V Receptor Antagonists for Treatment of Hyponatremia

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
New horizons in the pharmacologic approach to hyponatremia: The V2 receptor antagonists

Under normal circumstances, there is a balance between water intake and water excretion such that plasma osmolality and the serum sodium (Na+) concentration remain relatively constant. The principal mechanism responsible for prevention of hyponatremia and hyposmolality is renal water excretion. In all hyponatremic patients, water intake exceeds renal water excretion.

Excretion of water by the kidney is dependent on 3 factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be preserved so that the tubular fluid will be diluted. Lastly, arginine vasopressin (AVP) levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. Given the central role of AVP in limiting renal water excretion, AVP receptor antagonists represent a physiologic and rational method to increase renal water excretion.

AVP in Regulation of Plasma Osmolality

AVP is synthesized in the supraoptic and paraventricular nucleus of the hypothalamus and then stored in the neurohypophysis (reviewed in the article Diagnostic Approach and Management of Inpatient Hyponatremia in this supplement). The release of AVP is exquisitely sensitive to changes in plasma osmolality. AVP is not detectable in the plasma at an osmolality below approximately 280 mOsm/kg but increases in a nearly linear fashion beginning with as little as a 2% to 3% increase in osmolality above this value. The extreme sensitivity of this system allows for plasma osmolality to be maintained within a narrow range.

A second major determinant of AVP release is the effective arterial blood volume. While AVP levels are very sensitive to plasma osmolality, small changes of <10% in blood pressure or blood volume have no effect on AVP levels. However, once decreases in volume or pressure exceed this value, baroreceptor‐mediated signals provide persistent stimuli for AVP secretion. Baroreceptor‐mediated AVP release will continue even when plasma osmolality falls below 280 mOsm/kg. Teleologically, this system can be viewed as an emergency mechanism to defend blood pressure. Thus, small decreases in blood volume and blood pressure will cause the body to retain NaCl which will raise osmolality and lead to water retention. However, if NaCl is not available and if blood pressure and volume are becoming dangerously low (down 10%), the body behaves as if defense of blood pressure is more important than defense of osmolality, and AVP is secreted.1 The specific compartment whose volume is sensed in order to determine AVP secretion in this setting is the effective arterial volume. This overriding effect of volume explains the persistence of high AVP levels in hyponatremic patients with conditions such as heart failure and cirrhosis.

Other stimuli for the release of AVP include pain, nausea, and hypoxia. Inappropriate release of AVP can occur with a variety of central nervous system and pulmonary diseases as well as with drugs, particularly those that act within the central nervous system.2 Certain tumors can synthesize and release AVP.

AVP exerts its effects on cells through 3 receptors (Table 1). The V1A receptor is expressed in a variety of tissues but is primarily found on vascular smooth muscle cells. Stimulation of this receptor results in vasoconstriction, platelet aggregation, inotropic stimulation and myocardial protein synthesis. The V1B receptor is expressed in cells of the anterior pituitary and throughout the brain. Stimulation of this receptor results in release of adrenocorticotropin stimulating hormone (ACTH). Stimulation of the V1A and V1B receptors activate phospholipase C leading to increases in inositol trisphosphate and diacylglycerol with secondary increases in cell calcium and activation of protein kinase C.

The V2 receptor is found on the basolateral surface of the renal collecting duct and vascular endothelium where it mediates the antidiuretic effects of AVP and stimulates the release of von Willebrand factor respectively. Unlike the V1A and V1B receptors, binding of AVP to the V2 receptor activates the GS‐coupled adenyl cyclase system causing increased intracellular levels of cyclic adenosine monophosphate (cAMP). In the kidney, generation of cAMP stimulates protein kinase A which then phosphorylates preformed aquaporin‐2 water channels causing trafficking and insertion of the channels into the luminal membrane of the tubular cells.3 The insertion of the aquaporin‐2 protein renders the collecting duct selectively permeable to water, which is then reabsorbed from the tubular lumen into the blood driven by the osmotic driving force of the hypertonic interstitium. In the absence of AVP, aquaporin membrane insertion and apical membrane water permeability are dramatically reduced.

Physiologic Rationale for Use of AVP Antagonists

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of the aquaporin‐2 water channels into the luminal membrane. The increase in urine output is similar in quantity to diuretics but differs in content. V2 receptor antagonists increase water excretion with little to no change in urinary electrolytes. As a result, lowering of the serum K+ level, metabolic alkalosis, and increases in the serum creatinine and blood urea nitrogen concentration are avoided in contrast to diuretics such as furosemide and hydrochlorothiazide. In addition, orthostatic hypotension and activation of neurohumoral effectors such as angiotensin II, circulating catecholamines, and aldosterone are not features of V2 receptor blockade. These differences have lead to V2 receptor antagonists being characterized as aquaretic agents so as to distinguish them from diuretics.

The physiologic rationale for use of V2 receptor antagonists is best exemplified by considering the relationship between the serum Na+ concentration and the total body content of Na+, K+, and water approximated by the equation: 2 receptor antagonist) is approved by the Food and Drug Administration (FDA) for euvolemic and hypervolemic hyponatremia. The effectiveness of the drug was demonstrated in a randomized placebo controlled trial of 84 hospitalized patients with euvolemic or hypervolemic hyponatremia in which the serum Na+ ranged from 115 to <130 mEq/L.5 The patients were given a 20‐mg loading dose followed by a continuous infusion of either 40 or 80 mg daily for 4 days. Conivaptan significantly raised the serum Na+ concentration by 6.3 and 9.4 in the 40 and 80 mg/day arms respectively as compared to 0.8 mEq/L in the placebo group.

Conivaptan is also active as an oral formulation but its distribution has been restricted to parenteral use for short‐term (4 days maximum) in‐hospital administration only. This restriction is due to potent inhibitory effects of the drug on the hepatic cytochrome P450 3A4 enzyme system and the potential for untoward drug interactions. The inhibitory effects of other members of this class on the cytochrome P450 3A4 (CYP3A4) system are more limited, allowing for oral formulations to be used in a clinical setting.

Tolvaptan is the only other AVP antagonist currently approved by the FDA. Unlike conivaptan, tolvaptan is an oral agent with effects confined to the V2 receptor. The drug is indicated for the treatment of clinically significant hypervolemic and euvolemic hyponatremia.

