarditis in absence of heart failure, arrhythmias, or left ventricular dysfunction is prudent.

Colchicine is used mainly to address the pain associated with pericarditis. For patients who present with pericarditis without myocarditis, nonsteroidal anti-inflammatory drugs (NSAIDs) remain the first-line treatment, with the addition of colchicine leading to faster symptom resolution.30 The benefit of colchicine for isolated myocarditis is not well established, with only limited data showing some clinical effects.31

 

 

CASE CONTINUED

The patient was given colchicine 1.2 mg on the first day and then 0.6 mg daily. Within 2 days, his chest pain had resolved. He did not receive any immunosuppressive agents.

DISCHARGE INSTRUCTIONS

4. Before discharge, this patient should be instructed to do which of the following?

  • Take over-the-counter NSAIDs to supplement the effects of colchicine
  • Avoid competitive sports and athletics for at least 6 months
  • Call to schedule repeat cardiac MRI
  • No further instruction is needed

NSAIDs are used by themselves or in combination with colchicine in the treatment of pericarditis, but their use may be associated with worse outcomes in myocarditis.3,21 Thus, their use is not recommended in most cases.3

Excessive physical activity should be avoided for at least 6 months after the clinical syndrome resolves. This recommendation is included in the most recent ESC guidelines but is based mainly on expert opinion and murine models with coxsackievirus B.3 Periodic reassessment is indicated with exercise stress testing before return to strenuous activity.3,16,32 Testing should look for exercise tolerance, and exercise electrocardiography also helps to evaluate for clinically relevant arrythmias.

Cardiac MRI can help clarify the prognosis in myocarditis, but the role of repeat testing in guiding therapy is limited.3 Indications for repeat cardiac MRI include presence of 0 or 1 of the Lake Louise criteria (recall that 2 are necessary to make the diagnosis) with recurrence of symptoms and a high suspicion for myocardial inflammation.3,9 Repeat cardiac MRI was not performed for our patient.

CASE CONCLUDED

The patient was evaluated in the cardiology clinic within 1 week of discharge. At that time, he was in sinus tachycardia with a heart rate of 102 bpm, and he was instructed to avoid any exercise until further notice.

At 6-month follow-up, the sinus tachycardia had resolved. However, because persistent tachycardia had been noted at the first postdischarge visit, and in view of the extent of myocardial involvement, he underwent exercise treadmill testing to evaluate for ventricular arrhythmias. The study did show premature ventricular complexes and 1 ventricular couplet at submaximal exercise levels. As this indicated a higher risk of exercise-induced arrhythmias, he was asked to continue normal activity levels but to abstain from exercise until the next evaluation.

During his 1-year follow-up, a repeat treadmill test showed no ventricular ectopy. Holter monitoring was ordered and showed no premature ventricular complexes, supraventricular arrhythmias, or atrioventricular block within the 48-hour period.

At his 2-year evaluation, he had returned to playing basketball and soccer on weekends and reported no recurrence of his initial symptoms.

KEY POINTS

  • farid_myocarditis_f3.jpg
    %3Cp%3EFigure%203.%20Our%20suggested%20approach%20to%20suspected%20acute%20myocarditis.%3C%2Fp%3E
    Cardiac MRI has emerged as an excellent noninvasive imaging modality for the diagnosis of myocarditis.
  • Treatment of myocarditis depends on the cause and severity of the patient’s presentation, spanning the spectrum from conservative care to immunosuppressive agents and even heart failure therapy.
  • Excessive physical activity should be avoided for the first 6 months after disease diagnosis and treatment.
  • If myocarditis is associated with pericardial involvement, colchicine is the agent of choice, and NSAIDs should be avoided.

Our suggested strategy for approaching myocarditis is shown in Figure 3.

References
  1. Dennert R, Crijns HJ, Heymans S. Acute viral myocarditis. Eur Heart J 2008; 29(17):2073–2082. doi:10.1093/eurheartj/ehn296
  2. Mahfoud F, Gärtner B, Kindermann M, et al. Virus serology in patients with suspected myocarditis: utility or futility? Eur Heart J 2011; 32(7):897–903. doi:10.1093/eurheartj/ehq493
  3. Caforio AL, Pankuweit S, Arbustini E, et al; European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2013; 34(33):2636–2648, 2648a–2648d. doi:10.1093/eurheartj/eht210
  4. Donnelly JP, Hanna M. Cardiac amyloidosis: an update on diagnosis and treatment. Cleve Clin J Med 2017; 84(12 suppl 3):12–26. doi:10.3949/ccjm.84.s3.02
  5. Siddiqi OK, Ruberg FL. Cardiac amyloidosis: an update on pathophysiology, diagnosis, and treatment. Trends Cardiovasc Med 2018; 28(1):10–21. doi:10.1016/j.tcm.2017.07.004
  6. Gertz MA, Benson MD, Dyck PJ, et al. Diagnosis, prognosis, and therapy of transthyretin amyloidosis. J Am Coll Cardiol 2015; 66(21):2451–2466. doi:10.1016/j.jacc.2015.09.075
  7. Blankstein R, Osborne M, Naya M, et al. Cardiac positron emission tomography enhances prognostic assessments of patients with suspected cardiac sarcoidosis. J Am Coll Cardiol 2014; 63(4):329–336. doi:10.1016/j.jacc.2013.09.022
  8. Baccouche H, Mahrholtz H, Meinhardt G, et al. Diagnostic synergy of non-invasive cardiovascular magnetic resonance and invasive endomyocardial biopsy in troponin-positive patients without coronary artery disease. Eur Heart J 2009; 30(23):2869–2879. doi:10.1093/eurheartj/ehp328
  9. Friedrich MG, Sechtem U, Schulz-Menger J, et al; International Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC white paper. J Am Coll Cardiol 2009; 53(17):1475–1487. doi:10.1016/j.jacc.2009.02.007
  10. Kindermann I, Barth C, Mahfoud F, et al. Update on myocarditis. J Am Coll Cardiol 2012; 59(9):779–792. doi:10.1016/j.jacc.2011.09.074
  11. Mahrholdt H, Wagner A, Deluigi CC, et al. Presentation, patterns of myocardial damage, and clinical course of viral myocarditis. Circulation 2006; 114(15):1581–1590. doi:10.1161/CIRCULATIONAHA.105.606509
  12. Gräni C, Eichhorn C, Bière L, et al. Prognostic value of cardiac magnetic resonance tissue characterization in risk stratifying patients with suspected myocarditis. J Am Coll Cardiol 2017; 70(16):1964–1976. doi:10.1016/j.jacc.2017.08.050
  13. Lurz P, Luecke C, Eitel I, et al. Comprehensive cardiac magnetic resonance imaging in patients with suspected myocarditis: the MyoRacer-Trial. J Am Coll Cardiol 2016; 67(15):1800–1811. doi:10.1016/j.jacc.2016.02.013
  14. Gannon MP, Schaub E, Griens CL, Saba SG. State of the art: evaluation and prognostication of myocarditis using cardiac MRI. J Magn Reson Imaging 2019; 49(7):e122–e131. doi:10.1002/jmri.26611
  15. Cooper LT, Baughman KL, Feldman AM, et al. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology endorsed by the Heart Failure Society of America and the Heart Failure Association of the European Society of Cardiology. Eur Heart J 2007; 28(24):3076–3093. doi:10.1093/eurheartj/ehm456
  16. Sinagra G, Anzini M, Pereira NL, et al. Myocarditis in clinical practice. Mayo Clin Proc 2016; 91(9):1256–1266. doi:10.1016/j.mayocp.2016.05.013
  17. Cooper LT, Baughman KL, Feldman AM, et al; American Heart Association; American College of Cardiology; European Society of Cardiology. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology. Circulation 2007; 116(19):2216–2233. doi:10.1161/CIRCULATIONAHA.107.186093
  18. Leone O, Veinot JP, Angelini A, et al. 2011 consensus statement on endomyocardial biopsy from the Association for European Cardiovascular Pathology and the Society for Cardiovascular Pathology. Cardiovasc Pathol 2012; 21(4):245–274. doi:10.1016/j.carpath.2011.10.001
  19. Baughman KL. Diagnosis of myocarditis: death of Dallas criteria. Circulation 2006; 113(4):593–595. doi:10.1161/CIRCULATIONAHA.105.589663
  20. Alraies MC, Klein AL. Should we still use electrocardiography to diagnose pericardial disease? Cleve Clin J Med 2013; 80(2):97–100. doi:10.3949/ccjm.80a.11144
  21. Sagar S, Liu PP, Cooper LT Jr. Myocarditis. Lancet 2012; 379(9817):738–747. doi:10.1016/S0140-6736(11)60648-X
  22. Caforio AL, Marcolongo R, Basso C, Iliceto S. Clinical presentation and diagnosis of myocarditis. Heart 2015; 101(16):1332–1344. doi:10.1136/heartjnl-2014-306363
  23. Cooper LT Jr. Myocarditis. N Engl J Med 2009; 360(15):1526–1538. doi:10.1056/NEJMra0800028
  24. LeLeiko RM, Bower DJ, Larsen CP. MRSA-associated bacterial myocarditis causing ruptured ventricle and tamponade. Cardiology 2008; 111(3):188–190. doi:10.1159/000121602
  25. Wasi F, Shuter J. Primary bacterial infection of the myocardium. Front Biosci 2003; 8:s228–s231. pmid:12700039
  26. Al-Amoodi M, Rao K, Rao S, Brewer JH, Magalski A, Chhatriwalla AK. Fulminant myocarditis due to H1N1 influenza. Circ Heart Fail 2010; 3(3):e7–e9. doi:10.1161/CIRCHEARTFAILURE.110.938506
  27. Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA focused update on new pharmacological therapy for heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. J Am Coll Cardiol 2016; 68(13):1476–1488. doi:10.1016/j.jacc.2016.05.011
  28. Schmidt-Lucke C, Spillmann F, Bock T, et al. Interferon beta modulates endothelial damage in patients with cardiac persistence of human parvovirus b19 infection. J Infect Dis 2010; 201(6):936–945. doi:10.1086/650700
  29. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis: the Myocarditis Treatment Trial Investigators. N Engl J Med 1995; 333(5):269–275. doi:10.1056/NEJM199508033330501
  30. Imazio M, Bobbio M, Cecchi E, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the COlchicine for acute PEricarditis (COPE) trial. Circulation 2005; 112(13):2012–2016. doi:10.1161/CIRCULATIONAHA.105.542738
  31. Morgenstern D, Lisko J, Boniface NC, Mikolich BM, Mikolich JR. Myocarditis and colchicine: a new perspective from cardiac MRI. J Cardiovasc Magn Reson 2016; 18(suppl 1):0100.
  32. Maron BJ, Zipes DP, Kovacs RJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: preamble, principles, and general considerations: a scientific statement from the American Heart Association and American College of Cardiology. J Am Coll Cardiol 2015; 66(21):2343–2349. doi:10.1016/j.jacc.2015.09.032
Article PDF
Author and Disclosure Information

Amir Farid, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Neil Beri, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

David Torres-Barba, MD, PhD
Department of Cardiology, University of California San Diego

Charles Whitcomb, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Address: David Torres-Barba, MD, PhD, Department of Internal Medicine, University of California, Davis, 4150 V. Street, Sacramento, CA 95817; davidtorresbarba@gmail.com

Issue
Cleveland Clinic Journal of Medicine - 86(9)
Publications
Topics
Page Number
586-594
Legacy Keywords
chest pain, angina, myocarditis, pericarditis, ST-segment elevation, serologic testing, light chain, myocardial perfusion, magnetic resonance imaging, MRI, biopsy, amyloidosis, sarcoidosis, parvovirus B19, colchicine, Amir Farid, Neil Beri, David Torres-Barba, Charles Whitcomb
Sections
Author and Disclosure Information

Amir Farid, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Neil Beri, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

David Torres-Barba, MD, PhD
Department of Cardiology, University of California San Diego

Charles Whitcomb, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Address: David Torres-Barba, MD, PhD, Department of Internal Medicine, University of California, Davis, 4150 V. Street, Sacramento, CA 95817; davidtorresbarba@gmail.com

Author and Disclosure Information

Amir Farid, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Neil Beri, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

David Torres-Barba, MD, PhD
Department of Cardiology, University of California San Diego

Charles Whitcomb, MD
Department of Cardiology, University of California Davis Medical Center, Sacramento

Address: David Torres-Barba, MD, PhD, Department of Internal Medicine, University of California, Davis, 4150 V. Street, Sacramento, CA 95817; davidtorresbarba@gmail.com

Article PDF
Article PDF
Related Articles

An 18-year-old man without any significant medical history was transferred from another hospital for higher-level care after presenting with unremitting chest pain. He had been in his usual state of good health until 7 days before presentation, when he developed mild rhinorrhea and a sore throat, but not a cough. He went to an outpatient clinic, where a rapid test for group A streptococci was done; the result was negative, and he was sent home on supportive measures.

On the day of admission, he awoke with severe, pressure-like, midsternal, nonradiating pain, which he rated 10 on a scale of 10. The pain intensified in the supine position and improved with sitting. A complete review of systems was otherwise negative. He denied having had similar symptoms in the past, as well as sick contacts, recent travel, toxin exposure, illicit substance abuse, pets at home, or tick bites. His family history was negative for cardiac arrhythmias, premature coronary artery disease, thoracic aneurysms or dissection, and infiltrative disorders. His surgical and social histories were unremarkable. He said he had no drug allergies.

farid_myocarditis_f1.jpg
%3Cp%3EFigure%201.%20The%20patient%E2%80%99s%20electrocardiogram%20on%20presentation%20shows%20ST-segment%20elevation%20(arrows)%20over%20the%20lateral%20and%20inferior%20distribution%20(V%3Csub%3E4%3C%2Fsub%3E%E2%80%93V%3Csub%3E6%3C%2Fsub%3E%2C%20II%2C%20III%2C%20and%20aVF).%3C%2Fp%3E
An electrocardiogram was obtained (Figure 1). His troponin I level was 7.0 ng/mL (reference range < 0.04 ng/mL).

On examination, his temperature was 38.1°C (100.6°F), heart rate 101 beats per minute, blood pressure 142/78 mm Hg, respiratory rate 16 breaths per minute, and oxygen saturation 98% on room air. He appeared anxious but was in no acute distress. Neck examination showed no elevation in jugular venous pulsation, bruits, thyromegaly, or lymphadenopathy. Cardiac examination revealed tachycardia without murmurs, rubs, or gallops. Lungs were clear to auscultation. Examination of all 4 extremities found 2+ pulses (on a scale of 0 to 4+) throughout and no cyanosis, clubbing, or edema. Abdominal, neurologic, and dermatologic examinations were unremarkable.

Further blood testing revealed the following:

  • Troponin I (3 hours after the first level) 15.5 ng/mL
  • B-type natriuretic peptide 200 mg/dL (reference range 0–100 mg/dL)
  • C-reactive protein 0.9 mg/dL (reference range 0.0–0.8 mg/dL)
  • Erythrocyte sedimentation rate 10 mm/h (reference range < 15 mm/h).

Metabolic and hematologic assessments were unremarkable. A toxicology screen for drugs of abuse was negative. Viral serologic testing was not done.