The efficacy of tolvaptan was evaluated in 2 simultaneously conducted multicenter, randomized, double blind trials called Study of Ascending Levels of Tolvaptan in hyponatremia 1 and 2 (SALT 1 and SALT 2).6 The 2 studies together randomized 225 patients with euvolemic or hypervolemic hyponatremia to outpatient treatment with the study drug for 30 days and 223 patients to placebo. Patients with a serum Na+ of <120 mEq/L in association with neurologic impairment were excluded from the trial. Serum Na+ increased more in the tolvaptan group than in the placebo during the first 4 days (3.62 vs. 0.25 mEq/L). After 30 days of therapy serum Na+ concentrations were 6.22 higher in the tolvaptan group compared with 1.66 mEq/L in the placebo group. When tolvaptan was discontinued at study end, the serum Na+ fell over a seven day period to a value similar to that in the placebo treated patients. Four patients (1.8%) exceeded the study goal of limiting correction of the hyponatremia to <12 mEq/L in the first 24 hours of treatment, but none of these patients developed adverse clinical sequelae.

As previously mentioned conivaptan and tolvaptan are indicated for the treatment of hyponatremia in the setting of euvolemia or hypervolemia. These drugs should not be used in hypovolemic states since the increase in renal water excretion can potentially predispose to worsening hemodynamics in the setting of volume depletion. To be sure, any volume of water removed from the body is principally derived from the intracellular compartment (two‐thirds) and would not be expected to affect blood pressure to a major extent. However, one‐twelfth of any water volume loss is derived from the circulating compartment and could potentially aggravate a borderline low blood pressure in the setting of volume depletion.

This concern is particularly true for conivaptan and its blocking effects on the V1a receptor. AVP can cause peripheral vasoconstriction by stimulating the V1a receptor on the peripheral vasculature.7 However, circulating concentrations observed in euvolemic and hypervolemic conditions are not typically of a magnitude to elicit this effect thus explaining the lack of clinically significant hypotension in clinical trials. By contrast, AVP may reach a high enough concentration and play a contributory role in the maintenance of blood pressure under conditions of significant depletion of extracellular fluid volume (hemorrhage) or in states of generalized vasodilation such as sepsis or advanced cirrhosis. In these settings blockade of the V1a receptor may result in significant hypotension.

Another theoretical concern of blocking the V1a receptor with conivaptan is the potential to cause further sequestration of fluid in the splanchnic vascular bed and theoretically increase the risk of hepatorenal syndrome.8 The importance of splanchnic vasodilation in the genesis of renal hypoperfusion has been indirectly illustrated by the response to ornipressin, an analog of AVP that is a preferential splanchnic vasoconstrictor. The administration of ornipressin to patients with advanced cirrhosis leads to correction of many of the systemic and renal hemodynamic abnormalities that are present. These include an elevation in mean arterial pressure, reductions in plasma renin activity and norepinephrine concentration, and increases in renal blood flow, glomerular filtration rate, and urinary sodium excretion and volume. V1a receptor blockade has the potential to increase the degree of arterial vasodilation in the splanchnic arteriolar bed. Increasing degrees of splanchnic vasodilation contribute to a fall in mean arterial pressure and unloading of baroreceptors in the central circulation. As a result, central afferent sensors signal the activation of neurohumoral effectors which in turn decrease perfusion of other organs, particularly the kidney.

AVP Antagonists in Severe Symptomatic Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, restlessness, confusion and disorientation.9 Life threatening symptoms are those of impending brain herniation and include seizures, decreased levels consciousness, and obtundation. Hypertonic saline remains the treatment of choice in those patients who are clinically determined to be severely symptomatic.10 Vaptan therapy does result in a brisk and relatively prolonged water diuresis. Indeed, in the first 6 hours following the parenteral administration of conivaptan there is a several mEq/L increase in the serum Na+ concentration.5 Whether this time course of correction is sufficient to abort fatal hyponatremic encephalopathy is simply not known. Such patients to date have been excluded from clinical studies since randomization in a placebo‐controlled trial would be clearly unethical in such subjects, and not using hypertonic saline might also be considered unsafe

Although not well studied, one could coadminister conivaptan along with hypertonic saline and anticipate a more rapid initial rise in the serum Na+. Once stabilized, the hypertonic saline could be discontinued and the remainder of correction be accomplished by use of the receptor antagonist alone. This strategy would help to minimize the likelihood of volume overload due to the use of hypertonic saline but would require frequent monitoring of the serum Na+ to ensure the rate of correction was within accepted guidelines.

AVP Antagonists in Patients With Mild to Moderate Symptoms of Hyponatremia

Hospitalized patients with mild to moderate symptoms of hyponatremia can be considered ideal candidates for the use of V2 receptor antagonists. Both conivaptan and tolvaptan can be expected to increase the serum Na+ concentration to a greater extent and more predictably than fluid restriction alone. The superiority of these drugs would be particularly evident in those who require ongoing fluid administration for any number of reasons such as parenteral administration of antibiotics or proton pump inhibitors.11

In hyponatremic patients with decompensated heart failure there may be changes in mental status in which it is difficult to separate out the contribution of hyponatremia from the decrease in cerebral perfusion. Vaptan therapy offers a predictable way to improve the serum Na+ contribution over an acceptable period of time thereby removing one of the variables. Even if hyponatremia is not the direct cause of symptoms, it may lower the threshold for mental status changes resulting from poor cerebral perfusion. A similar argument for vaptan therapy can be made in hyponatremic patients with cirrhosis. In this population mental status changes can be due to the hyponatremia, the underlying liver disease, or both. If no improvement occurred following the correction of the hyponatremia, therapy could then be focused on the underlying liver disease. In fact, in any delirious patient with hyponatremia, correction of the underlying electrolyte disorder can help to clarify the degree to which the hyponatremia is exacerbating the altered mental status.

There are no head to head studies comparing conivaptan and tolvaptan in patients with mild to moderate symptoms of hyponatremia. Presumably both drugs would work with similar efficacy. Conivaptan therapy can be complicated by phlebitis when administered through a peripheral vein due to the irritative effects of polypropylene glycol which serves as a diluent for the drug.12 For this reason the drug is often given through a central vein. The theoretical risk of conivaptan's V1a receptor blocking effects in patients with cirrhosis has already been discussed. Tolvaptan is given orally starting at 15 mg daily. Depending on the response the dose can be increased to 30 mg and ultimately to 60 mg daily. Few patients in the SALT 1 and 2 trials required the 60 mg dose.

Measurement of the urine osmolality may be useful in predicting how responsive a patient will be to the administration of a vaptan. In this regard, urine osmolality can be thought as a biomarker of the effect of AVP on the renal collecting duct.13 The higher the urine osmolality, presumably the higher the serum AVP level or the greater the effect of AVP is on the tubule. By contrast, those with a lower urine osmolality presumably have less AVP effect on the tubule and therefore will manifest a less robust response to a V2 receptor blocker.

There are circumstances unique to the hospitalized patient where one may consider use of a V2 receptor antagonist. Consider a patient admitted with pneumonia complicated by hyponatremia (Na+ = 128 mEq/L) due to syndrome of inappropriate antidiuretic hormone secretion (SIADH) who is treated with intravenous antibiotics, stabilized, and is now being considered for discharge. Despite fluid restriction the serum Na+ concentration is currently 126 mEq/L and the admitting physician is reluctant to discharge the patient for fear the Na+ may fall further and result in symptomatic hyponatremia. The hospital discharge is postponed and fluid restriction is intensified. Two days later the serum Na+ concentration has risen to 131 mEq/L and the patient is discharged. While one can question the wisdom of postponing the original planned discharge based on the lab value alone, this type of scenario is common in clinical practice. Fluid restriction is poorly tolerated, difficult to enforce, and often unpredictable in response. The administration of a single dose of a V2 receptor antagonist is a reliable way to increase the serum Na+ concentration over a 24 hour period and in this case could have potentially shortened the hospital stay.

Consider another patient admitted with a hip fracture who is found to have a serum Na+ concentration of 126 meq/L attributed to ongoing use of an antidepressant agent. The patient is otherwise medically stable but the anesthesiologist is unwilling to accept the patient for surgery until the serum Na+ is at least >130 mEq/L. Fluid restriction is prescribed and only 2 to 3 days later has the serum Na+ reached a value of 131 mEq/L. Once again, administration of a vaptan would have almost certainly increased the Na+ concentration to the desired threshold over a 24 hour period, allowing the patient to undergo the surgical procedure without experiencing the undue delay.

In each of these scenarios short term use of a vaptan would provide for a more rapid and predictable increase in the serum Na+ concentration and potentially decrease the length of hospital stay in comparison to fluid restriction alone. In addition to being less predictable and often poorly tolerated, fluid restriction is often ineffective due to the obligatory fluid administration hospitalized patients require for other therapies or nutrition. Demeclocycline and lithium have been used to antagonize the effects of AVP on the tubule but these drugs take several days before any demonstrable increase in renal water excretion is seen. Demeclocycline antagonizes the effect of AVP through nephrotoxic effects on the tubular cell whereas vaptan's are competitive inhibitors of the V2 receptor and are not associated with nephrotoxicity. Lithium interferes in the intracellular signaling pathways by which AVP causes insertion of water channels into the apical membrane. The doses required to illicit an increase in renal water excretion are near those which can result in lithium levels sufficient to cause neurotoxicity. The primary side effect of vaptan therapy are those one would predict from inducing an aquaretic effect and include thirst, increased urinary frequency, and increased urinary volume.

The decision to use a vaptan on a more prolonged basis is made on a case by case basis. When the underlying cause of increased AVP is deemed to be chronic and irreversible then therapy can be extended into the outpatient setting. Consideration for more chronic therapy would be appropriate for patients with SIADH due to underlying cancer, or those with severe chronic congestive heart failure or advanced cirrhosis. If the cause of the increased AVP is transient, then 1 to 2 doses of a vaptan while in the hospital may be all that is required. Transient causes of increased AVP would include drug related causes, acute pneumonia, hypoxia associated with respiratory failure, and acute decompensated heart failure. Once again vaptan therapy should not be used when the cause of increased AVP is due to total body salt depletion.

AVP Antagonists in Asymptomatic Hyponatremic Patients

One remaining question concerning the use of vaptan therapy is whether they are helpful in hyponatremic patients who are asymptomatic. There are several observations which raise the possibility that patients deemed to be otherwise asymptomatic do in fact have subtle abnormalities attributable to the low Na+ concentration and therefore could benefit if the hyponatremia was corrected. First, some patients with a serum Na+ in the range of 120 mEq/L to 129 mEq/L have subtle neurologic changes that improve when the serum Na+ concentration is increased.14 These include scores on tests of mental and social functioning. In the SALT 1 and 2 trials a general health survey filled out by the patients showed tolvaptan therapy was associated with improvements in the mental health component of the instrument assessing parameters such as vitality, social functioning, emotionally limited accomplishments, calmness, and sadness.6 Response time is delayed and the error rate is increased in response to various stimuli while patients are hyponatremic, suggesting hyponatremia is associated with reversible impairment in attention. It is certainly reasonable to speculate that correction of hyponatremia in an elderly patient with SIADH might improve that individual's quality of life by allowing them to better enjoy any number of activities such as reading or completing a crossword puzzle. The presence of concomitant neuropsychiatric disturbance (delirium and dementia in particular) may also be relevant, since even mild hyponatremia may increase vulnerability to further alterations in mental status in patients who are impaired at baseline.

Second, asymptomatic hyponatremic patients exhibit subtle disturbances in gait that improve following correction of the serum Na+ concentration.14, 15 In this regard, case control studies have shown an association between hyponatremia and risk of falls and fractures particularly in the elderly population.15, 16 Chronic hyponatremia has been shown to cause a reduction in bone mass in an experimental model of SIADH and hyponatremia is associated with osteoporosis in cross‐sectional human data.17 Fall‐related injury is associated with substantial adverse psychological and physical outcomes and is a cause of both death and disability in this population. It would be of considerable socioeconomic benefit if treatment of asymptomatic hyponatremia were able to reduce the risk of this complication.

Lastly, hyponatremia is associated with increased morbidity and mortality but generally the low Na+ concentration is merely thought to be marker of the severity of the underlying disease and not a direct contributor to the adverse outcome.18, 19 For example in decompensated heart failure there is an inverse relationship between the degree of hyponatremia and the extent of neurohormonal activation. Adverse effects resulting from persistent activation of the renin‐angiotensin‐aldosterone system and sympathetic nerve activity are thought to be the mechanism underlying the increase in mortality in hyponatremic patients with heart failure. Nevertheless, it has been suggested that hyponatremia itself, the associated hypotonicity, and/or elevated levels of AVP might exert adverse effects on the cardiovascular system or other organ systems and therefore play a contributory role in patient morbidity and mortality. While it remains speculative as to whether correction of the hyponatremia per se will improve patient outcomes, the V2 receptor antagonists offer an opportunity to test this uncertainty in patients with euvolemic and hypervolemic hyponatremia.0, 0

Location and Function of AVP Receptor Subtypes
Subtype Location Function
  • Abbreviations: ACTH, adrenocorticotropin stimulating hormone; AVP, arginine vasopressin.

V1A Vascular smooth muscle to include splanchnic bed Vasoconstriction
V1B Anterior pituitary Release of ACTH
V2 Basolateral surface of renal collecting duct, vascular endothelium, vascular smooth muscle Insertion of aquaporin channel into luminal membrane, release of von Willebrand factor, vasodilation
AVP Receptor Antagonists Currently Approved by FDA
Parameter Tolvaptan Conivaptan
  • Abbreviations: AVP: arginine vasopressin; CYP3A4, cytochrome P450 3A4; FDA, Food and Drug Administration.