A chest radiograph showed no acute cardiopulmonary processes.

Given his presenting symptoms, persistent tachycardia, rapidly rising troponin I level, and electrocardiogram showing diffuse ST elevation, he was taken for urgent cardiac catheterization. Coronary angiography revealed no evidence of atherosclerotic disease, acute thrombosis, dissection, or aneurysm. Echocardiography 2 hours after the procedure showed a normal ejection fraction and no regional wall-motion abnormalities or valvular heart disease.

 

 

FURTHER TESTING

1. Which test should be done next to further evaluate this patient’s chest pain?

  • Serum viral serologic testing
  • Serum free light chain assay
  • Nuclear myocardial perfusion study
  • Cardiac magnetic resonance imaging (MRI)
  • Endomyocardial biopsy

In this patient without ischemic coronary disease or valvular heart disease, the recent upper respiratory tract prodrome, active positional chest pain, and diffuse electrocardiographic changes raise the possibility of myocarditis with pericardial involvement.

Viral serologic tests

Viral serologic tests are often obtained in the workup of myocarditis as a noninvasive means of detecting an infectious cause.

However, this approach has several problems. First, a positive serologic result is a signal of the peripheral immune response to a pathogen but does not necessarily indicate active myocardial inflammation. Additionally, circulating immunoglobulin G against cardiotropic viruses is commonly found, even in the absence of myocarditis.1 This is often the result of a high prevalence and exposure to these viruses in the general population. Further, trials have shown no correlation between serologic results and organisms identified by endomyocardial biopsy.2

Thus, serologic testing seems to be of limited utility, reserved for testing for infection with Borrelia burgdorferi (Lyme disease) in endemic areas, hepatitis C virus, human immunodeficiency virus in patients at high risk, Rickettsia conorii, and Rickettsia rickettsii.3

Serum free light chain testing for amyloidosis

Serum free light chain testing is replacing serum and urine protein electrophoresis in the workup of cardiac amyloidosis,4 as electrophoresis has poor sensitivity.4,5

Cardiac amyloidosis often affects older persons, although in rare cases it can affect young patients who carry mutations in the transthyretin gene (ATTR amyloidosis).6 This diagnosis is unlikely in our patient, as he has no other affected organ systems (amyloidosis often affects the renal and neurologic systems), normal QRS voltages on electrocardiography (which are often but not always low in amyloidosis), and no left ventricular hypertrophy or diastolic dysfunction on echocardiography (which are often seen in amyloidosis).4

Nuclear perfusion imaging for sarcoidosis

Nuclear imaging has a limited role in evaluating myocarditis,3 but positron-emission tomography with fluorine-18 fluorodeoxyglucose has a diagnostic role in sarcoidosis, an immune-mediated cause of myocarditis.7

Based on the acuity of the patient’s presentation, preceded by upper respiratory tract symptoms, sarcoidosis is less likely. Sarcoidosis is difficult to diagnose, although when it is the cause of myocarditis, some clues exist, as patients usually present with heart failure symptoms, a second- or third-degree atrioventricular block, or a dilated left ventricle on echocardiography.3 All of these were absent in our patient.

Cardiac MRI

Cardiac MRI has undergone many advances, making it an extremely useful noninvasive test. It has excellent utility as a stand-alone test in diagnosing myocarditis and has synergistic value when combined with endomyocardial biopsy.8 It is indicated in hemodynamically stable patients with a clinical suspicion of myocarditis, persistent symptoms, absence of heart failure, and when imaging findings will change management. It is particularly useful to help elucidate a cause and guide tailored therapy.9 Therefore, it is a reasonable next step in the diagnostic pathway for this patient.10

Cardiac MRI also allows for concurrent assessment of scar. In myocardial infarction, the late gadolinium enhancement is subendocardial or transmural. In myocarditis, the pattern differs, being found in the subepicardial lateral free wall (in most patients with parvovirus B19) and mid-myocardial septum (in most patients with herpesvirus 6).9,11 Cardiac MRI also confers prognostic information for patients with suspected myocarditis.12

The Lake Louise criteria9 for the diagnosis of myocarditis require 2 of the following:

  • Evidence of myocardial edema
  • Increased ratio of early gadolinium enhancement between myocardium and skeletal muscle (indicates hyperemia)
  • At least 1 focal lesion with nonischemic late gadolinium enhancement (indicates cardiac myocyte injury or scarring).

The Lake Louise criteria may be replaced by T1 and T2 mapping, which was found to be considerably better for diagnosing myocarditis when the 2 were compared.9,13,14

Endomyocardial biopsy

Endomyocardial biopsy should not be delayed while waiting for cardiac MRI in patients who are hemodynamically unstable or present with life-threatening features (ventricular arrhythmia, left ventricular failure, or resuscitation after sudden cardiac death).3,10

The indications for endomyocardial biopsy have been highly debated. The 2013 guidelines from the European Society of Cardiology (ESC) recommending endomyocardial biopsy  in all clinically suspected cases of myocarditis have only heightened the controversy.3 The American Heart Association (AHA) guidelines reserve biopsy for patients with suspected myocarditis who have acute or subacute heart failure symptoms or who do not respond to standard medical therapy.15 Other reasonable indications may include the following: myocarditis with life-threatening ventricular arrhythmias, suspicion of giant cell myocarditis, necrotizing eosinophilic myocarditis, or cardiac sarcoidosis.16

Endomyocardial biopsy is the only way to make a definitive diagnosis of myocarditis.3 However, given the patchy distribution of myocardial involvement, a negative result does not rule out myocarditis. The diagnostic utility can be improved by increasing the number of samples taken (at least 3 but up to 10), obtaining samples from both ventricles, and using cardiac MRI data to determine which sites to biopsy.3,13,17,18

Noninvasive testing such as cardiac MRI does not distinguish cell type or etiology (viral vs nonviral).3 Further, endomyocardial biopsy must be performed before immunosuppressive therapy can be safely started.3,16 At experienced centers, the complication rate is 0% to 0.8%.3 The addition of immunohistochemical testing and viral genomic detection by polymerase chain reaction testing have increased the sensitivity of this technique.19 Finally, endomyocardial biopsy can help rule out some of the other possibilities in the differential diagnosis for myocarditis, including infiltrative and storage diseases, and possibly cardiac tumors.3

Of additional note, the diffuse ST-segment elevation seen on the patient’s electrocardiogram (Figure 1) is indicative of subepicardial inflammation. Since the distribution involves more than one epicardial coronary territory, this helps to differentiate the changes from those that occur with myocardial infarction.20

 

 

CASE CONTINUED

farid_myocarditis_f2.jpg
%3Cp%3EFigure%202.%20Cardiac%20magnetic%20resonance%20imaging%20shows%20areas%20of%20patchy%20subepicardial%20late%20gadolinium%20enhancement%20(arrows).%3C%2Fp%3E
The patient underwent cardiac MRI, which showed myocardial edema and patchy areas of late gadolinium enhancement, raising suspicion for myocarditis (Figure 2).

Causes of myocarditis are numerous (Table 1),3,21,22 but viral and postinfectious etiologies remain the most common causes of acute myocarditis.23

farid_myocarditis_t1.jpg
2. What is the most likely causative infectious agent?

  • Parvovirus B19
  • Coxsackievirus B
  • Adenovirus species
  • Human herpesvirus 6
  • Staphylococcus aureus
  • Corynebacterium diphtheria
  • Trypanosoma cruzi
  • Influenza H1/N1

INFECTIOUS CAUSES OF MYOCARDITIS

Coxsackievirus B was the agent most often linked to this condition from the 1950s through the 1990s. However, in the last 2 decades, adenovirus species and human herpesvirus 6 have been increasingly encountered, and recently, parvovirus B19 has been credited as the most common culprit,11,23 at least in the Western world. In developing nations, T cruzi and C diphtheria are the most common offenders.21

S aureus is a common cause of endocarditis, but it rarely plays a role in myocarditis. When it does, the myocarditis is often the sequela of profound bacteremia. This was much more common before antibiotics were invented.24,25

Influenza H1/N1 is not among the most common causes of viral myocarditis, but it should be considered during flu season, given its ability to result in fulminant myocarditis.3,26

TREATMENT FOR MYOCARDITIS

3. Which treatment is the most appropriate at this time?

  • Intravenous immunoglobulin
  • Interferon beta
  • Acyclovir
  • Prednisone
  • Colchicine

Treatment for myocarditis depends on the cause but always includes supportive care to address the constellation of presenting symptoms. Standard therapies for tachy- or bradyarrhythmias, heart failure, and hemodynamic derangement should be started.

Supportive care

In patients with severe left ventricular dysfunction, an implantable cardiac electronic device, left ventricular assist device, or heart transplant may ultimately be needed. However, if possible these should be deferred for several months to determine response to treatment, since the myocardium can possibly recover.16

Diuretics, beta-blockers, angiotensin II receptor blockers, angiotensin-converting enzyme inhibitors, and aldosterone antagonists should be given as part of guideline-directed medical therapy for patients with heart failure and reduced ejection fraction.3,27 However, whether and how the patient should be weaned from these agents after disease recovery are unknown.3

Intravenous immunoglobulin

Intravenous immunoglobulin in high doses has had mixed results. Its efficacy is well documented in children,21 but limited supportive data are available in adults.3 As such, recent ESC guidelines do not provide recommendations regarding its use in adults.3

Interferon beta

Interferon beta has shown promise in improving New York Heart Association class and left ventricular ejection fraction.3 This is attributed to its effects on eliminating adenoviral species and enteroviruses. Treatment of enteroviral organisms in particular has been associated with improved 10-year prognosis.3 Interferon beta also has in vitro data showing efficacy at diminishing apoptosis from parvovirus B19.28

Nucleoside analogues

Empiric treatment with nucleoside analogues (acyclovir, ganciclovir, and valacyclovir) has been tried for patients in whom human herpesvirus is suspected as the causative organism, although with unconfirmed effects.3 Consultation with an infectious disease specialist is recommended before starting these agents, and biopsy is often needed beforehand.3

Immunosuppressive agents

Immunosuppressive agents such as prednisone, azathioprine, and cyclosporine can be used in cases of biopsy-proven disease with manifestations of severe heart failure, especially if biopsy results reveal sarcoidosis, giant cell myocarditis, or necrotizing eosinophilic myocarditis. Although the results were neutral in the Myocarditis Treatment Trial,29 the cause of myocarditis in this trial was unknown. Therapy with such agents should be initiated after active infection is ruled out, which also would require a biopsy.

Colchicine

Mechanisms of chest pain in myocarditis include associated pericarditis and coronary artery vasospasm.3,23 Our patient’s chest pain changed when he changed position, possibly indicating associated pericarditis. In myocarditis with accompanying pericarditis symptoms, colchicine (1–2 mg as an initial dose and then 0.6 mg daily for up to 3 months) can be helpful in alleviating symptoms.21,30 Thus, starting this agent in a patient who presents with myocarditis in absence of heart failure, arrhythmias, or left ventricular dysfunction is prudent.

Colchicine is used mainly to address the pain associated with pericarditis. For patients who present with pericarditis without myocarditis, nonsteroidal anti-inflammatory drugs (NSAIDs) remain the first-line treatment, with the addition of colchicine leading to faster symptom resolution.30 The benefit of colchicine for isolated myocarditis is not well established, with only limited data showing some clinical effects.31

 

 

CASE CONTINUED

The patient was given colchicine 1.2 mg on the first day and then 0.6 mg daily. Within 2 days, his chest pain had resolved. He did not receive any immunosuppressive agents.

DISCHARGE INSTRUCTIONS

4. Before discharge, this patient should be instructed to do which of the following?

  • Take over-the-counter NSAIDs to supplement the effects of colchicine
  • Avoid competitive sports and athletics for at least 6 months
  • Call to schedule repeat cardiac MRI
  • No further instruction is needed

NSAIDs are used by themselves or in combination with colchicine in the treatment of pericarditis, but their use may be associated with worse outcomes in myocarditis.3,21 Thus, their use is not recommended in most cases.3

Excessive physical activity should be avoided for at least 6 months after the clinical syndrome resolves. This recommendation is included in the most recent ESC guidelines but is based mainly on expert opinion and murine models with coxsackievirus B.3 Periodic reassessment is indicated with exercise stress testing before return to strenuous activity.3,16,32 Testing should look for exercise tolerance, and exercise electrocardiography also helps to evaluate for clinically relevant arrythmias.

Cardiac MRI can help clarify the prognosis in myocarditis, but the role of repeat testing in guiding therapy is limited.3 Indications for repeat cardiac MRI include presence of 0 or 1 of the Lake Louise criteria (recall that 2 are necessary to make the diagnosis) with recurrence of symptoms and a high suspicion for myocardial inflammation.3,9 Repeat cardiac MRI was not performed for our patient.

CASE CONCLUDED

The patient was evaluated in the cardiology clinic within 1 week of discharge. At that time, he was in sinus tachycardia with a heart rate of 102 bpm, and he was instructed to avoid any exercise until further notice.

At 6-month follow-up, the sinus tachycardia had resolved. However, because persistent tachycardia had been noted at the first postdischarge visit, and in view of the extent of myocardial involvement, he underwent exercise treadmill testing to evaluate for ventricular arrhythmias. The study did show premature ventricular complexes and 1 ventricular couplet at submaximal exercise levels. As this indicated a higher risk of exercise-induced arrhythmias, he was asked to continue normal activity levels but to abstain from exercise until the next evaluation.

During his 1-year follow-up, a repeat treadmill test showed no ventricular ectopy. Holter monitoring was ordered and showed no premature ventricular complexes, supraventricular arrhythmias, or atrioventricular block within the 48-hour period.

At his 2-year evaluation, he had returned to playing basketball and soccer on weekends and reported no recurrence of his initial symptoms.

KEY POINTS

  • farid_myocarditis_f3.jpg
    %3Cp%3EFigure%203.%20Our%20suggested%20approach%20to%20suspected%20acute%20myocarditis.%3C%2Fp%3E
    Cardiac MRI has emerged as an excellent noninvasive imaging modality for the diagnosis of myocarditis.
  • Treatment of myocarditis depends on the cause and severity of the patient’s presentation, spanning the spectrum from conservative care to immunosuppressive agents and even heart failure therapy.
  • Excessive physical activity should be avoided for the first 6 months after disease diagnosis and treatment.
  • If myocarditis is associated with pericardial involvement, colchicine is the agent of choice, and NSAIDs should be avoided.

Our suggested strategy for approaching myocarditis is shown in Figure 3.

An 18-year-old man without any significant medical history was transferred from another hospital for higher-level care after presenting with unremitting chest pain. He had been in his usual state of good health until 7 days before presentation, when he developed mild rhinorrhea and a sore throat, but not a cough. He went to an outpatient clinic, where a rapid test for group A streptococci was done; the result was negative, and he was sent home on supportive measures.