Trade name Samsca Vaprisol
Administration Oral Intravenous
Dose 1560 mg daily 20 mg loading dose followed by 2040 mg continuous infusion
Receptor V2 V1A and V2
Protein binding (%) 99 98.5
Half life 68 hours 38 hours
Metabolism Hepatic (CYP3A4) Hepatic (CYP3A4)
Elimination Feces Feces
References
  1. Palmer BF,Alpern RJ.Integrated Response to Abnormalities in Tonicity. In: Seldin DW, Giebisch G, eds.Clinical Disturbances of Water Metabolism.New York, New York:Raven Press Ltd;1993:273295.
  2. Palmer BF,Gates JR,Lader M.Causes and management of hyponatremia.Ann Pharmacother.2003;37:16941702.
  3. Nedvetsky P,Tamma G,Beulshausen S,Valenti G,Rosenthal W,Klussmann E.Regulation of aquaporin‐2 trafficking.Handb Exp Pharmacol.2009;190:133157.
  4. Chen S,Jalandhara N,Batlle D.Evaluation and management of hyponatremia: an emerging role for vasopressin receptor antagonists.Nat Clin Pract Nephrol.2007;3:8295.
  5. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
  6. Schrier R,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:2099112.
  7. Koshimizu T,Nasa Y,Tanoue A, et al.V1a vasopressin receptors maintain normal blood pressure by regulating circulating blood volume and baroreflex sensitivity.Proc Natl Acad Sci.2006;103:78077812.
  8. Palmer B.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  9. Adrogue H,Madias N.Hyponatremia.N Engl J Med.2000;342:15811589.
  10. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  11. Palmer BF.Hyponatremia in the intensive care unit.Semin Nephrol.2009;29:257270.
  12. Ghali J,Farah J,Daifallah S,Zabalawi H,Zmily H.Conivaptan and its role in the treatment of hyponatremia.Drug Des Devel Ther.2009;3:253268.
  13. Verbalis J.Vaptans for the treatment of hyponatremia: how who when and why.Nephrol Self Assess Program.2007;6:199209.
  14. Decaux G.The syndrome of inappropriate secretion of antidiuretic hormone (SIADH).Semin Nephrol.2009;29(3):239256.
  15. Kengne F,Andres C,Sattar L,Melot C,Decaux C.Mild hyponatremia and risk of fracture in the ambulatory elderly.Q J Med.2008;101:583588.
  16. Kinsella S,Moran S,Sullivan M,Molloy M,Eustace J.Hyponatremia independent of osteoporosis is associated with fracture occurrence.Clin J Am Soc Nephrol.2010 (in press).
  17. Verbalis J,Barsony J,Sugimura Y, et al.Hyponatremia‐induced osteoporosis.J Bone Miner Res.2009;999:137.
  18. Wald R,Jaber B,Price L,Upadhyay A,Madias, N.Impact of hospital‐associated hyponatremia on selected outcomes.Arch Intern Med.2010;170(3):294302.
  19. Waikar S,Mount D,Curhan G, et al.Mortality after hospitalization with mild, moderate, and severe hyponatremia.Am J Med.2009;122:857865.
Article PDF
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S27-S32
Legacy Keywords
arginine vasopressin, AVP receptor antagonists, conivaptan, tolvaptan, hyponatremia
Sections
Article PDF
Article PDF

Under normal circumstances, there is a balance between water intake and water excretion such that plasma osmolality and the serum sodium (Na+) concentration remain relatively constant. The principal mechanism responsible for prevention of hyponatremia and hyposmolality is renal water excretion. In all hyponatremic patients, water intake exceeds renal water excretion.

Excretion of water by the kidney is dependent on 3 factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be preserved so that the tubular fluid will be diluted. Lastly, arginine vasopressin (AVP) levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. Given the central role of AVP in limiting renal water excretion, AVP receptor antagonists represent a physiologic and rational method to increase renal water excretion.

AVP in Regulation of Plasma Osmolality

AVP is synthesized in the supraoptic and paraventricular nucleus of the hypothalamus and then stored in the neurohypophysis (reviewed in the article Diagnostic Approach and Management of Inpatient Hyponatremia in this supplement). The release of AVP is exquisitely sensitive to changes in plasma osmolality. AVP is not detectable in the plasma at an osmolality below approximately 280 mOsm/kg but increases in a nearly linear fashion beginning with as little as a 2% to 3% increase in osmolality above this value. The extreme sensitivity of this system allows for plasma osmolality to be maintained within a narrow range.

A second major determinant of AVP release is the effective arterial blood volume. While AVP levels are very sensitive to plasma osmolality, small changes of <10% in blood pressure or blood volume have no effect on AVP levels. However, once decreases in volume or pressure exceed this value, baroreceptor‐mediated signals provide persistent stimuli for AVP secretion. Baroreceptor‐mediated AVP release will continue even when plasma osmolality falls below 280 mOsm/kg. Teleologically, this system can be viewed as an emergency mechanism to defend blood pressure. Thus, small decreases in blood volume and blood pressure will cause the body to retain NaCl which will raise osmolality and lead to water retention. However, if NaCl is not available and if blood pressure and volume are becoming dangerously low (down 10%), the body behaves as if defense of blood pressure is more important than defense of osmolality, and AVP is secreted.1 The specific compartment whose volume is sensed in order to determine AVP secretion in this setting is the effective arterial volume. This overriding effect of volume explains the persistence of high AVP levels in hyponatremic patients with conditions such as heart failure and cirrhosis.

Other stimuli for the release of AVP include pain, nausea, and hypoxia. Inappropriate release of AVP can occur with a variety of central nervous system and pulmonary diseases as well as with drugs, particularly those that act within the central nervous system.2 Certain tumors can synthesize and release AVP.

AVP exerts its effects on cells through 3 receptors (Table 1). The V1A receptor is expressed in a variety of tissues but is primarily found on vascular smooth muscle cells. Stimulation of this receptor results in vasoconstriction, platelet aggregation, inotropic stimulation and myocardial protein synthesis. The V1B receptor is expressed in cells of the anterior pituitary and throughout the brain. Stimulation of this receptor results in release of adrenocorticotropin stimulating hormone (ACTH). Stimulation of the V1A and V1B receptors activate phospholipase C leading to increases in inositol trisphosphate and diacylglycerol with secondary increases in cell calcium and activation of protein kinase C.

The V2 receptor is found on the basolateral surface of the renal collecting duct and vascular endothelium where it mediates the antidiuretic effects of AVP and stimulates the release of von Willebrand factor respectively. Unlike the V1A and V1B receptors, binding of AVP to the V2 receptor activates the GS‐coupled adenyl cyclase system causing increased intracellular levels of cyclic adenosine monophosphate (cAMP). In the kidney, generation of cAMP stimulates protein kinase A which then phosphorylates preformed aquaporin‐2 water channels causing trafficking and insertion of the channels into the luminal membrane of the tubular cells.3 The insertion of the aquaporin‐2 protein renders the collecting duct selectively permeable to water, which is then reabsorbed from the tubular lumen into the blood driven by the osmotic driving force of the hypertonic interstitium. In the absence of AVP, aquaporin membrane insertion and apical membrane water permeability are dramatically reduced.

Physiologic Rationale for Use of AVP Antagonists

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of the aquaporin‐2 water channels into the luminal membrane. The increase in urine output is similar in quantity to diuretics but differs in content. V2 receptor antagonists increase water excretion with little to no change in urinary electrolytes. As a result, lowering of the serum K+ level, metabolic alkalosis, and increases in the serum creatinine and blood urea nitrogen concentration are avoided in contrast to diuretics such as furosemide and hydrochlorothiazide. In addition, orthostatic hypotension and activation of neurohumoral effectors such as angiotensin II, circulating catecholamines, and aldosterone are not features of V2 receptor blockade. These differences have lead to V2 receptor antagonists being characterized as aquaretic agents so as to distinguish them from diuretics.

The physiologic rationale for use of V2 receptor antagonists is best exemplified by considering the relationship between the serum Na+ concentration and the total body content of Na+, K+, and water approximated by the equation: 2 receptor antagonist) is approved by the Food and Drug Administration (FDA) for euvolemic and hypervolemic hyponatremia. The effectiveness of the drug was demonstrated in a randomized placebo controlled trial of 84 hospitalized patients with euvolemic or hypervolemic hyponatremia in which the serum Na+ ranged from 115 to <130 mEq/L.5 The patients were given a 20‐mg loading dose followed by a continuous infusion of either 40 or 80 mg daily for 4 days. Conivaptan significantly raised the serum Na+ concentration by 6.3 and 9.4 in the 40 and 80 mg/day arms respectively as compared to 0.8 mEq/L in the placebo group.

Conivaptan is also active as an oral formulation but its distribution has been restricted to parenteral use for short‐term (4 days maximum) in‐hospital administration only. This restriction is due to potent inhibitory effects of the drug on the hepatic cytochrome P450 3A4 enzyme system and the potential for untoward drug interactions. The inhibitory effects of other members of this class on the cytochrome P450 3A4 (CYP3A4) system are more limited, allowing for oral formulations to be used in a clinical setting.

Tolvaptan is the only other AVP antagonist currently approved by the FDA. Unlike conivaptan, tolvaptan is an oral agent with effects confined to the V2 receptor. The drug is indicated for the treatment of clinically significant hypervolemic and euvolemic hyponatremia.

The efficacy of tolvaptan was evaluated in 2 simultaneously conducted multicenter, randomized, double blind trials called Study of Ascending Levels of Tolvaptan in hyponatremia 1 and 2 (SALT 1 and SALT 2).6 The 2 studies together randomized 225 patients with euvolemic or hypervolemic hyponatremia to outpatient treatment with the study drug for 30 days and 223 patients to placebo. Patients with a serum Na+ of <120 mEq/L in association with neurologic impairment were excluded from the trial. Serum Na+ increased more in the tolvaptan group than in the placebo during the first 4 days (3.62 vs. 0.25 mEq/L). After 30 days of therapy serum Na+ concentrations were 6.22 higher in the tolvaptan group compared with 1.66 mEq/L in the placebo group. When tolvaptan was discontinued at study end, the serum Na+ fell over a seven day period to a value similar to that in the placebo treated patients. Four patients (1.8%) exceeded the study goal of limiting correction of the hyponatremia to <12 mEq/L in the first 24 hours of treatment, but none of these patients developed adverse clinical sequelae.

As previously mentioned conivaptan and tolvaptan are indicated for the treatment of hyponatremia in the setting of euvolemia or hypervolemia. These drugs should not be used in hypovolemic states since the increase in renal water excretion can potentially predispose to worsening hemodynamics in the setting of volume depletion. To be sure, any volume of water removed from the body is principally derived from the intracellular compartment (two‐thirds) and would not be expected to affect blood pressure to a major extent. However, one‐twelfth of any water volume loss is derived from the circulating compartment and could potentially aggravate a borderline low blood pressure in the setting of volume depletion.

This concern is particularly true for conivaptan and its blocking effects on the V1a receptor. AVP can cause peripheral vasoconstriction by stimulating the V1a receptor on the peripheral vasculature.7 However, circulating concentrations observed in euvolemic and hypervolemic conditions are not typically of a magnitude to elicit this effect thus explaining the lack of clinically significant hypotension in clinical trials. By contrast, AVP may reach a high enough concentration and play a contributory role in the maintenance of blood pressure under conditions of significant depletion of extracellular fluid volume (hemorrhage) or in states of generalized vasodilation such as sepsis or advanced cirrhosis. In these settings blockade of the V1a receptor may result in significant hypotension.

Another theoretical concern of blocking the V1a receptor with conivaptan is the potential to cause further sequestration of fluid in the splanchnic vascular bed and theoretically increase the risk of hepatorenal syndrome.8 The importance of splanchnic vasodilation in the genesis of renal hypoperfusion has been indirectly illustrated by the response to ornipressin, an analog of AVP that is a preferential splanchnic vasoconstrictor. The administration of ornipressin to patients with advanced cirrhosis leads to correction of many of the systemic and renal hemodynamic abnormalities that are present. These include an elevation in mean arterial pressure, reductions in plasma renin activity and norepinephrine concentration, and increases in renal blood flow, glomerular filtration rate, and urinary sodium excretion and volume. V1a receptor blockade has the potential to increase the degree of arterial vasodilation in the splanchnic arteriolar bed. Increasing degrees of splanchnic vasodilation contribute to a fall in mean arterial pressure and unloading of baroreceptors in the central circulation. As a result, central afferent sensors signal the activation of neurohumoral effectors which in turn decrease perfusion of other organs, particularly the kidney.