On the day of admission, he awoke with severe, pressure-like, midsternal, nonradiating pain, which he rated 10 on a scale of 10. The pain intensified in the supine position and improved with sitting. A complete review of systems was otherwise negative. He denied having had similar symptoms in the past, as well as sick contacts, recent travel, toxin exposure, illicit substance abuse, pets at home, or tick bites. His family history was negative for cardiac arrhythmias, premature coronary artery disease, thoracic aneurysms or dissection, and infiltrative disorders. His surgical and social histories were unremarkable. He said he had no drug allergies.

farid_myocarditis_f1.jpg
%3Cp%3EFigure%201.%20The%20patient%E2%80%99s%20electrocardiogram%20on%20presentation%20shows%20ST-segment%20elevation%20(arrows)%20over%20the%20lateral%20and%20inferior%20distribution%20(V%3Csub%3E4%3C%2Fsub%3E%E2%80%93V%3Csub%3E6%3C%2Fsub%3E%2C%20II%2C%20III%2C%20and%20aVF).%3C%2Fp%3E
An electrocardiogram was obtained (Figure 1). His troponin I level was 7.0 ng/mL (reference range < 0.04 ng/mL).

On examination, his temperature was 38.1°C (100.6°F), heart rate 101 beats per minute, blood pressure 142/78 mm Hg, respiratory rate 16 breaths per minute, and oxygen saturation 98% on room air. He appeared anxious but was in no acute distress. Neck examination showed no elevation in jugular venous pulsation, bruits, thyromegaly, or lymphadenopathy. Cardiac examination revealed tachycardia without murmurs, rubs, or gallops. Lungs were clear to auscultation. Examination of all 4 extremities found 2+ pulses (on a scale of 0 to 4+) throughout and no cyanosis, clubbing, or edema. Abdominal, neurologic, and dermatologic examinations were unremarkable.

Further blood testing revealed the following:

  • Troponin I (3 hours after the first level) 15.5 ng/mL
  • B-type natriuretic peptide 200 mg/dL (reference range 0–100 mg/dL)
  • C-reactive protein 0.9 mg/dL (reference range 0.0–0.8 mg/dL)
  • Erythrocyte sedimentation rate 10 mm/h (reference range < 15 mm/h).

Metabolic and hematologic assessments were unremarkable. A toxicology screen for drugs of abuse was negative. Viral serologic testing was not done.

A chest radiograph showed no acute cardiopulmonary processes.

Given his presenting symptoms, persistent tachycardia, rapidly rising troponin I level, and electrocardiogram showing diffuse ST elevation, he was taken for urgent cardiac catheterization. Coronary angiography revealed no evidence of atherosclerotic disease, acute thrombosis, dissection, or aneurysm. Echocardiography 2 hours after the procedure showed a normal ejection fraction and no regional wall-motion abnormalities or valvular heart disease.

 

 

FURTHER TESTING

1. Which test should be done next to further evaluate this patient’s chest pain?

  • Serum viral serologic testing
  • Serum free light chain assay
  • Nuclear myocardial perfusion study
  • Cardiac magnetic resonance imaging (MRI)
  • Endomyocardial biopsy

In this patient without ischemic coronary disease or valvular heart disease, the recent upper respiratory tract prodrome, active positional chest pain, and diffuse electrocardiographic changes raise the possibility of myocarditis with pericardial involvement.

Viral serologic tests

Viral serologic tests are often obtained in the workup of myocarditis as a noninvasive means of detecting an infectious cause.

However, this approach has several problems. First, a positive serologic result is a signal of the peripheral immune response to a pathogen but does not necessarily indicate active myocardial inflammation. Additionally, circulating immunoglobulin G against cardiotropic viruses is commonly found, even in the absence of myocarditis.1 This is often the result of a high prevalence and exposure to these viruses in the general population. Further, trials have shown no correlation between serologic results and organisms identified by endomyocardial biopsy.2

Thus, serologic testing seems to be of limited utility, reserved for testing for infection with Borrelia burgdorferi (Lyme disease) in endemic areas, hepatitis C virus, human immunodeficiency virus in patients at high risk, Rickettsia conorii, and Rickettsia rickettsii.3

Serum free light chain testing for amyloidosis

Serum free light chain testing is replacing serum and urine protein electrophoresis in the workup of cardiac amyloidosis,4 as electrophoresis has poor sensitivity.4,5

Cardiac amyloidosis often affects older persons, although in rare cases it can affect young patients who carry mutations in the transthyretin gene (ATTR amyloidosis).6 This diagnosis is unlikely in our patient, as he has no other affected organ systems (amyloidosis often affects the renal and neurologic systems), normal QRS voltages on electrocardiography (which are often but not always low in amyloidosis), and no left ventricular hypertrophy or diastolic dysfunction on echocardiography (which are often seen in amyloidosis).4

Nuclear perfusion imaging for sarcoidosis

Nuclear imaging has a limited role in evaluating myocarditis,3 but positron-emission tomography with fluorine-18 fluorodeoxyglucose has a diagnostic role in sarcoidosis, an immune-mediated cause of myocarditis.7

Based on the acuity of the patient’s presentation, preceded by upper respiratory tract symptoms, sarcoidosis is less likely. Sarcoidosis is difficult to diagnose, although when it is the cause of myocarditis, some clues exist, as patients usually present with heart failure symptoms, a second- or third-degree atrioventricular block, or a dilated left ventricle on echocardiography.3 All of these were absent in our patient.

Cardiac MRI

Cardiac MRI has undergone many advances, making it an extremely useful noninvasive test. It has excellent utility as a stand-alone test in diagnosing myocarditis and has synergistic value when combined with endomyocardial biopsy.8 It is indicated in hemodynamically stable patients with a clinical suspicion of myocarditis, persistent symptoms, absence of heart failure, and when imaging findings will change management. It is particularly useful to help elucidate a cause and guide tailored therapy.9 Therefore, it is a reasonable next step in the diagnostic pathway for this patient.10

Cardiac MRI also allows for concurrent assessment of scar. In myocardial infarction, the late gadolinium enhancement is subendocardial or transmural. In myocarditis, the pattern differs, being found in the subepicardial lateral free wall (in most patients with parvovirus B19) and mid-myocardial septum (in most patients with herpesvirus 6).9,11 Cardiac MRI also confers prognostic information for patients with suspected myocarditis.12

The Lake Louise criteria9 for the diagnosis of myocarditis require 2 of the following:

  • Evidence of myocardial edema
  • Increased ratio of early gadolinium enhancement between myocardium and skeletal muscle (indicates hyperemia)
  • At least 1 focal lesion with nonischemic late gadolinium enhancement (indicates cardiac myocyte injury or scarring).

The Lake Louise criteria may be replaced by T1 and T2 mapping, which was found to be considerably better for diagnosing myocarditis when the 2 were compared.9,13,14

Endomyocardial biopsy

Endomyocardial biopsy should not be delayed while waiting for cardiac MRI in patients who are hemodynamically unstable or present with life-threatening features (ventricular arrhythmia, left ventricular failure, or resuscitation after sudden cardiac death).3,10

The indications for endomyocardial biopsy have been highly debated. The 2013 guidelines from the European Society of Cardiology (ESC) recommending endomyocardial biopsy  in all clinically suspected cases of myocarditis have only heightened the controversy.3 The American Heart Association (AHA) guidelines reserve biopsy for patients with suspected myocarditis who have acute or subacute heart failure symptoms or who do not respond to standard medical therapy.15 Other reasonable indications may include the following: myocarditis with life-threatening ventricular arrhythmias, suspicion of giant cell myocarditis, necrotizing eosinophilic myocarditis, or cardiac sarcoidosis.16

Endomyocardial biopsy is the only way to make a definitive diagnosis of myocarditis.3 However, given the patchy distribution of myocardial involvement, a negative result does not rule out myocarditis. The diagnostic utility can be improved by increasing the number of samples taken (at least 3 but up to 10), obtaining samples from both ventricles, and using cardiac MRI data to determine which sites to biopsy.3,13,17,18

Noninvasive testing such as cardiac MRI does not distinguish cell type or etiology (viral vs nonviral).3 Further, endomyocardial biopsy must be performed before immunosuppressive therapy can be safely started.3,16 At experienced centers, the complication rate is 0% to 0.8%.3 The addition of immunohistochemical testing and viral genomic detection by polymerase chain reaction testing have increased the sensitivity of this technique.19 Finally, endomyocardial biopsy can help rule out some of the other possibilities in the differential diagnosis for myocarditis, including infiltrative and storage diseases, and possibly cardiac tumors.3

Of additional note, the diffuse ST-segment elevation seen on the patient’s electrocardiogram (Figure 1) is indicative of subepicardial inflammation. Since the distribution involves more than one epicardial coronary territory, this helps to differentiate the changes from those that occur with myocardial infarction.20

 

 

CASE CONTINUED

farid_myocarditis_f2.jpg
%3Cp%3EFigure%202.%20Cardiac%20magnetic%20resonance%20imaging%20shows%20areas%20of%20patchy%20subepicardial%20late%20gadolinium%20enhancement%20(arrows).%3C%2Fp%3E
The patient underwent cardiac MRI, which showed myocardial edema and patchy areas of late gadolinium enhancement, raising suspicion for myocarditis (Figure 2).

Causes of myocarditis are numerous (Table 1),3,21,22 but viral and postinfectious etiologies remain the most common causes of acute myocarditis.23

farid_myocarditis_t1.jpg
2. What is the most likely causative infectious agent?

  • Parvovirus B19
  • Coxsackievirus B
  • Adenovirus species
  • Human herpesvirus 6
  • Staphylococcus aureus
  • Corynebacterium diphtheria
  • Trypanosoma cruzi
  • Influenza H1/N1

INFECTIOUS CAUSES OF MYOCARDITIS

Coxsackievirus B was the agent most often linked to this condition from the 1950s through the 1990s. However, in the last 2 decades, adenovirus species and human herpesvirus 6 have been increasingly encountered, and recently, parvovirus B19 has been credited as the most common culprit,11,23 at least in the Western world. In developing nations, T cruzi and C diphtheria are the most common offenders.21

S aureus is a common cause of endocarditis, but it rarely plays a role in myocarditis. When it does, the myocarditis is often the sequela of profound bacteremia. This was much more common before antibiotics were invented.24,25

Influenza H1/N1 is not among the most common causes of viral myocarditis, but it should be considered during flu season, given its ability to result in fulminant myocarditis.3,26

TREATMENT FOR MYOCARDITIS

3. Which treatment is the most appropriate at this time?

  • Intravenous immunoglobulin
  • Interferon beta
  • Acyclovir
  • Prednisone
  • Colchicine

Treatment for myocarditis depends on the cause but always includes supportive care to address the constellation of presenting symptoms. Standard therapies for tachy- or bradyarrhythmias, heart failure, and hemodynamic derangement should be started.

Supportive care

In patients with severe left ventricular dysfunction, an implantable cardiac electronic device, left ventricular assist device, or heart transplant may ultimately be needed. However, if possible these should be deferred for several months to determine response to treatment, since the myocardium can possibly recover.16

Diuretics, beta-blockers, angiotensin II receptor blockers, angiotensin-converting enzyme inhibitors, and aldosterone antagonists should be given as part of guideline-directed medical therapy for patients with heart failure and reduced ejection fraction.3,27 However, whether and how the patient should be weaned from these agents after disease recovery are unknown.3

Intravenous immunoglobulin

Intravenous immunoglobulin in high doses has had mixed results. Its efficacy is well documented in children,21 but limited supportive data are available in adults.3 As such, recent ESC guidelines do not provide recommendations regarding its use in adults.3

Interferon beta

Interferon beta has shown promise in improving New York Heart Association class and left ventricular ejection fraction.3 This is attributed to its effects on eliminating adenoviral species and enteroviruses. Treatment of enteroviral organisms in particular has been associated with improved 10-year prognosis.3 Interferon beta also has in vitro data showing efficacy at diminishing apoptosis from parvovirus B19.28

Nucleoside analogues

Empiric treatment with nucleoside analogues (acyclovir, ganciclovir, and valacyclovir) has been tried for patients in whom human herpesvirus is suspected as the causative organism, although with unconfirmed effects.3 Consultation with an infectious disease specialist is recommended before starting these agents, and biopsy is often needed beforehand.3

Immunosuppressive agents

Immunosuppressive agents such as prednisone, azathioprine, and cyclosporine can be used in cases of biopsy-proven disease with manifestations of severe heart failure, especially if biopsy results reveal sarcoidosis, giant cell myocarditis, or necrotizing eosinophilic myocarditis. Although the results were neutral in the Myocarditis Treatment Trial,29 the cause of myocarditis in this trial was unknown. Therapy with such agents should be initiated after active infection is ruled out, which also would require a biopsy.

Colchicine

Mechanisms of chest pain in myocarditis include associated pericarditis and coronary artery vasospasm.3,23 Our patient’s chest pain changed when he changed position, possibly indicating associated pericarditis. In myocarditis with accompanying pericarditis symptoms, colchicine (1–2 mg as an initial dose and then 0.6 mg daily for up to 3 months) can be helpful in alleviating symptoms.21,30 Thus, starting this agent in a patient who presents with myocarditis in absence of heart failure, arrhythmias, or left ventricular dysfunction is prudent.

Colchicine is used mainly to address the pain associated with pericarditis. For patients who present with pericarditis without myocarditis, nonsteroidal anti-inflammatory drugs (NSAIDs) remain the first-line treatment, with the addition of colchicine leading to faster symptom resolution.30 The benefit of colchicine for isolated myocarditis is not well established, with only limited data showing some clinical effects.31

 

 

CASE CONTINUED

The patient was given colchicine 1.2 mg on the first day and then 0.6 mg daily. Within 2 days, his chest pain had resolved. He did not receive any immunosuppressive agents.

DISCHARGE INSTRUCTIONS

4. Before discharge, this patient should be instructed to do which of the following?

  • Take over-the-counter NSAIDs to supplement the effects of colchicine
  • Avoid competitive sports and athletics for at least 6 months
  • Call to schedule repeat cardiac MRI
  • No further instruction is needed

NSAIDs are used by themselves or in combination with colchicine in the treatment of pericarditis, but their use may be associated with worse outcomes in myocarditis.3,21 Thus, their use is not recommended in most cases.3

Excessive physical activity should be avoided for at least 6 months after the clinical syndrome resolves. This recommendation is included in the most recent ESC guidelines but is based mainly on expert opinion and murine models with coxsackievirus B.3 Periodic reassessment is indicated with exercise stress testing before return to strenuous activity.3,16,32 Testing should look for exercise tolerance, and exercise electrocardiography also helps to evaluate for clinically relevant arrythmias.

Cardiac MRI can help clarify the prognosis in myocarditis, but the role of repeat testing in guiding therapy is limited.3 Indications for repeat cardiac MRI include presence of 0 or 1 of the Lake Louise criteria (recall that 2 are necessary to make the diagnosis) with recurrence of symptoms and a high suspicion for myocardial inflammation.3,9 Repeat cardiac MRI was not performed for our patient.

CASE CONCLUDED

The patient was evaluated in the cardiology clinic within 1 week of discharge. At that time, he was in sinus tachycardia with a heart rate of 102 bpm, and he was instructed to avoid any exercise until further notice.

At 6-month follow-up, the sinus tachycardia had resolved. However, because persistent tachycardia had been noted at the first postdischarge visit, and in view of the extent of myocardial involvement, he underwent exercise treadmill testing to evaluate for ventricular arrhythmias. The study did show premature ventricular complexes and 1 ventricular couplet at submaximal exercise levels. As this indicated a higher risk of exercise-induced arrhythmias, he was asked to continue normal activity levels but to abstain from exercise until the next evaluation.