AVP Antagonists in Severe Symptomatic Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, restlessness, confusion and disorientation.9 Life threatening symptoms are those of impending brain herniation and include seizures, decreased levels consciousness, and obtundation. Hypertonic saline remains the treatment of choice in those patients who are clinically determined to be severely symptomatic.10 Vaptan therapy does result in a brisk and relatively prolonged water diuresis. Indeed, in the first 6 hours following the parenteral administration of conivaptan there is a several mEq/L increase in the serum Na+ concentration.5 Whether this time course of correction is sufficient to abort fatal hyponatremic encephalopathy is simply not known. Such patients to date have been excluded from clinical studies since randomization in a placebo‐controlled trial would be clearly unethical in such subjects, and not using hypertonic saline might also be considered unsafe

Although not well studied, one could coadminister conivaptan along with hypertonic saline and anticipate a more rapid initial rise in the serum Na+. Once stabilized, the hypertonic saline could be discontinued and the remainder of correction be accomplished by use of the receptor antagonist alone. This strategy would help to minimize the likelihood of volume overload due to the use of hypertonic saline but would require frequent monitoring of the serum Na+ to ensure the rate of correction was within accepted guidelines.

AVP Antagonists in Patients With Mild to Moderate Symptoms of Hyponatremia

Hospitalized patients with mild to moderate symptoms of hyponatremia can be considered ideal candidates for the use of V2 receptor antagonists. Both conivaptan and tolvaptan can be expected to increase the serum Na+ concentration to a greater extent and more predictably than fluid restriction alone. The superiority of these drugs would be particularly evident in those who require ongoing fluid administration for any number of reasons such as parenteral administration of antibiotics or proton pump inhibitors.11

In hyponatremic patients with decompensated heart failure there may be changes in mental status in which it is difficult to separate out the contribution of hyponatremia from the decrease in cerebral perfusion. Vaptan therapy offers a predictable way to improve the serum Na+ contribution over an acceptable period of time thereby removing one of the variables. Even if hyponatremia is not the direct cause of symptoms, it may lower the threshold for mental status changes resulting from poor cerebral perfusion. A similar argument for vaptan therapy can be made in hyponatremic patients with cirrhosis. In this population mental status changes can be due to the hyponatremia, the underlying liver disease, or both. If no improvement occurred following the correction of the hyponatremia, therapy could then be focused on the underlying liver disease. In fact, in any delirious patient with hyponatremia, correction of the underlying electrolyte disorder can help to clarify the degree to which the hyponatremia is exacerbating the altered mental status.

There are no head to head studies comparing conivaptan and tolvaptan in patients with mild to moderate symptoms of hyponatremia. Presumably both drugs would work with similar efficacy. Conivaptan therapy can be complicated by phlebitis when administered through a peripheral vein due to the irritative effects of polypropylene glycol which serves as a diluent for the drug.12 For this reason the drug is often given through a central vein. The theoretical risk of conivaptan's V1a receptor blocking effects in patients with cirrhosis has already been discussed. Tolvaptan is given orally starting at 15 mg daily. Depending on the response the dose can be increased to 30 mg and ultimately to 60 mg daily. Few patients in the SALT 1 and 2 trials required the 60 mg dose.

Measurement of the urine osmolality may be useful in predicting how responsive a patient will be to the administration of a vaptan. In this regard, urine osmolality can be thought as a biomarker of the effect of AVP on the renal collecting duct.13 The higher the urine osmolality, presumably the higher the serum AVP level or the greater the effect of AVP is on the tubule. By contrast, those with a lower urine osmolality presumably have less AVP effect on the tubule and therefore will manifest a less robust response to a V2 receptor blocker.

There are circumstances unique to the hospitalized patient where one may consider use of a V2 receptor antagonist. Consider a patient admitted with pneumonia complicated by hyponatremia (Na+ = 128 mEq/L) due to syndrome of inappropriate antidiuretic hormone secretion (SIADH) who is treated with intravenous antibiotics, stabilized, and is now being considered for discharge. Despite fluid restriction the serum Na+ concentration is currently 126 mEq/L and the admitting physician is reluctant to discharge the patient for fear the Na+ may fall further and result in symptomatic hyponatremia. The hospital discharge is postponed and fluid restriction is intensified. Two days later the serum Na+ concentration has risen to 131 mEq/L and the patient is discharged. While one can question the wisdom of postponing the original planned discharge based on the lab value alone, this type of scenario is common in clinical practice. Fluid restriction is poorly tolerated, difficult to enforce, and often unpredictable in response. The administration of a single dose of a V2 receptor antagonist is a reliable way to increase the serum Na+ concentration over a 24 hour period and in this case could have potentially shortened the hospital stay.

Consider another patient admitted with a hip fracture who is found to have a serum Na+ concentration of 126 meq/L attributed to ongoing use of an antidepressant agent. The patient is otherwise medically stable but the anesthesiologist is unwilling to accept the patient for surgery until the serum Na+ is at least >130 mEq/L. Fluid restriction is prescribed and only 2 to 3 days later has the serum Na+ reached a value of 131 mEq/L. Once again, administration of a vaptan would have almost certainly increased the Na+ concentration to the desired threshold over a 24 hour period, allowing the patient to undergo the surgical procedure without experiencing the undue delay.

In each of these scenarios short term use of a vaptan would provide for a more rapid and predictable increase in the serum Na+ concentration and potentially decrease the length of hospital stay in comparison to fluid restriction alone. In addition to being less predictable and often poorly tolerated, fluid restriction is often ineffective due to the obligatory fluid administration hospitalized patients require for other therapies or nutrition. Demeclocycline and lithium have been used to antagonize the effects of AVP on the tubule but these drugs take several days before any demonstrable increase in renal water excretion is seen. Demeclocycline antagonizes the effect of AVP through nephrotoxic effects on the tubular cell whereas vaptan's are competitive inhibitors of the V2 receptor and are not associated with nephrotoxicity. Lithium interferes in the intracellular signaling pathways by which AVP causes insertion of water channels into the apical membrane. The doses required to illicit an increase in renal water excretion are near those which can result in lithium levels sufficient to cause neurotoxicity. The primary side effect of vaptan therapy are those one would predict from inducing an aquaretic effect and include thirst, increased urinary frequency, and increased urinary volume.

The decision to use a vaptan on a more prolonged basis is made on a case by case basis. When the underlying cause of increased AVP is deemed to be chronic and irreversible then therapy can be extended into the outpatient setting. Consideration for more chronic therapy would be appropriate for patients with SIADH due to underlying cancer, or those with severe chronic congestive heart failure or advanced cirrhosis. If the cause of the increased AVP is transient, then 1 to 2 doses of a vaptan while in the hospital may be all that is required. Transient causes of increased AVP would include drug related causes, acute pneumonia, hypoxia associated with respiratory failure, and acute decompensated heart failure. Once again vaptan therapy should not be used when the cause of increased AVP is due to total body salt depletion.