During his 1-year follow-up, a repeat treadmill test showed no ventricular ectopy. Holter monitoring was ordered and showed no premature ventricular complexes, supraventricular arrhythmias, or atrioventricular block within the 48-hour period.

At his 2-year evaluation, he had returned to playing basketball and soccer on weekends and reported no recurrence of his initial symptoms.

KEY POINTS

  • farid_myocarditis_f3.jpg
    %3Cp%3EFigure%203.%20Our%20suggested%20approach%20to%20suspected%20acute%20myocarditis.%3C%2Fp%3E
    Cardiac MRI has emerged as an excellent noninvasive imaging modality for the diagnosis of myocarditis.
  • Treatment of myocarditis depends on the cause and severity of the patient’s presentation, spanning the spectrum from conservative care to immunosuppressive agents and even heart failure therapy.
  • Excessive physical activity should be avoided for the first 6 months after disease diagnosis and treatment.
  • If myocarditis is associated with pericardial involvement, colchicine is the agent of choice, and NSAIDs should be avoided.

Our suggested strategy for approaching myocarditis is shown in Figure 3.

References
  1. Dennert R, Crijns HJ, Heymans S. Acute viral myocarditis. Eur Heart J 2008; 29(17):2073–2082. doi:10.1093/eurheartj/ehn296
  2. Mahfoud F, Gärtner B, Kindermann M, et al. Virus serology in patients with suspected myocarditis: utility or futility? Eur Heart J 2011; 32(7):897–903. doi:10.1093/eurheartj/ehq493
  3. Caforio AL, Pankuweit S, Arbustini E, et al; European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2013; 34(33):2636–2648, 2648a–2648d. doi:10.1093/eurheartj/eht210
  4. Donnelly JP, Hanna M. Cardiac amyloidosis: an update on diagnosis and treatment. Cleve Clin J Med 2017; 84(12 suppl 3):12–26. doi:10.3949/ccjm.84.s3.02
  5. Siddiqi OK, Ruberg FL. Cardiac amyloidosis: an update on pathophysiology, diagnosis, and treatment. Trends Cardiovasc Med 2018; 28(1):10–21. doi:10.1016/j.tcm.2017.07.004
  6. Gertz MA, Benson MD, Dyck PJ, et al. Diagnosis, prognosis, and therapy of transthyretin amyloidosis. J Am Coll Cardiol 2015; 66(21):2451–2466. doi:10.1016/j.jacc.2015.09.075
  7. Blankstein R, Osborne M, Naya M, et al. Cardiac positron emission tomography enhances prognostic assessments of patients with suspected cardiac sarcoidosis. J Am Coll Cardiol 2014; 63(4):329–336. doi:10.1016/j.jacc.2013.09.022
  8. Baccouche H, Mahrholtz H, Meinhardt G, et al. Diagnostic synergy of non-invasive cardiovascular magnetic resonance and invasive endomyocardial biopsy in troponin-positive patients without coronary artery disease. Eur Heart J 2009; 30(23):2869–2879. doi:10.1093/eurheartj/ehp328
  9. Friedrich MG, Sechtem U, Schulz-Menger J, et al; International Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC white paper. J Am Coll Cardiol 2009; 53(17):1475–1487. doi:10.1016/j.jacc.2009.02.007
  10. Kindermann I, Barth C, Mahfoud F, et al. Update on myocarditis. J Am Coll Cardiol 2012; 59(9):779–792. doi:10.1016/j.jacc.2011.09.074
  11. Mahrholdt H, Wagner A, Deluigi CC, et al. Presentation, patterns of myocardial damage, and clinical course of viral myocarditis. Circulation 2006; 114(15):1581–1590. doi:10.1161/CIRCULATIONAHA.105.606509
  12. Gräni C, Eichhorn C, Bière L, et al. Prognostic value of cardiac magnetic resonance tissue characterization in risk stratifying patients with suspected myocarditis. J Am Coll Cardiol 2017; 70(16):1964–1976. doi:10.1016/j.jacc.2017.08.050
  13. Lurz P, Luecke C, Eitel I, et al. Comprehensive cardiac magnetic resonance imaging in patients with suspected myocarditis: the MyoRacer-Trial. J Am Coll Cardiol 2016; 67(15):1800–1811. doi:10.1016/j.jacc.2016.02.013
  14. Gannon MP, Schaub E, Griens CL, Saba SG. State of the art: evaluation and prognostication of myocarditis using cardiac MRI. J Magn Reson Imaging 2019; 49(7):e122–e131. doi:10.1002/jmri.26611
  15. Cooper LT, Baughman KL, Feldman AM, et al. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology endorsed by the Heart Failure Society of America and the Heart Failure Association of the European Society of Cardiology. Eur Heart J 2007; 28(24):3076–3093. doi:10.1093/eurheartj/ehm456
  16. Sinagra G, Anzini M, Pereira NL, et al. Myocarditis in clinical practice. Mayo Clin Proc 2016; 91(9):1256–1266. doi:10.1016/j.mayocp.2016.05.013
  17. Cooper LT, Baughman KL, Feldman AM, et al; American Heart Association; American College of Cardiology; European Society of Cardiology. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology. Circulation 2007; 116(19):2216–2233. doi:10.1161/CIRCULATIONAHA.107.186093
  18. Leone O, Veinot JP, Angelini A, et al. 2011 consensus statement on endomyocardial biopsy from the Association for European Cardiovascular Pathology and the Society for Cardiovascular Pathology. Cardiovasc Pathol 2012; 21(4):245–274. doi:10.1016/j.carpath.2011.10.001
  19. Baughman KL. Diagnosis of myocarditis: death of Dallas criteria. Circulation 2006; 113(4):593–595. doi:10.1161/CIRCULATIONAHA.105.589663
  20. Alraies MC, Klein AL. Should we still use electrocardiography to diagnose pericardial disease? Cleve Clin J Med 2013; 80(2):97–100. doi:10.3949/ccjm.80a.11144
  21. Sagar S, Liu PP, Cooper LT Jr. Myocarditis. Lancet 2012; 379(9817):738–747. doi:10.1016/S0140-6736(11)60648-X
  22. Caforio AL, Marcolongo R, Basso C, Iliceto S. Clinical presentation and diagnosis of myocarditis. Heart 2015; 101(16):1332–1344. doi:10.1136/heartjnl-2014-306363
  23. Cooper LT Jr. Myocarditis. N Engl J Med 2009; 360(15):1526–1538. doi:10.1056/NEJMra0800028
  24. LeLeiko RM, Bower DJ, Larsen CP. MRSA-associated bacterial myocarditis causing ruptured ventricle and tamponade. Cardiology 2008; 111(3):188–190. doi:10.1159/000121602
  25. Wasi F, Shuter J. Primary bacterial infection of the myocardium. Front Biosci 2003; 8:s228–s231. pmid:12700039
  26. Al-Amoodi M, Rao K, Rao S, Brewer JH, Magalski A, Chhatriwalla AK. Fulminant myocarditis due to H1N1 influenza. Circ Heart Fail 2010; 3(3):e7–e9. doi:10.1161/CIRCHEARTFAILURE.110.938506
  27. Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA focused update on new pharmacological therapy for heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. J Am Coll Cardiol 2016; 68(13):1476–1488. doi:10.1016/j.jacc.2016.05.011
  28. Schmidt-Lucke C, Spillmann F, Bock T, et al. Interferon beta modulates endothelial damage in patients with cardiac persistence of human parvovirus b19 infection. J Infect Dis 2010; 201(6):936–945. doi:10.1086/650700
  29. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis: the Myocarditis Treatment Trial Investigators. N Engl J Med 1995; 333(5):269–275. doi:10.1056/NEJM199508033330501
  30. Imazio M, Bobbio M, Cecchi E, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the COlchicine for acute PEricarditis (COPE) trial. Circulation 2005; 112(13):2012–2016. doi:10.1161/CIRCULATIONAHA.105.542738
  31. Morgenstern D, Lisko J, Boniface NC, Mikolich BM, Mikolich JR. Myocarditis and colchicine: a new perspective from cardiac MRI. J Cardiovasc Magn Reson 2016; 18(suppl 1):0100.
  32. Maron BJ, Zipes DP, Kovacs RJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: preamble, principles, and general considerations: a scientific statement from the American Heart Association and American College of Cardiology. J Am Coll Cardiol 2015; 66(21):2343–2349. doi:10.1016/j.jacc.2015.09.032
References
  1. Dennert R, Crijns HJ, Heymans S. Acute viral myocarditis. Eur Heart J 2008; 29(17):2073–2082. doi:10.1093/eurheartj/ehn296
  2. Mahfoud F, Gärtner B, Kindermann M, et al. Virus serology in patients with suspected myocarditis: utility or futility? Eur Heart J 2011; 32(7):897–903. doi:10.1093/eurheartj/ehq493
  3. Caforio AL, Pankuweit S, Arbustini E, et al; European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2013; 34(33):2636–2648, 2648a–2648d. doi:10.1093/eurheartj/eht210
  4. Donnelly JP, Hanna M. Cardiac amyloidosis: an update on diagnosis and treatment. Cleve Clin J Med 2017; 84(12 suppl 3):12–26. doi:10.3949/ccjm.84.s3.02
  5. Siddiqi OK, Ruberg FL. Cardiac amyloidosis: an update on pathophysiology, diagnosis, and treatment. Trends Cardiovasc Med 2018; 28(1):10–21. doi:10.1016/j.tcm.2017.07.004
  6. Gertz MA, Benson MD, Dyck PJ, et al. Diagnosis, prognosis, and therapy of transthyretin amyloidosis. J Am Coll Cardiol 2015; 66(21):2451–2466. doi:10.1016/j.jacc.2015.09.075
  7. Blankstein R, Osborne M, Naya M, et al. Cardiac positron emission tomography enhances prognostic assessments of patients with suspected cardiac sarcoidosis. J Am Coll Cardiol 2014; 63(4):329–336. doi:10.1016/j.jacc.2013.09.022
  8. Baccouche H, Mahrholtz H, Meinhardt G, et al. Diagnostic synergy of non-invasive cardiovascular magnetic resonance and invasive endomyocardial biopsy in troponin-positive patients without coronary artery disease. Eur Heart J 2009; 30(23):2869–2879. doi:10.1093/eurheartj/ehp328
  9. Friedrich MG, Sechtem U, Schulz-Menger J, et al; International Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis. Cardiovascular magnetic resonance in myocarditis: a JACC white paper. J Am Coll Cardiol 2009; 53(17):1475–1487. doi:10.1016/j.jacc.2009.02.007
  10. Kindermann I, Barth C, Mahfoud F, et al. Update on myocarditis. J Am Coll Cardiol 2012; 59(9):779–792. doi:10.1016/j.jacc.2011.09.074
  11. Mahrholdt H, Wagner A, Deluigi CC, et al. Presentation, patterns of myocardial damage, and clinical course of viral myocarditis. Circulation 2006; 114(15):1581–1590. doi:10.1161/CIRCULATIONAHA.105.606509
  12. Gräni C, Eichhorn C, Bière L, et al. Prognostic value of cardiac magnetic resonance tissue characterization in risk stratifying patients with suspected myocarditis. J Am Coll Cardiol 2017; 70(16):1964–1976. doi:10.1016/j.jacc.2017.08.050
  13. Lurz P, Luecke C, Eitel I, et al. Comprehensive cardiac magnetic resonance imaging in patients with suspected myocarditis: the MyoRacer-Trial. J Am Coll Cardiol 2016; 67(15):1800–1811. doi:10.1016/j.jacc.2016.02.013
  14. Gannon MP, Schaub E, Griens CL, Saba SG. State of the art: evaluation and prognostication of myocarditis using cardiac MRI. J Magn Reson Imaging 2019; 49(7):e122–e131. doi:10.1002/jmri.26611
  15. Cooper LT, Baughman KL, Feldman AM, et al. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology endorsed by the Heart Failure Society of America and the Heart Failure Association of the European Society of Cardiology. Eur Heart J 2007; 28(24):3076–3093. doi:10.1093/eurheartj/ehm456
  16. Sinagra G, Anzini M, Pereira NL, et al. Myocarditis in clinical practice. Mayo Clin Proc 2016; 91(9):1256–1266. doi:10.1016/j.mayocp.2016.05.013
  17. Cooper LT, Baughman KL, Feldman AM, et al; American Heart Association; American College of Cardiology; European Society of Cardiology. The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology. Circulation 2007; 116(19):2216–2233. doi:10.1161/CIRCULATIONAHA.107.186093
  18. Leone O, Veinot JP, Angelini A, et al. 2011 consensus statement on endomyocardial biopsy from the Association for European Cardiovascular Pathology and the Society for Cardiovascular Pathology. Cardiovasc Pathol 2012; 21(4):245–274. doi:10.1016/j.carpath.2011.10.001
  19. Baughman KL. Diagnosis of myocarditis: death of Dallas criteria. Circulation 2006; 113(4):593–595. doi:10.1161/CIRCULATIONAHA.105.589663
  20. Alraies MC, Klein AL. Should we still use electrocardiography to diagnose pericardial disease? Cleve Clin J Med 2013; 80(2):97–100. doi:10.3949/ccjm.80a.11144
  21. Sagar S, Liu PP, Cooper LT Jr. Myocarditis. Lancet 2012; 379(9817):738–747. doi:10.1016/S0140-6736(11)60648-X
  22. Caforio AL, Marcolongo R, Basso C, Iliceto S. Clinical presentation and diagnosis of myocarditis. Heart 2015; 101(16):1332–1344. doi:10.1136/heartjnl-2014-306363
  23. Cooper LT Jr. Myocarditis. N Engl J Med 2009; 360(15):1526–1538. doi:10.1056/NEJMra0800028
  24. LeLeiko RM, Bower DJ, Larsen CP. MRSA-associated bacterial myocarditis causing ruptured ventricle and tamponade. Cardiology 2008; 111(3):188–190. doi:10.1159/000121602
  25. Wasi F, Shuter J. Primary bacterial infection of the myocardium. Front Biosci 2003; 8:s228–s231. pmid:12700039
  26. Al-Amoodi M, Rao K, Rao S, Brewer JH, Magalski A, Chhatriwalla AK. Fulminant myocarditis due to H1N1 influenza. Circ Heart Fail 2010; 3(3):e7–e9. doi:10.1161/CIRCHEARTFAILURE.110.938506
  27. Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA focused update on new pharmacological therapy for heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. J Am Coll Cardiol 2016; 68(13):1476–1488. doi:10.1016/j.jacc.2016.05.011
  28. Schmidt-Lucke C, Spillmann F, Bock T, et al. Interferon beta modulates endothelial damage in patients with cardiac persistence of human parvovirus b19 infection. J Infect Dis 2010; 201(6):936–945. doi:10.1086/650700
  29. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis: the Myocarditis Treatment Trial Investigators. N Engl J Med 1995; 333(5):269–275. doi:10.1056/NEJM199508033330501
  30. Imazio M, Bobbio M, Cecchi E, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the COlchicine for acute PEricarditis (COPE) trial. Circulation 2005; 112(13):2012–2016. doi:10.1161/CIRCULATIONAHA.105.542738
  31. Morgenstern D, Lisko J, Boniface NC, Mikolich BM, Mikolich JR. Myocarditis and colchicine: a new perspective from cardiac MRI. J Cardiovasc Magn Reson 2016; 18(suppl 1):0100.
  32. Maron BJ, Zipes DP, Kovacs RJ. Eligibility and disqualification recommendations for competitive athletes with cardiovascular abnormalities: preamble, principles, and general considerations: a scientific statement from the American Heart Association and American College of Cardiology. J Am Coll Cardiol 2015; 66(21):2343–2349. doi:10.1016/j.jacc.2015.09.032
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Page Number
586-594
Page Number
586-594
Publications
Publications
Topics
Article Type
Display Headline
A young man with acute chest pain
Display Headline
A young man with acute chest pain
Legacy Keywords
chest pain, angina, myocarditis, pericarditis, ST-segment elevation, serologic testing, light chain, myocardial perfusion, magnetic resonance imaging, MRI, biopsy, amyloidosis, sarcoidosis, parvovirus B19, colchicine, Amir Farid, Neil Beri, David Torres-Barba, Charles Whitcomb
Legacy Keywords
chest pain, angina, myocarditis, pericarditis, ST-segment elevation, serologic testing, light chain, myocardial perfusion, magnetic resonance imaging, MRI, biopsy, amyloidosis, sarcoidosis, parvovirus B19, colchicine, Amir Farid, Neil Beri, David Torres-Barba, Charles Whitcomb
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/22/2019 - 09:15
Un-Gate On Date
Thu, 08/22/2019 - 09:15
Use ProPublica
CFC Schedule Remove Status
Thu, 08/22/2019 - 09:15
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Diabetes management: Beyond hemoglobin A1c

Article Type
Changed
Tue, 05/03/2022 - 15:13
Display Headline
Diabetes management: Beyond hemoglobin A1c

When scientists discovered the band of hemoglobin A1c during electrophoresis in the 1950s and 1960s and discerned it was elevated in patients with diabetes, little did they know the important role it would play in the diagnosis and treatment of diabetes in the decades to come.1–3 Despite some caveats, a hemoglobin A1c level of 6.5% or higher is diagnostic of diabetes across most populations, and hemoglobin A1c goals ranging from 6.5% to 7.5% have been set for different subsets of patients depending on comorbidities, complications, risk of hypoglycemia, life expectancy, disease duration, patient preferences, and available resources.4

makin_diabetestargets_t1.jpg

With a growing number of medications for diabetes—insulin in its various formulations and 11 other classes—hemoglobin A1c targets can now be tailored to fit individual patient profiles. Although helping patients attain their glycemic goals is paramount, other factors should be considered when prescribing or changing a drug treatment regimen, such as cardiovascular risk reduction, weight control, avoidance of hypoglycemia, and minimizing out-of-pocket drug costs (Table 1).

CARDIOVASCULAR BENEFIT

Patients with type 2 diabetes have a 2 to 3 times higher risk of clinical atherosclerotic disease, according to 20 years of surveillance data from the Framingham cohort.5

Mixed results with intensive treatment

Reducing cardiovascular risk remains an important goal in diabetes management, but unfortunately, data from the long-term clinical trials aimed at reducing macrovascular risk with intensive glycemic management have been conflicting.

The United Kingdom Prospective Diabetes Study (UKPDS),6 which enrolled more than 4,000 patients with newly diagnosed type 2 diabetes, did not initially show a statistically significant difference in the incidence of myocardial infarction with intensive control vs conventional control, although intensive treatment did reduce the incidence of microvascular disease. However, 10 years after the trial ended, the incidence was 15% lower in the intensive-treatment group than in the conventional-treatment group, and the difference was statistically significant.7

A 10-year follow-up analysis of the Veterans Affairs Diabetes Trial (VADT)8 showed that patients who had been randomly assigned to intensive glucose control for 5.6 years had 8.6 fewer major cardiovascular events per 1,000 person-years than those assigned to standard therapy, but no improvement in median overall survival. The hemoglobin A1c levels achieved during the trial were 6.9% and 8.4%, respectively.

In 2008, the US Food and Drug Administration (FDA)9 mandated that all new applications for diabetes drugs must include cardiovascular outcome studies. Therefore, we now have data on the cardiovascular benefits of two antihyperglycemic drug classes—incretins and sodium-glucose cotransporter 2 (SGLT2) inhibitors, making them attractive medications to target both cardiac and glucose concerns.

Incretins

The incretin drugs comprise 2 classes, glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors.

Liraglutide. The Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial10 compared liraglutide (a GLP-1 receptor agonist) and placebo in 9,000 patients with diabetes who either had or were at high risk of cardiovascular disease. Patients in the liraglutide group had a lower risk of the primary composite end point of death from cardiovascular causes or the first episode of nonfatal (including silent) myocardial infarction or nonfatal stroke, and a lower risk of cardiovascular death, all-cause mortality, and microvascular events than those in the placebo group. The number of patients who would need to be treated to prevent 1 event in 3 years was 66 in the analysis of the primary outcome and 98 in the analysis of death from any cause.9

Lixisenatide. The Evaluation of Lixisenatide in Acute Coronary Syndrome (ELIXA) trial11 studied the effect of the once-daily GLP-1 receptor agonist lixisenatide on cardiovascular outcomes in 6,000 patients with type 2 diabetes with a recent coronary event. In contrast to LEADER, ELIXA did not show a cardiovascular benefit over placebo.

Exenatide. The Exenatide Study of Cardiovascular Event Lowering (EXSCEL)12 assessed another GLP-1 extended-release drug, exenatide, in 14,000 patients, 73% of whom had established cardiovascular disease. In those patients, the drug had a modest benefit in terms of first occurrence of any component of the composite outcome of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke (3-component major adverse cardiac event [MACE] outcome) in a time-to-event analysis, but the results were not statistically significant. However, the drug did significantly reduce all-cause mortality.

Semaglutide, another GLP-1 receptor agonist recently approved by the FDA, also showed benefit in patients who had cardiovascular disease or were at high risk, with significant reduction in the primary composite end point of death from cardiovascular causes or the first occurrence of nonfatal myocardial infarction (including silent) or nonfatal stroke.13

Dulaglutide, a newer GLP-1 drug, was associated with significantly reduced major adverse cardiovascular events (a composite end point of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) in about 9,900 patients with diabetes, with a median follow-up of more than 5 years. Only 31% of the patients in the trial had established cardiovascular disease.14

Comment. GLP-1 drugs as a class are a good option for patients with diabetes who require weight loss, and liraglutide is now FDA-approved for reduction of cardiovascular events in patients with type 2 diabetes with established cardiovascular disease. However, other factors should be considered when prescribing these drugs: they have adverse gastrointestinal effects, the cardiovascular benefit was not a class effect, they are relatively expensive, and they must be injected. Also, they should not be prescribed concurrently with a DPP-4 inhibitor because they target the same pathway.

 

 

SGLT2 inhibitors

The other class of diabetes drugs that have shown cardiovascular benefit are the SGLT2 inhibitors.

Empagliflozin. The Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG)15 compared the efficacy of empagliflozin vs placebo in 7,000 patients with diabetes and cardiovascular disease and showed relative risk reductions of 38% in death from cardiovascular death, 31% in sudden death, and 35% in heart failure hospitalizations. Empagliflozin also showed benefit in terms of progression of kidney disease and occurrence of clinically relevant renal events in this population.16

Canagliflozin also has cardiovascular outcome data and showed significant benefit when compared with placebo in the primary outcome of the composite of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke, but no significant effects on cardiovascular death or all-cause mortality.17 Data from this trial also suggested a nonsignificant benefit of canagliflozin in decreasing progression of albuminuria and in the composite outcome of a sustained 40% reduction in the estimated glomerular filtration rate (eGFR), the need for renal replacement therapy, or death from renal causes.

The above data led to an additional indication from the FDA for empagliflozin—and recently, canagliflozin—to prevent cardiovascular death in patients with diabetes with established disease, but other factors should be considered when prescribing them. Patients taking canagliflozin showed a significantly increased risk of amputation. SGLT2 inhibitors as a class also increase the risk of genital infections in men and women; this is an important consideration since patients with diabetes complain of vaginal fungal and urinary tract infections even without the use of these drugs. A higher incidence of fractures with canagliflozin should also be considered when using these medications in elderly and osteoporosis-prone patients at high risk of falling.

Dapagliflozin, the third drug in this class, was associated with a lower rate of hospitalization for heart failure in about 17,160 patients—including 10,186 without atherosclerotic cardiovascular disease—who were followed for a median of 4.2 years.18 It did not show benefit for the primary safety outcome, a composite of major adverse cardiovascular events defined as cardiovascular death, myocardial infarction, or ischemic stroke.

WEIGHT MANAGEMENT

Weight loss can help overweight patients reach their hemoglobin A1c target.

Metformin should be continued as other drugs are added because it does not induce weight gain and may help with weight loss of up to 2 kg as shown in the Diabetes Prevention Program Outcomes Study.19

GLP-1 receptor agonists and SGLT2 inhibitors help with weight loss and are good additions to a basal insulin regimen to minimize weight gain.

Liraglutide was associated with a mean weight loss of 2.3 kg over 36 months of treatment compared with placebo in the LEADER trial.10

In the Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes (SUSTAIN-6),20 the mean body weight in the semaglutide group, compared with the placebo group, was 2.9 kg lower in the group receiving a lower dose and 4.3 kg lower in the group receiving a higher dose of the drug.

In a 24-week trial in 182 patients with type 2 diabetes inadequately controlled on metformin, dapagliflozin produced a statistically significant weight reduction of 2.08 kg (95% confidence interval 2.84–1.31; P < .0001) compared with placebo.21

Lifestyle changes aimed at weight management should be emphasized and discussed at every visit.

HYPOGLYCEMIA RISK

Hypoglycemia is a major consideration when tailoring hemoglobin A1c targets. In the Action to Control Cardiovascular Risk (ACCORD) trial,22 severe, symptomatic hypoglycemia increased the risk of death in both the intensive and conventional treatment groups. In VADT, the occurrence of a recent severe hypoglycemic event was the strongest independent predictor of death within 90 days. Further analysis showed that even though serious hypoglycemia occurred more often in the intensive therapy group, it was associated with progression of coronary artery calcification in the standard therapy group.23 Hence, it is imperative that tight glycemic control not be achieved at the cost of severe or recurrent hypoglycemia.

In terms of hypoglycemia, metformin is an excellent medication. The American Diabetes Association24 recommends metformin as the first-line therapy for newly diagnosed diabetes. Long-term follow-up data from UKPDS showed that metformin decreased mortality and the incidence of myocardial infarction and lowered treatment costs as well as the overall risk of hypoglycemia.25 When prescribed, it should be titrated to the highest dose.

The FDA26 has changed the prescribing information for metformin in patients with renal impairment. Metformin should not be started if the eGFR is less than 45 mL/min/1.73 m2, but it can be continued if the patient is already receiving it and the eGFR is between 30 and 45. Previously, creatinine levels were used to define renal impairment and suitability for metformin. This change has increased the number of patients who can benefit from this medication.

In patients who have a contraindication to metformin, DPP-4 inhibitors can be considered, as they carry a low risk of hypoglycemia as well. Sulfonylureas should be used with caution in these patients, especially if their oral intake is variable. When sulfonylureas were compared to the DPP-4 inhibitor sitagliptin as an add-on to metformin, the rate of hypoglycemia was 32% in the sulfonylurea group vs 5% in the sitagliptin group.27

Of the sulfonylureas, glipizide and glimepiride are better than glyburide because of a comparatively lower risk of hypoglycemia and a higher selectivity for binding the KATP channel on the pancreatic beta cell.28

Meglitinides can be a good option for patients who skip meals, but they are more expensive than other generic oral hypoglycemic agents and require multiple daily dosing.

GLP-1 analogues also have a low risk of hypoglycemia but are only available in injectable formulations. Patients must be willing and able to perform the injections themselves.29

 

 

LOOSER TARGETS FOR OLDER PATIENTS

In 2010, among US residents age 65 and older, 10.9 million (about 27%) had diabetes,30 and this number is projected to increase to 26.7 million by 2050.31 This population is prone to hypoglycemia when treated with insulin and sulfonylureas. An injury sustained by a fall induced by hypoglycemia can be life-altering. In addition, no randomized clinical trials show the effect of tight glycemic control on complications in older patients with diabetes because patients older than 80 are often excluded.

A reasonable goal suggested by the European Diabetes Working Party for Older People 201132 and reiterated by the American Geriatrics Society in 201333 is a hemoglobin A1c between 7% and 7.5% for relatively healthy older patients and 7.5% to 8% or 8.5% in frail elderly patients with diabetes.

Consider prescribing medications that carry a low risk of hypoglycemia, can be dose-adjusted for kidney function, and do not rely on manual dexterity for administration (ie, do not require patients to give themselves injections). These include metformin and DPP-4 inhibitors.

DRUG COMBINATIONS

Polypharmacy is a concern for all patients with diabetes, especially since it increases the risk of drug interactions and adverse effects, increases out-of-pocket costs, and decreases the likelihood that patients will remain adherent to their treatment regimen. The use of combination medications can reduce the number of pills or injections required, as well as copayments.

Due to concern for multiple drug-drug interactions (and also due to the progressive nature of diabetes), many people with type 2 diabetes are given insulin in lieu of pills to lower their blood glucose. In addition to premixed insulin combinations (such as combinations of neutral protamine Hagedorn and regular insulin or combinations of insulin analogues), long-acting basal insulins can now be prescribed with a GLP-1 drug in fixed-dose combinations such as insulin glargine plus lixisenatide and insulin degludec plus liraglutide.

COST CONSIDERATIONS

It is important to discuss medication cost with patients, because many newer diabetic drugs are expensive and add to the financial burden of patients already paying for multiple medications, such as antihypertensives and statins.

Metformin and sulfonylureas are less expensive alternatives for patients who cannot afford GLP-1 analogues or SGLT2 inhibitors. Even within the same drug class, the formulary-preferred drug may be cheaper than the nonformulary alternative. Thus, it is helpful to research formulary alternatives before discussing treatment regimens with patients.