AVP Antagonists in Asymptomatic Hyponatremic Patients

One remaining question concerning the use of vaptan therapy is whether they are helpful in hyponatremic patients who are asymptomatic. There are several observations which raise the possibility that patients deemed to be otherwise asymptomatic do in fact have subtle abnormalities attributable to the low Na+ concentration and therefore could benefit if the hyponatremia was corrected. First, some patients with a serum Na+ in the range of 120 mEq/L to 129 mEq/L have subtle neurologic changes that improve when the serum Na+ concentration is increased.14 These include scores on tests of mental and social functioning. In the SALT 1 and 2 trials a general health survey filled out by the patients showed tolvaptan therapy was associated with improvements in the mental health component of the instrument assessing parameters such as vitality, social functioning, emotionally limited accomplishments, calmness, and sadness.6 Response time is delayed and the error rate is increased in response to various stimuli while patients are hyponatremic, suggesting hyponatremia is associated with reversible impairment in attention. It is certainly reasonable to speculate that correction of hyponatremia in an elderly patient with SIADH might improve that individual's quality of life by allowing them to better enjoy any number of activities such as reading or completing a crossword puzzle. The presence of concomitant neuropsychiatric disturbance (delirium and dementia in particular) may also be relevant, since even mild hyponatremia may increase vulnerability to further alterations in mental status in patients who are impaired at baseline.

Second, asymptomatic hyponatremic patients exhibit subtle disturbances in gait that improve following correction of the serum Na+ concentration.14, 15 In this regard, case control studies have shown an association between hyponatremia and risk of falls and fractures particularly in the elderly population.15, 16 Chronic hyponatremia has been shown to cause a reduction in bone mass in an experimental model of SIADH and hyponatremia is associated with osteoporosis in cross‐sectional human data.17 Fall‐related injury is associated with substantial adverse psychological and physical outcomes and is a cause of both death and disability in this population. It would be of considerable socioeconomic benefit if treatment of asymptomatic hyponatremia were able to reduce the risk of this complication.

Lastly, hyponatremia is associated with increased morbidity and mortality but generally the low Na+ concentration is merely thought to be marker of the severity of the underlying disease and not a direct contributor to the adverse outcome.18, 19 For example in decompensated heart failure there is an inverse relationship between the degree of hyponatremia and the extent of neurohormonal activation. Adverse effects resulting from persistent activation of the renin‐angiotensin‐aldosterone system and sympathetic nerve activity are thought to be the mechanism underlying the increase in mortality in hyponatremic patients with heart failure. Nevertheless, it has been suggested that hyponatremia itself, the associated hypotonicity, and/or elevated levels of AVP might exert adverse effects on the cardiovascular system or other organ systems and therefore play a contributory role in patient morbidity and mortality. While it remains speculative as to whether correction of the hyponatremia per se will improve patient outcomes, the V2 receptor antagonists offer an opportunity to test this uncertainty in patients with euvolemic and hypervolemic hyponatremia.0, 0

Location and Function of AVP Receptor Subtypes
Subtype Location Function
  • Abbreviations: ACTH, adrenocorticotropin stimulating hormone; AVP, arginine vasopressin.

V1A Vascular smooth muscle to include splanchnic bed Vasoconstriction
V1B Anterior pituitary Release of ACTH
V2 Basolateral surface of renal collecting duct, vascular endothelium, vascular smooth muscle Insertion of aquaporin channel into luminal membrane, release of von Willebrand factor, vasodilation
AVP Receptor Antagonists Currently Approved by FDA
Parameter Tolvaptan Conivaptan
  • Abbreviations: AVP: arginine vasopressin; CYP3A4, cytochrome P450 3A4; FDA, Food and Drug Administration.

Trade name Samsca Vaprisol
Administration Oral Intravenous
Dose 1560 mg daily 20 mg loading dose followed by 2040 mg continuous infusion
Receptor V2 V1A and V2
Protein binding (%) 99 98.5
Half life 68 hours 38 hours
Metabolism Hepatic (CYP3A4) Hepatic (CYP3A4)
Elimination Feces Feces

Under normal circumstances, there is a balance between water intake and water excretion such that plasma osmolality and the serum sodium (Na+) concentration remain relatively constant. The principal mechanism responsible for prevention of hyponatremia and hyposmolality is renal water excretion. In all hyponatremic patients, water intake exceeds renal water excretion.

Excretion of water by the kidney is dependent on 3 factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be preserved so that the tubular fluid will be diluted. Lastly, arginine vasopressin (AVP) levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. Given the central role of AVP in limiting renal water excretion, AVP receptor antagonists represent a physiologic and rational method to increase renal water excretion.

AVP in Regulation of Plasma Osmolality

AVP is synthesized in the supraoptic and paraventricular nucleus of the hypothalamus and then stored in the neurohypophysis (reviewed in the article Diagnostic Approach and Management of Inpatient Hyponatremia in this supplement). The release of AVP is exquisitely sensitive to changes in plasma osmolality. AVP is not detectable in the plasma at an osmolality below approximately 280 mOsm/kg but increases in a nearly linear fashion beginning with as little as a 2% to 3% increase in osmolality above this value. The extreme sensitivity of this system allows for plasma osmolality to be maintained within a narrow range.

A second major determinant of AVP release is the effective arterial blood volume. While AVP levels are very sensitive to plasma osmolality, small changes of <10% in blood pressure or blood volume have no effect on AVP levels. However, once decreases in volume or pressure exceed this value, baroreceptor‐mediated signals provide persistent stimuli for AVP secretion. Baroreceptor‐mediated AVP release will continue even when plasma osmolality falls below 280 mOsm/kg. Teleologically, this system can be viewed as an emergency mechanism to defend blood pressure. Thus, small decreases in blood volume and blood pressure will cause the body to retain NaCl which will raise osmolality and lead to water retention. However, if NaCl is not available and if blood pressure and volume are becoming dangerously low (down 10%), the body behaves as if defense of blood pressure is more important than defense of osmolality, and AVP is secreted.1 The specific compartment whose volume is sensed in order to determine AVP secretion in this setting is the effective arterial volume. This overriding effect of volume explains the persistence of high AVP levels in hyponatremic patients with conditions such as heart failure and cirrhosis.

Other stimuli for the release of AVP include pain, nausea, and hypoxia. Inappropriate release of AVP can occur with a variety of central nervous system and pulmonary diseases as well as with drugs, particularly those that act within the central nervous system.2 Certain tumors can synthesize and release AVP.