References
  1. Allen DW, Schroeder WA, Balog J. Observations on the chromatographic heterogeneity of normal adult and fetal human hemoglobin: a study of the effects of crystallization and chromatography on the heterogeneity and isoleucine content. J Amer Chem Soc 1958; 80(7):1628–1634. doi:10.1021/ja01540a030
  2. Huisman TH, Dozy AM. Studies on the heterogeneity of hemoglobin. V. Binding of hemoglobin with oxidized glutathione. J Lab Clin Med 1962; 60:302–319. pmid:14449875
  3. Rahbar S, Blumenfeld O, Ranney HM. Studies of an unusual hemoglobin in patients with diabetes mellitus. Biochem Biophys Res Commun 1969; 36(5):838–843. pmid:5808299
  4. American Diabetes Association. 6. Glycemic targets: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S55–S64. doi:10.2337/dc18-S006
  5. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241(19):2035–2038. pmid:430798
  6. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352(9131):837–853. [Erratum in Lancet 1999; 354:602.] pmid:9742976
  7. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  8. Hayward RA, Reaven PD, Wiitala WL, et al; VADT Investigators. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015; 372(23):2197–2206. doi:10.1056/NEJMoa1414266
  9. US Food and Drug Administration. Guidance for industry: diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. https://www.govinfo.gov/content/pkg/FR-2008-12-19/pdf/E8-30086.pdf. Accessed August 6, 2019.
  10. Marso SP, Daniels GH, Brown-Frandsen K, et al; LEADER Steering Committee; LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375(4):311–322. doi:10.1056/NEJMoa1603827
  11. Pfeffer MA, Claggett B, Diaz R, et al; ELIXA Investigators. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015; 373(23):2247–2257. doi:10.1056/NEJMoa1509225
  12. Holman RR, Bethel MA, Mentz RJ, et al; EXSCEL Study Group. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017; 377(13):1228–1239. doi:10.1056/NEJMoa1612917
  13. Cosmi F, Laini R, Nicolucci A. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2017; 376(9):890. doi:10.1056/NEJMc1615712
  14. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394(10193):121–130. doi:10.1016/S0140-6736(19)31149-3
  15. Zinman B, Wanner C, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015; 373(22):2117–2128. doi:10.1056/NEJMoa1504720
  16. Wanner C, Inzucchi SE, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016; 375(4):323–334. doi:10.1056/NEJMoa1515920
  17. Neal B, Perkovic V, Mahaffey KW, et al; CANVAS Program Collaborative Group. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017; 377(7):644–657. doi:10.1056/NEJMoa1611925
  18. Wiviott SD, Raz I, Bonaca MP, et al; DECLARE–TIMI 58 Investigators. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2018. [Epub ahead of print] doi:10.1056/NEJMoa1812389
  19. Diabetes Prevention Program Research Group; Knowler WC, Fowler SE, Hamman RF, et al. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 2009; 374(9702):1677–1686. doi:10.1016/S0140-6736(09)61457-4
  20. Marso SP, Bain SC, Consoli A, et al, for the SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016; 375:1834–1844. doi:10.1056/NEJMoa1607141
  21. Bolinder J, Ljunggren Ö, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab 2012; 97(3):1020–1031. doi:10.1210/jc.2011-2260
  22. Bonds DE, Miller ME, Bergenstal RM, et al. The association between symptomatic, severe hypoglycaemia and mortality in type 2 diabetes: retrospective epidemiological analysis of the ACCORD study. BMJ 2010; 340:b4909. doi:10.1136/bmj.b4909
  23. Saremi A, Bahn GD, Reaven PD; Veterans Affairs Diabetes Trial (VADT). A link between hypoglycemia and progression of atherosclerosis in the Veterans Affairs Diabetes Trial (VADT). Diabetes Care 2016; 39(3):448–454. doi:10.2337/dc15-2107
  24. American Diabetes Association. 8. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S73–S85. doi:10.2337/dc18-S008
  25. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  26. US Food and Drug Administration. FDA drug safety communication: FDA revises warnings regarding use of the diabetes medicine metformin in certain patients with reduced kidney function. www.fda.gov/Drugs/DrugSafety/ucm493244.htm. Accessed August 5, 2019.
  27. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9(2):194–205. doi:10.1111/j.1463-1326.2006.00704.x
  28. Gangji AS, Cukierman T, Gerstein HC, Goldsmith CH, Clase CM. A systematic review and meta-analysis of hypoglycemia and cardiovascular events: a comparison of glyburide with other secretagogues and with insulin. Diabetes Care 2007; 30(2):389–394. doi:10.2337/dc06-1789
  29. Nauck M, Frid A, Hermansen K, et al; LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32(1):84–90. doi:10.2337/dc08-1355
  30. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Accessed August 5, 2019.
  31. Boyle JP, Thompson TJ, Gregg EW, Barker LE, Williamson DF. Projection of the year 2050 burden of diabetes in the US adult population: dynamic modeling of incidence, mortality, and prediabetes prevalence. Popul Health Metr 2010; 8:29. doi:10.1186/1478-7954-8-29
  32. Sinclair AJ, Paolisso G, Castro M, Bourdel-Marchasson I, Gadsby R, Rodriguez Mañas L; European Diabetes Working Party for Older People. European Diabetes Working Party for Older People 2011 clinical guidelines for type 2 diabetes mellitus. Executive summary. Diabetes Metab 2011; 37(suppl 3):S27–S38. doi:10.1016/S1262-3636(11)70962-4
  33. American Geriatrics Society Expert Panel on Care of Older Adults with Diabetes Mellitus; Moreno G, Mangione CM, Kimbro L, Vaisberg E. Guidelines abstracted from the American Geriatrics Society Guidelines for Improving the Care of Older Adults with Diabetes Mellitus: 2013 update. J Am Geriatr Soc 2013; 61(11):2020–2026. doi:10.1111/jgs.12514
Article PDF
Author and Disclosure Information

Vinni Makin, MBBS, MD, FACE
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Assistant Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Clinical Assistant Professor, Ohio Heritage College of Osteopathic Medicine, Cleveland, OH

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Service; Chair, Cleveland Clinic Health Systems Diabetes Care Committee; Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Vinni Makin, MBBS, MD, FACE, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; makinv@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 86(9)
Publications
Topics
Page Number
595-600
Legacy Keywords
diabetes, drugs, hemoglobin A1c,management, glucagon-like peptide 1, GLP-1, GLP-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors, SGLT2 inhibitors, metformin, dipeptidyl peptidase 4 inhibitors, DPP-4 inhibitors, cardiovascular benefit, sulfonylureas, meglitinides, thiazolidinediones, incretins, liraglutide, lixisenatide, exenatide, semaglutide, dulaglutide, empagliflozin, canagliflozin, dapaagliflozin, weight management, hypoglycemia risk, Vinni Makin, M. Cecilia Lansang
Sections
Author and Disclosure Information

Vinni Makin, MBBS, MD, FACE
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Assistant Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Clinical Assistant Professor, Ohio Heritage College of Osteopathic Medicine, Cleveland, OH

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Service; Chair, Cleveland Clinic Health Systems Diabetes Care Committee; Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Vinni Makin, MBBS, MD, FACE, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; makinv@ccf.org

Author and Disclosure Information

Vinni Makin, MBBS, MD, FACE
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Assistant Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Clinical Assistant Professor, Ohio Heritage College of Osteopathic Medicine, Cleveland, OH

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Service; Chair, Cleveland Clinic Health Systems Diabetes Care Committee; Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic; Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Vinni Makin, MBBS, MD, FACE, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; makinv@ccf.org

Article PDF
Article PDF
Related Articles

When scientists discovered the band of hemoglobin A1c during electrophoresis in the 1950s and 1960s and discerned it was elevated in patients with diabetes, little did they know the important role it would play in the diagnosis and treatment of diabetes in the decades to come.1–3 Despite some caveats, a hemoglobin A1c level of 6.5% or higher is diagnostic of diabetes across most populations, and hemoglobin A1c goals ranging from 6.5% to 7.5% have been set for different subsets of patients depending on comorbidities, complications, risk of hypoglycemia, life expectancy, disease duration, patient preferences, and available resources.4

makin_diabetestargets_t1.jpg

With a growing number of medications for diabetes—insulin in its various formulations and 11 other classes—hemoglobin A1c targets can now be tailored to fit individual patient profiles. Although helping patients attain their glycemic goals is paramount, other factors should be considered when prescribing or changing a drug treatment regimen, such as cardiovascular risk reduction, weight control, avoidance of hypoglycemia, and minimizing out-of-pocket drug costs (Table 1).

CARDIOVASCULAR BENEFIT

Patients with type 2 diabetes have a 2 to 3 times higher risk of clinical atherosclerotic disease, according to 20 years of surveillance data from the Framingham cohort.5

Mixed results with intensive treatment

Reducing cardiovascular risk remains an important goal in diabetes management, but unfortunately, data from the long-term clinical trials aimed at reducing macrovascular risk with intensive glycemic management have been conflicting.

The United Kingdom Prospective Diabetes Study (UKPDS),6 which enrolled more than 4,000 patients with newly diagnosed type 2 diabetes, did not initially show a statistically significant difference in the incidence of myocardial infarction with intensive control vs conventional control, although intensive treatment did reduce the incidence of microvascular disease. However, 10 years after the trial ended, the incidence was 15% lower in the intensive-treatment group than in the conventional-treatment group, and the difference was statistically significant.7

A 10-year follow-up analysis of the Veterans Affairs Diabetes Trial (VADT)8 showed that patients who had been randomly assigned to intensive glucose control for 5.6 years had 8.6 fewer major cardiovascular events per 1,000 person-years than those assigned to standard therapy, but no improvement in median overall survival. The hemoglobin A1c levels achieved during the trial were 6.9% and 8.4%, respectively.

In 2008, the US Food and Drug Administration (FDA)9 mandated that all new applications for diabetes drugs must include cardiovascular outcome studies. Therefore, we now have data on the cardiovascular benefits of two antihyperglycemic drug classes—incretins and sodium-glucose cotransporter 2 (SGLT2) inhibitors, making them attractive medications to target both cardiac and glucose concerns.

Incretins

The incretin drugs comprise 2 classes, glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors.

Liraglutide. The Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial10 compared liraglutide (a GLP-1 receptor agonist) and placebo in 9,000 patients with diabetes who either had or were at high risk of cardiovascular disease. Patients in the liraglutide group had a lower risk of the primary composite end point of death from cardiovascular causes or the first episode of nonfatal (including silent) myocardial infarction or nonfatal stroke, and a lower risk of cardiovascular death, all-cause mortality, and microvascular events than those in the placebo group. The number of patients who would need to be treated to prevent 1 event in 3 years was 66 in the analysis of the primary outcome and 98 in the analysis of death from any cause.9

Lixisenatide. The Evaluation of Lixisenatide in Acute Coronary Syndrome (ELIXA) trial11 studied the effect of the once-daily GLP-1 receptor agonist lixisenatide on cardiovascular outcomes in 6,000 patients with type 2 diabetes with a recent coronary event. In contrast to LEADER, ELIXA did not show a cardiovascular benefit over placebo.

Exenatide. The Exenatide Study of Cardiovascular Event Lowering (EXSCEL)12 assessed another GLP-1 extended-release drug, exenatide, in 14,000 patients, 73% of whom had established cardiovascular disease. In those patients, the drug had a modest benefit in terms of first occurrence of any component of the composite outcome of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke (3-component major adverse cardiac event [MACE] outcome) in a time-to-event analysis, but the results were not statistically significant. However, the drug did significantly reduce all-cause mortality.

Semaglutide, another GLP-1 receptor agonist recently approved by the FDA, also showed benefit in patients who had cardiovascular disease or were at high risk, with significant reduction in the primary composite end point of death from cardiovascular causes or the first occurrence of nonfatal myocardial infarction (including silent) or nonfatal stroke.13

Dulaglutide, a newer GLP-1 drug, was associated with significantly reduced major adverse cardiovascular events (a composite end point of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) in about 9,900 patients with diabetes, with a median follow-up of more than 5 years. Only 31% of the patients in the trial had established cardiovascular disease.14

Comment. GLP-1 drugs as a class are a good option for patients with diabetes who require weight loss, and liraglutide is now FDA-approved for reduction of cardiovascular events in patients with type 2 diabetes with established cardiovascular disease. However, other factors should be considered when prescribing these drugs: they have adverse gastrointestinal effects, the cardiovascular benefit was not a class effect, they are relatively expensive, and they must be injected. Also, they should not be prescribed concurrently with a DPP-4 inhibitor because they target the same pathway.

 

 

SGLT2 inhibitors

The other class of diabetes drugs that have shown cardiovascular benefit are the SGLT2 inhibitors.

Empagliflozin. The Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG)15 compared the efficacy of empagliflozin vs placebo in 7,000 patients with diabetes and cardiovascular disease and showed relative risk reductions of 38% in death from cardiovascular death, 31% in sudden death, and 35% in heart failure hospitalizations. Empagliflozin also showed benefit in terms of progression of kidney disease and occurrence of clinically relevant renal events in this population.16

Canagliflozin also has cardiovascular outcome data and showed significant benefit when compared with placebo in the primary outcome of the composite of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke, but no significant effects on cardiovascular death or all-cause mortality.17 Data from this trial also suggested a nonsignificant benefit of canagliflozin in decreasing progression of albuminuria and in the composite outcome of a sustained 40% reduction in the estimated glomerular filtration rate (eGFR), the need for renal replacement therapy, or death from renal causes.

The above data led to an additional indication from the FDA for empagliflozin—and recently, canagliflozin—to prevent cardiovascular death in patients with diabetes with established disease, but other factors should be considered when prescribing them. Patients taking canagliflozin showed a significantly increased risk of amputation. SGLT2 inhibitors as a class also increase the risk of genital infections in men and women; this is an important consideration since patients with diabetes complain of vaginal fungal and urinary tract infections even without the use of these drugs. A higher incidence of fractures with canagliflozin should also be considered when using these medications in elderly and osteoporosis-prone patients at high risk of falling.

Dapagliflozin, the third drug in this class, was associated with a lower rate of hospitalization for heart failure in about 17,160 patients—including 10,186 without atherosclerotic cardiovascular disease—who were followed for a median of 4.2 years.18 It did not show benefit for the primary safety outcome, a composite of major adverse cardiovascular events defined as cardiovascular death, myocardial infarction, or ischemic stroke.

WEIGHT MANAGEMENT

Weight loss can help overweight patients reach their hemoglobin A1c target.

Metformin should be continued as other drugs are added because it does not induce weight gain and may help with weight loss of up to 2 kg as shown in the Diabetes Prevention Program Outcomes Study.19

GLP-1 receptor agonists and SGLT2 inhibitors help with weight loss and are good additions to a basal insulin regimen to minimize weight gain.

Liraglutide was associated with a mean weight loss of 2.3 kg over 36 months of treatment compared with placebo in the LEADER trial.10

In the Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes (SUSTAIN-6),20 the mean body weight in the semaglutide group, compared with the placebo group, was 2.9 kg lower in the group receiving a lower dose and 4.3 kg lower in the group receiving a higher dose of the drug.

In a 24-week trial in 182 patients with type 2 diabetes inadequately controlled on metformin, dapagliflozin produced a statistically significant weight reduction of 2.08 kg (95% confidence interval 2.84–1.31; P < .0001) compared with placebo.21

Lifestyle changes aimed at weight management should be emphasized and discussed at every visit.

HYPOGLYCEMIA RISK

Hypoglycemia is a major consideration when tailoring hemoglobin A1c targets. In the Action to Control Cardiovascular Risk (ACCORD) trial,22 severe, symptomatic hypoglycemia increased the risk of death in both the intensive and conventional treatment groups. In VADT, the occurrence of a recent severe hypoglycemic event was the strongest independent predictor of death within 90 days. Further analysis showed that even though serious hypoglycemia occurred more often in the intensive therapy group, it was associated with progression of coronary artery calcification in the standard therapy group.23 Hence, it is imperative that tight glycemic control not be achieved at the cost of severe or recurrent hypoglycemia.

In terms of hypoglycemia, metformin is an excellent medication. The American Diabetes Association24 recommends metformin as the first-line therapy for newly diagnosed diabetes. Long-term follow-up data from UKPDS showed that metformin decreased mortality and the incidence of myocardial infarction and lowered treatment costs as well as the overall risk of hypoglycemia.25 When prescribed, it should be titrated to the highest dose.

The FDA26 has changed the prescribing information for metformin in patients with renal impairment. Metformin should not be started if the eGFR is less than 45 mL/min/1.73 m2, but it can be continued if the patient is already receiving it and the eGFR is between 30 and 45. Previously, creatinine levels were used to define renal impairment and suitability for metformin. This change has increased the number of patients who can benefit from this medication.

In patients who have a contraindication to metformin, DPP-4 inhibitors can be considered, as they carry a low risk of hypoglycemia as well. Sulfonylureas should be used with caution in these patients, especially if their oral intake is variable. When sulfonylureas were compared to the DPP-4 inhibitor sitagliptin as an add-on to metformin, the rate of hypoglycemia was 32% in the sulfonylurea group vs 5% in the sitagliptin group.27

Of the sulfonylureas, glipizide and glimepiride are better than glyburide because of a comparatively lower risk of hypoglycemia and a higher selectivity for binding the KATP channel on the pancreatic beta cell.28

Meglitinides can be a good option for patients who skip meals, but they are more expensive than other generic oral hypoglycemic agents and require multiple daily dosing.

GLP-1 analogues also have a low risk of hypoglycemia but are only available in injectable formulations. Patients must be willing and able to perform the injections themselves.29

 

 

LOOSER TARGETS FOR OLDER PATIENTS

In 2010, among US residents age 65 and older, 10.9 million (about 27%) had diabetes,30 and this number is projected to increase to 26.7 million by 2050.31 This population is prone to hypoglycemia when treated with insulin and sulfonylureas. An injury sustained by a fall induced by hypoglycemia can be life-altering. In addition, no randomized clinical trials show the effect of tight glycemic control on complications in older patients with diabetes because patients older than 80 are often excluded.

A reasonable goal suggested by the European Diabetes Working Party for Older People 201132 and reiterated by the American Geriatrics Society in 201333 is a hemoglobin A1c between 7% and 7.5% for relatively healthy older patients and 7.5% to 8% or 8.5% in frail elderly patients with diabetes.

Consider prescribing medications that carry a low risk of hypoglycemia, can be dose-adjusted for kidney function, and do not rely on manual dexterity for administration (ie, do not require patients to give themselves injections). These include metformin and DPP-4 inhibitors.

DRUG COMBINATIONS

Polypharmacy is a concern for all patients with diabetes, especially since it increases the risk of drug interactions and adverse effects, increases out-of-pocket costs, and decreases the likelihood that patients will remain adherent to their treatment regimen. The use of combination medications can reduce the number of pills or injections required, as well as copayments.

Due to concern for multiple drug-drug interactions (and also due to the progressive nature of diabetes), many people with type 2 diabetes are given insulin in lieu of pills to lower their blood glucose. In addition to premixed insulin combinations (such as combinations of neutral protamine Hagedorn and regular insulin or combinations of insulin analogues), long-acting basal insulins can now be prescribed with a GLP-1 drug in fixed-dose combinations such as insulin glargine plus lixisenatide and insulin degludec plus liraglutide.

COST CONSIDERATIONS

It is important to discuss medication cost with patients, because many newer diabetic drugs are expensive and add to the financial burden of patients already paying for multiple medications, such as antihypertensives and statins.

Metformin and sulfonylureas are less expensive alternatives for patients who cannot afford GLP-1 analogues or SGLT2 inhibitors. Even within the same drug class, the formulary-preferred drug may be cheaper than the nonformulary alternative. Thus, it is helpful to research formulary alternatives before discussing treatment regimens with patients.

When scientists discovered the band of hemoglobin A1c during electrophoresis in the 1950s and 1960s and discerned it was elevated in patients with diabetes, little did they know the important role it would play in the diagnosis and treatment of diabetes in the decades to come.1–3 Despite some caveats, a hemoglobin A1c level of 6.5% or higher is diagnostic of diabetes across most populations, and hemoglobin A1c goals ranging from 6.5% to 7.5% have been set for different subsets of patients depending on comorbidities, complications, risk of hypoglycemia, life expectancy, disease duration, patient preferences, and available resources.4

makin_diabetestargets_t1.jpg

With a growing number of medications for diabetes—insulin in its various formulations and 11 other classes—hemoglobin A1c targets can now be tailored to fit individual patient profiles. Although helping patients attain their glycemic goals is paramount, other factors should be considered when prescribing or changing a drug treatment regimen, such as cardiovascular risk reduction, weight control, avoidance of hypoglycemia, and minimizing out-of-pocket drug costs (Table 1).

CARDIOVASCULAR BENEFIT

Patients with type 2 diabetes have a 2 to 3 times higher risk of clinical atherosclerotic disease, according to 20 years of surveillance data from the Framingham cohort.5

Mixed results with intensive treatment

Reducing cardiovascular risk remains an important goal in diabetes management, but unfortunately, data from the long-term clinical trials aimed at reducing macrovascular risk with intensive glycemic management have been conflicting.

The United Kingdom Prospective Diabetes Study (UKPDS),6 which enrolled more than 4,000 patients with newly diagnosed type 2 diabetes, did not initially show a statistically significant difference in the incidence of myocardial infarction with intensive control vs conventional control, although intensive treatment did reduce the incidence of microvascular disease. However, 10 years after the trial ended, the incidence was 15% lower in the intensive-treatment group than in the conventional-treatment group, and the difference was statistically significant.7

A 10-year follow-up analysis of the Veterans Affairs Diabetes Trial (VADT)8 showed that patients who had been randomly assigned to intensive glucose control for 5.6 years had 8.6 fewer major cardiovascular events per 1,000 person-years than those assigned to standard therapy, but no improvement in median overall survival. The hemoglobin A1c levels achieved during the trial were 6.9% and 8.4%, respectively.

In 2008, the US Food and Drug Administration (FDA)9 mandated that all new applications for diabetes drugs must include cardiovascular outcome studies. Therefore, we now have data on the cardiovascular benefits of two antihyperglycemic drug classes—incretins and sodium-glucose cotransporter 2 (SGLT2) inhibitors, making them attractive medications to target both cardiac and glucose concerns.

Incretins

The incretin drugs comprise 2 classes, glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase 4 (DPP-4) inhibitors.

Liraglutide. The Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial10 compared liraglutide (a GLP-1 receptor agonist) and placebo in 9,000 patients with diabetes who either had or were at high risk of cardiovascular disease. Patients in the liraglutide group had a lower risk of the primary composite end point of death from cardiovascular causes or the first episode of nonfatal (including silent) myocardial infarction or nonfatal stroke, and a lower risk of cardiovascular death, all-cause mortality, and microvascular events than those in the placebo group. The number of patients who would need to be treated to prevent 1 event in 3 years was 66 in the analysis of the primary outcome and 98 in the analysis of death from any cause.9

Lixisenatide. The Evaluation of Lixisenatide in Acute Coronary Syndrome (ELIXA) trial11 studied the effect of the once-daily GLP-1 receptor agonist lixisenatide on cardiovascular outcomes in 6,000 patients with type 2 diabetes with a recent coronary event. In contrast to LEADER, ELIXA did not show a cardiovascular benefit over placebo.

Exenatide. The Exenatide Study of Cardiovascular Event Lowering (EXSCEL)12 assessed another GLP-1 extended-release drug, exenatide, in 14,000 patients, 73% of whom had established cardiovascular disease. In those patients, the drug had a modest benefit in terms of first occurrence of any component of the composite outcome of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke (3-component major adverse cardiac event [MACE] outcome) in a time-to-event analysis, but the results were not statistically significant. However, the drug did significantly reduce all-cause mortality.

Semaglutide, another GLP-1 receptor agonist recently approved by the FDA, also showed benefit in patients who had cardiovascular disease or were at high risk, with significant reduction in the primary composite end point of death from cardiovascular causes or the first occurrence of nonfatal myocardial infarction (including silent) or nonfatal stroke.13

Dulaglutide, a newer GLP-1 drug, was associated with significantly reduced major adverse cardiovascular events (a composite end point of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) in about 9,900 patients with diabetes, with a median follow-up of more than 5 years. Only 31% of the patients in the trial had established cardiovascular disease.14

Comment. GLP-1 drugs as a class are a good option for patients with diabetes who require weight loss, and liraglutide is now FDA-approved for reduction of cardiovascular events in patients with type 2 diabetes with established cardiovascular disease. However, other factors should be considered when prescribing these drugs: they have adverse gastrointestinal effects, the cardiovascular benefit was not a class effect, they are relatively expensive, and they must be injected. Also, they should not be prescribed concurrently with a DPP-4 inhibitor because they target the same pathway.

 

 

SGLT2 inhibitors

The other class of diabetes drugs that have shown cardiovascular benefit are the SGLT2 inhibitors.

Empagliflozin. The Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG)15 compared the efficacy of empagliflozin vs placebo in 7,000 patients with diabetes and cardiovascular disease and showed relative risk reductions of 38% in death from cardiovascular death, 31% in sudden death, and 35% in heart failure hospitalizations. Empagliflozin also showed benefit in terms of progression of kidney disease and occurrence of clinically relevant renal events in this population.16

Canagliflozin also has cardiovascular outcome data and showed significant benefit when compared with placebo in the primary outcome of the composite of death from cardiovascular causes, nonfatal myocardial infarction, or nonfatal stroke, but no significant effects on cardiovascular death or all-cause mortality.17 Data from this trial also suggested a nonsignificant benefit of canagliflozin in decreasing progression of albuminuria and in the composite outcome of a sustained 40% reduction in the estimated glomerular filtration rate (eGFR), the need for renal replacement therapy, or death from renal causes.

The above data led to an additional indication from the FDA for empagliflozin—and recently, canagliflozin—to prevent cardiovascular death in patients with diabetes with established disease, but other factors should be considered when prescribing them. Patients taking canagliflozin showed a significantly increased risk of amputation. SGLT2 inhibitors as a class also increase the risk of genital infections in men and women; this is an important consideration since patients with diabetes complain of vaginal fungal and urinary tract infections even without the use of these drugs. A higher incidence of fractures with canagliflozin should also be considered when using these medications in elderly and osteoporosis-prone patients at high risk of falling.

Dapagliflozin, the third drug in this class, was associated with a lower rate of hospitalization for heart failure in about 17,160 patients—including 10,186 without atherosclerotic cardiovascular disease—who were followed for a median of 4.2 years.18 It did not show benefit for the primary safety outcome, a composite of major adverse cardiovascular events defined as cardiovascular death, myocardial infarction, or ischemic stroke.

WEIGHT MANAGEMENT

Weight loss can help overweight patients reach their hemoglobin A1c target.

Metformin should be continued as other drugs are added because it does not induce weight gain and may help with weight loss of up to 2 kg as shown in the Diabetes Prevention Program Outcomes Study.19

GLP-1 receptor agonists and SGLT2 inhibitors help with weight loss and are good additions to a basal insulin regimen to minimize weight gain.

Liraglutide was associated with a mean weight loss of 2.3 kg over 36 months of treatment compared with placebo in the LEADER trial.10

In the Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes (SUSTAIN-6),20 the mean body weight in the semaglutide group, compared with the placebo group, was 2.9 kg lower in the group receiving a lower dose and 4.3 kg lower in the group receiving a higher dose of the drug.

In a 24-week trial in 182 patients with type 2 diabetes inadequately controlled on metformin, dapagliflozin produced a statistically significant weight reduction of 2.08 kg (95% confidence interval 2.84–1.31; P < .0001) compared with placebo.21

Lifestyle changes aimed at weight management should be emphasized and discussed at every visit.

HYPOGLYCEMIA RISK

Hypoglycemia is a major consideration when tailoring hemoglobin A1c targets. In the Action to Control Cardiovascular Risk (ACCORD) trial,22 severe, symptomatic hypoglycemia increased the risk of death in both the intensive and conventional treatment groups. In VADT, the occurrence of a recent severe hypoglycemic event was the strongest independent predictor of death within 90 days. Further analysis showed that even though serious hypoglycemia occurred more often in the intensive therapy group, it was associated with progression of coronary artery calcification in the standard therapy group.23 Hence, it is imperative that tight glycemic control not be achieved at the cost of severe or recurrent hypoglycemia.

In terms of hypoglycemia, metformin is an excellent medication. The American Diabetes Association24 recommends metformin as the first-line therapy for newly diagnosed diabetes. Long-term follow-up data from UKPDS showed that metformin decreased mortality and the incidence of myocardial infarction and lowered treatment costs as well as the overall risk of hypoglycemia.25 When prescribed, it should be titrated to the highest dose.

The FDA26 has changed the prescribing information for metformin in patients with renal impairment. Metformin should not be started if the eGFR is less than 45 mL/min/1.73 m2, but it can be continued if the patient is already receiving it and the eGFR is between 30 and 45. Previously, creatinine levels were used to define renal impairment and suitability for metformin. This change has increased the number of patients who can benefit from this medication.

In patients who have a contraindication to metformin, DPP-4 inhibitors can be considered, as they carry a low risk of hypoglycemia as well. Sulfonylureas should be used with caution in these patients, especially if their oral intake is variable. When sulfonylureas were compared to the DPP-4 inhibitor sitagliptin as an add-on to metformin, the rate of hypoglycemia was 32% in the sulfonylurea group vs 5% in the sitagliptin group.27

Of the sulfonylureas, glipizide and glimepiride are better than glyburide because of a comparatively lower risk of hypoglycemia and a higher selectivity for binding the KATP channel on the pancreatic beta cell.28

Meglitinides can be a good option for patients who skip meals, but they are more expensive than other generic oral hypoglycemic agents and require multiple daily dosing.

GLP-1 analogues also have a low risk of hypoglycemia but are only available in injectable formulations. Patients must be willing and able to perform the injections themselves.29

 

 

LOOSER TARGETS FOR OLDER PATIENTS

In 2010, among US residents age 65 and older, 10.9 million (about 27%) had diabetes,30 and this number is projected to increase to 26.7 million by 2050.31 This population is prone to hypoglycemia when treated with insulin and sulfonylureas. An injury sustained by a fall induced by hypoglycemia can be life-altering. In addition, no randomized clinical trials show the effect of tight glycemic control on complications in older patients with diabetes because patients older than 80 are often excluded.

A reasonable goal suggested by the European Diabetes Working Party for Older People 201132 and reiterated by the American Geriatrics Society in 201333 is a hemoglobin A1c between 7% and 7.5% for relatively healthy older patients and 7.5% to 8% or 8.5% in frail elderly patients with diabetes.

Consider prescribing medications that carry a low risk of hypoglycemia, can be dose-adjusted for kidney function, and do not rely on manual dexterity for administration (ie, do not require patients to give themselves injections). These include metformin and DPP-4 inhibitors.

DRUG COMBINATIONS

Polypharmacy is a concern for all patients with diabetes, especially since it increases the risk of drug interactions and adverse effects, increases out-of-pocket costs, and decreases the likelihood that patients will remain adherent to their treatment regimen. The use of combination medications can reduce the number of pills or injections required, as well as copayments.

Due to concern for multiple drug-drug interactions (and also due to the progressive nature of diabetes), many people with type 2 diabetes are given insulin in lieu of pills to lower their blood glucose. In addition to premixed insulin combinations (such as combinations of neutral protamine Hagedorn and regular insulin or combinations of insulin analogues), long-acting basal insulins can now be prescribed with a GLP-1 drug in fixed-dose combinations such as insulin glargine plus lixisenatide and insulin degludec plus liraglutide.

COST CONSIDERATIONS

It is important to discuss medication cost with patients, because many newer diabetic drugs are expensive and add to the financial burden of patients already paying for multiple medications, such as antihypertensives and statins.

Metformin and sulfonylureas are less expensive alternatives for patients who cannot afford GLP-1 analogues or SGLT2 inhibitors. Even within the same drug class, the formulary-preferred drug may be cheaper than the nonformulary alternative. Thus, it is helpful to research formulary alternatives before discussing treatment regimens with patients.

References
  1. Allen DW, Schroeder WA, Balog J. Observations on the chromatographic heterogeneity of normal adult and fetal human hemoglobin: a study of the effects of crystallization and chromatography on the heterogeneity and isoleucine content. J Amer Chem Soc 1958; 80(7):1628–1634. doi:10.1021/ja01540a030
  2. Huisman TH, Dozy AM. Studies on the heterogeneity of hemoglobin. V. Binding of hemoglobin with oxidized glutathione. J Lab Clin Med 1962; 60:302–319. pmid:14449875
  3. Rahbar S, Blumenfeld O, Ranney HM. Studies of an unusual hemoglobin in patients with diabetes mellitus. Biochem Biophys Res Commun 1969; 36(5):838–843. pmid:5808299
  4. American Diabetes Association. 6. Glycemic targets: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S55–S64. doi:10.2337/dc18-S006
  5. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241(19):2035–2038. pmid:430798
  6. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352(9131):837–853. [Erratum in Lancet 1999; 354:602.] pmid:9742976
  7. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  8. Hayward RA, Reaven PD, Wiitala WL, et al; VADT Investigators. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015; 372(23):2197–2206. doi:10.1056/NEJMoa1414266
  9. US Food and Drug Administration. Guidance for industry: diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. https://www.govinfo.gov/content/pkg/FR-2008-12-19/pdf/E8-30086.pdf. Accessed August 6, 2019.
  10. Marso SP, Daniels GH, Brown-Frandsen K, et al; LEADER Steering Committee; LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375(4):311–322. doi:10.1056/NEJMoa1603827
  11. Pfeffer MA, Claggett B, Diaz R, et al; ELIXA Investigators. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015; 373(23):2247–2257. doi:10.1056/NEJMoa1509225
  12. Holman RR, Bethel MA, Mentz RJ, et al; EXSCEL Study Group. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017; 377(13):1228–1239. doi:10.1056/NEJMoa1612917
  13. Cosmi F, Laini R, Nicolucci A. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2017; 376(9):890. doi:10.1056/NEJMc1615712
  14. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394(10193):121–130. doi:10.1016/S0140-6736(19)31149-3
  15. Zinman B, Wanner C, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015; 373(22):2117–2128. doi:10.1056/NEJMoa1504720
  16. Wanner C, Inzucchi SE, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016; 375(4):323–334. doi:10.1056/NEJMoa1515920
  17. Neal B, Perkovic V, Mahaffey KW, et al; CANVAS Program Collaborative Group. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017; 377(7):644–657. doi:10.1056/NEJMoa1611925
  18. Wiviott SD, Raz I, Bonaca MP, et al; DECLARE–TIMI 58 Investigators. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2018. [Epub ahead of print] doi:10.1056/NEJMoa1812389
  19. Diabetes Prevention Program Research Group; Knowler WC, Fowler SE, Hamman RF, et al. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 2009; 374(9702):1677–1686. doi:10.1016/S0140-6736(09)61457-4
  20. Marso SP, Bain SC, Consoli A, et al, for the SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016; 375:1834–1844. doi:10.1056/NEJMoa1607141
  21. Bolinder J, Ljunggren Ö, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab 2012; 97(3):1020–1031. doi:10.1210/jc.2011-2260
  22. Bonds DE, Miller ME, Bergenstal RM, et al. The association between symptomatic, severe hypoglycaemia and mortality in type 2 diabetes: retrospective epidemiological analysis of the ACCORD study. BMJ 2010; 340:b4909. doi:10.1136/bmj.b4909
  23. Saremi A, Bahn GD, Reaven PD; Veterans Affairs Diabetes Trial (VADT). A link between hypoglycemia and progression of atherosclerosis in the Veterans Affairs Diabetes Trial (VADT). Diabetes Care 2016; 39(3):448–454. doi:10.2337/dc15-2107
  24. American Diabetes Association. 8. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S73–S85. doi:10.2337/dc18-S008
  25. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  26. US Food and Drug Administration. FDA drug safety communication: FDA revises warnings regarding use of the diabetes medicine metformin in certain patients with reduced kidney function. www.fda.gov/Drugs/DrugSafety/ucm493244.htm. Accessed August 5, 2019.
  27. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9(2):194–205. doi:10.1111/j.1463-1326.2006.00704.x
  28. Gangji AS, Cukierman T, Gerstein HC, Goldsmith CH, Clase CM. A systematic review and meta-analysis of hypoglycemia and cardiovascular events: a comparison of glyburide with other secretagogues and with insulin. Diabetes Care 2007; 30(2):389–394. doi:10.2337/dc06-1789
  29. Nauck M, Frid A, Hermansen K, et al; LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32(1):84–90. doi:10.2337/dc08-1355
  30. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Accessed August 5, 2019.
  31. Boyle JP, Thompson TJ, Gregg EW, Barker LE, Williamson DF. Projection of the year 2050 burden of diabetes in the US adult population: dynamic modeling of incidence, mortality, and prediabetes prevalence. Popul Health Metr 2010; 8:29. doi:10.1186/1478-7954-8-29
  32. Sinclair AJ, Paolisso G, Castro M, Bourdel-Marchasson I, Gadsby R, Rodriguez Mañas L; European Diabetes Working Party for Older People. European Diabetes Working Party for Older People 2011 clinical guidelines for type 2 diabetes mellitus. Executive summary. Diabetes Metab 2011; 37(suppl 3):S27–S38. doi:10.1016/S1262-3636(11)70962-4
  33. American Geriatrics Society Expert Panel on Care of Older Adults with Diabetes Mellitus; Moreno G, Mangione CM, Kimbro L, Vaisberg E. Guidelines abstracted from the American Geriatrics Society Guidelines for Improving the Care of Older Adults with Diabetes Mellitus: 2013 update. J Am Geriatr Soc 2013; 61(11):2020–2026. doi:10.1111/jgs.12514
References
  1. Allen DW, Schroeder WA, Balog J. Observations on the chromatographic heterogeneity of normal adult and fetal human hemoglobin: a study of the effects of crystallization and chromatography on the heterogeneity and isoleucine content. J Amer Chem Soc 1958; 80(7):1628–1634. doi:10.1021/ja01540a030
  2. Huisman TH, Dozy AM. Studies on the heterogeneity of hemoglobin. V. Binding of hemoglobin with oxidized glutathione. J Lab Clin Med 1962; 60:302–319. pmid:14449875
  3. Rahbar S, Blumenfeld O, Ranney HM. Studies of an unusual hemoglobin in patients with diabetes mellitus. Biochem Biophys Res Commun 1969; 36(5):838–843. pmid:5808299
  4. American Diabetes Association. 6. Glycemic targets: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S55–S64. doi:10.2337/dc18-S006
  5. Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA 1979; 241(19):2035–2038. pmid:430798
  6. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352(9131):837–853. [Erratum in Lancet 1999; 354:602.] pmid:9742976
  7. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  8. Hayward RA, Reaven PD, Wiitala WL, et al; VADT Investigators. Follow-up of glycemic control and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015; 372(23):2197–2206. doi:10.1056/NEJMoa1414266
  9. US Food and Drug Administration. Guidance for industry: diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. https://www.govinfo.gov/content/pkg/FR-2008-12-19/pdf/E8-30086.pdf. Accessed August 6, 2019.
  10. Marso SP, Daniels GH, Brown-Frandsen K, et al; LEADER Steering Committee; LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375(4):311–322. doi:10.1056/NEJMoa1603827
  11. Pfeffer MA, Claggett B, Diaz R, et al; ELIXA Investigators. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015; 373(23):2247–2257. doi:10.1056/NEJMoa1509225
  12. Holman RR, Bethel MA, Mentz RJ, et al; EXSCEL Study Group. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017; 377(13):1228–1239. doi:10.1056/NEJMoa1612917
  13. Cosmi F, Laini R, Nicolucci A. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2017; 376(9):890. doi:10.1056/NEJMc1615712
  14. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394(10193):121–130. doi:10.1016/S0140-6736(19)31149-3
  15. Zinman B, Wanner C, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015; 373(22):2117–2128. doi:10.1056/NEJMoa1504720
  16. Wanner C, Inzucchi SE, Lachin JM, et al; EMPA-REG OUTCOME Investigators. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016; 375(4):323–334. doi:10.1056/NEJMoa1515920
  17. Neal B, Perkovic V, Mahaffey KW, et al; CANVAS Program Collaborative Group. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017; 377(7):644–657. doi:10.1056/NEJMoa1611925
  18. Wiviott SD, Raz I, Bonaca MP, et al; DECLARE–TIMI 58 Investigators. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2018. [Epub ahead of print] doi:10.1056/NEJMoa1812389
  19. Diabetes Prevention Program Research Group; Knowler WC, Fowler SE, Hamman RF, et al. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 2009; 374(9702):1677–1686. doi:10.1016/S0140-6736(09)61457-4
  20. Marso SP, Bain SC, Consoli A, et al, for the SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016; 375:1834–1844. doi:10.1056/NEJMoa1607141
  21. Bolinder J, Ljunggren Ö, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab 2012; 97(3):1020–1031. doi:10.1210/jc.2011-2260
  22. Bonds DE, Miller ME, Bergenstal RM, et al. The association between symptomatic, severe hypoglycaemia and mortality in type 2 diabetes: retrospective epidemiological analysis of the ACCORD study. BMJ 2010; 340:b4909. doi:10.1136/bmj.b4909
  23. Saremi A, Bahn GD, Reaven PD; Veterans Affairs Diabetes Trial (VADT). A link between hypoglycemia and progression of atherosclerosis in the Veterans Affairs Diabetes Trial (VADT). Diabetes Care 2016; 39(3):448–454. doi:10.2337/dc15-2107
  24. American Diabetes Association. 8. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes—2018. Diabetes Care 2018; 41(suppl 1):S73–S85. doi:10.2337/dc18-S008
  25. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 2008; 359(15):1577–1589. doi:10.1056/NEJMoa0806470
  26. US Food and Drug Administration. FDA drug safety communication: FDA revises warnings regarding use of the diabetes medicine metformin in certain patients with reduced kidney function. www.fda.gov/Drugs/DrugSafety/ucm493244.htm. Accessed August 5, 2019.
  27. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab 2007; 9(2):194–205. doi:10.1111/j.1463-1326.2006.00704.x
  28. Gangji AS, Cukierman T, Gerstein HC, Goldsmith CH, Clase CM. A systematic review and meta-analysis of hypoglycemia and cardiovascular events: a comparison of glyburide with other secretagogues and with insulin. Diabetes Care 2007; 30(2):389–394. doi:10.2337/dc06-1789
  29. Nauck M, Frid A, Hermansen K, et al; LEAD-2 Study Group. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009; 32(1):84–90. doi:10.2337/dc08-1355
  30. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Accessed August 5, 2019.
  31. Boyle JP, Thompson TJ, Gregg EW, Barker LE, Williamson DF. Projection of the year 2050 burden of diabetes in the US adult population: dynamic modeling of incidence, mortality, and prediabetes prevalence. Popul Health Metr 2010; 8:29. doi:10.1186/1478-7954-8-29
  32. Sinclair AJ, Paolisso G, Castro M, Bourdel-Marchasson I, Gadsby R, Rodriguez Mañas L; European Diabetes Working Party for Older People. European Diabetes Working Party for Older People 2011 clinical guidelines for type 2 diabetes mellitus. Executive summary. Diabetes Metab 2011; 37(suppl 3):S27–S38. doi:10.1016/S1262-3636(11)70962-4
  33. American Geriatrics Society Expert Panel on Care of Older Adults with Diabetes Mellitus; Moreno G, Mangione CM, Kimbro L, Vaisberg E. Guidelines abstracted from the American Geriatrics Society Guidelines for Improving the Care of Older Adults with Diabetes Mellitus: 2013 update. J Am Geriatr Soc 2013; 61(11):2020–2026. doi:10.1111/jgs.12514
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Issue
Cleveland Clinic Journal of Medicine - 86(9)
Page Number
595-600
Page Number
595-600
Publications
Publications
Topics
Article Type
Display Headline
Diabetes management: Beyond hemoglobin A1c
Display Headline
Diabetes management: Beyond hemoglobin A1c
Legacy Keywords
diabetes, drugs, hemoglobin A1c,management, glucagon-like peptide 1, GLP-1, GLP-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors, SGLT2 inhibitors, metformin, dipeptidyl peptidase 4 inhibitors, DPP-4 inhibitors, cardiovascular benefit, sulfonylureas, meglitinides, thiazolidinediones, incretins, liraglutide, lixisenatide, exenatide, semaglutide, dulaglutide, empagliflozin, canagliflozin, dapaagliflozin, weight management, hypoglycemia risk, Vinni Makin, M. Cecilia Lansang
Legacy Keywords
diabetes, drugs, hemoglobin A1c,management, glucagon-like peptide 1, GLP-1, GLP-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors, SGLT2 inhibitors, metformin, dipeptidyl peptidase 4 inhibitors, DPP-4 inhibitors, cardiovascular benefit, sulfonylureas, meglitinides, thiazolidinediones, incretins, liraglutide, lixisenatide, exenatide, semaglutide, dulaglutide, empagliflozin, canagliflozin, dapaagliflozin, weight management, hypoglycemia risk, Vinni Makin, M. Cecilia Lansang
Sections
Inside the Article

KEY POINTS

  • Some glucagon-like peptide 1 (GLP-1) receptor agonists have been shown to reduce cardiovascular risk, and liraglutide carries an indication for this use.
  • The sodium-glucose cotransporter 2 inhibitors empaglifozin and canaglifozin carry indications to prevent cardiovascular death in patients with diabetes with established cardiovascular disease.
  • Metformin, GLP-1 receptor agonists, and dipeptidyl peptidase 4 inhibitors are beneficial in terms of promoting weight loss—or at least not causing weight gain.
  • Disadvantages and adverse effects of various drugs must also be considered.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/22/2019 - 09:00
Un-Gate On Date
Thu, 08/22/2019 - 09:00
Use ProPublica
CFC Schedule Remove Status
Thu, 08/22/2019 - 09:00
Hide sidebar & use full width
render the right sidebar.
Article PDF Media