AVP exerts its effects on cells through 3 receptors (Table 1). The V1A receptor is expressed in a variety of tissues but is primarily found on vascular smooth muscle cells. Stimulation of this receptor results in vasoconstriction, platelet aggregation, inotropic stimulation and myocardial protein synthesis. The V1B receptor is expressed in cells of the anterior pituitary and throughout the brain. Stimulation of this receptor results in release of adrenocorticotropin stimulating hormone (ACTH). Stimulation of the V1A and V1B receptors activate phospholipase C leading to increases in inositol trisphosphate and diacylglycerol with secondary increases in cell calcium and activation of protein kinase C.

The V2 receptor is found on the basolateral surface of the renal collecting duct and vascular endothelium where it mediates the antidiuretic effects of AVP and stimulates the release of von Willebrand factor respectively. Unlike the V1A and V1B receptors, binding of AVP to the V2 receptor activates the GS‐coupled adenyl cyclase system causing increased intracellular levels of cyclic adenosine monophosphate (cAMP). In the kidney, generation of cAMP stimulates protein kinase A which then phosphorylates preformed aquaporin‐2 water channels causing trafficking and insertion of the channels into the luminal membrane of the tubular cells.3 The insertion of the aquaporin‐2 protein renders the collecting duct selectively permeable to water, which is then reabsorbed from the tubular lumen into the blood driven by the osmotic driving force of the hypertonic interstitium. In the absence of AVP, aquaporin membrane insertion and apical membrane water permeability are dramatically reduced.

Physiologic Rationale for Use of AVP Antagonists

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of the aquaporin‐2 water channels into the luminal membrane. The increase in urine output is similar in quantity to diuretics but differs in content. V2 receptor antagonists increase water excretion with little to no change in urinary electrolytes. As a result, lowering of the serum K+ level, metabolic alkalosis, and increases in the serum creatinine and blood urea nitrogen concentration are avoided in contrast to diuretics such as furosemide and hydrochlorothiazide. In addition, orthostatic hypotension and activation of neurohumoral effectors such as angiotensin II, circulating catecholamines, and aldosterone are not features of V2 receptor blockade. These differences have lead to V2 receptor antagonists being characterized as aquaretic agents so as to distinguish them from diuretics.

The physiologic rationale for use of V2 receptor antagonists is best exemplified by considering the relationship between the serum Na+ concentration and the total body content of Na+, K+, and water approximated by the equation: 2 receptor antagonist) is approved by the Food and Drug Administration (FDA) for euvolemic and hypervolemic hyponatremia. The effectiveness of the drug was demonstrated in a randomized placebo controlled trial of 84 hospitalized patients with euvolemic or hypervolemic hyponatremia in which the serum Na+ ranged from 115 to <130 mEq/L.5 The patients were given a 20‐mg loading dose followed by a continuous infusion of either 40 or 80 mg daily for 4 days. Conivaptan significantly raised the serum Na+ concentration by 6.3 and 9.4 in the 40 and 80 mg/day arms respectively as compared to 0.8 mEq/L in the placebo group.

Conivaptan is also active as an oral formulation but its distribution has been restricted to parenteral use for short‐term (4 days maximum) in‐hospital administration only. This restriction is due to potent inhibitory effects of the drug on the hepatic cytochrome P450 3A4 enzyme system and the potential for untoward drug interactions. The inhibitory effects of other members of this class on the cytochrome P450 3A4 (CYP3A4) system are more limited, allowing for oral formulations to be used in a clinical setting.

Tolvaptan is the only other AVP antagonist currently approved by the FDA. Unlike conivaptan, tolvaptan is an oral agent with effects confined to the V2 receptor. The drug is indicated for the treatment of clinically significant hypervolemic and euvolemic hyponatremia.

The efficacy of tolvaptan was evaluated in 2 simultaneously conducted multicenter, randomized, double blind trials called Study of Ascending Levels of Tolvaptan in hyponatremia 1 and 2 (SALT 1 and SALT 2).6 The 2 studies together randomized 225 patients with euvolemic or hypervolemic hyponatremia to outpatient treatment with the study drug for 30 days and 223 patients to placebo. Patients with a serum Na+ of <120 mEq/L in association with neurologic impairment were excluded from the trial. Serum Na+ increased more in the tolvaptan group than in the placebo during the first 4 days (3.62 vs. 0.25 mEq/L). After 30 days of therapy serum Na+ concentrations were 6.22 higher in the tolvaptan group compared with 1.66 mEq/L in the placebo group. When tolvaptan was discontinued at study end, the serum Na+ fell over a seven day period to a value similar to that in the placebo treated patients. Four patients (1.8%) exceeded the study goal of limiting correction of the hyponatremia to <12 mEq/L in the first 24 hours of treatment, but none of these patients developed adverse clinical sequelae.

As previously mentioned conivaptan and tolvaptan are indicated for the treatment of hyponatremia in the setting of euvolemia or hypervolemia. These drugs should not be used in hypovolemic states since the increase in renal water excretion can potentially predispose to worsening hemodynamics in the setting of volume depletion. To be sure, any volume of water removed from the body is principally derived from the intracellular compartment (two‐thirds) and would not be expected to affect blood pressure to a major extent. However, one‐twelfth of any water volume loss is derived from the circulating compartment and could potentially aggravate a borderline low blood pressure in the setting of volume depletion.

This concern is particularly true for conivaptan and its blocking effects on the V1a receptor. AVP can cause peripheral vasoconstriction by stimulating the V1a receptor on the peripheral vasculature.7 However, circulating concentrations observed in euvolemic and hypervolemic conditions are not typically of a magnitude to elicit this effect thus explaining the lack of clinically significant hypotension in clinical trials. By contrast, AVP may reach a high enough concentration and play a contributory role in the maintenance of blood pressure under conditions of significant depletion of extracellular fluid volume (hemorrhage) or in states of generalized vasodilation such as sepsis or advanced cirrhosis. In these settings blockade of the V1a receptor may result in significant hypotension.

Another theoretical concern of blocking the V1a receptor with conivaptan is the potential to cause further sequestration of fluid in the splanchnic vascular bed and theoretically increase the risk of hepatorenal syndrome.8 The importance of splanchnic vasodilation in the genesis of renal hypoperfusion has been indirectly illustrated by the response to ornipressin, an analog of AVP that is a preferential splanchnic vasoconstrictor. The administration of ornipressin to patients with advanced cirrhosis leads to correction of many of the systemic and renal hemodynamic abnormalities that are present. These include an elevation in mean arterial pressure, reductions in plasma renin activity and norepinephrine concentration, and increases in renal blood flow, glomerular filtration rate, and urinary sodium excretion and volume. V1a receptor blockade has the potential to increase the degree of arterial vasodilation in the splanchnic arteriolar bed. Increasing degrees of splanchnic vasodilation contribute to a fall in mean arterial pressure and unloading of baroreceptors in the central circulation. As a result, central afferent sensors signal the activation of neurohumoral effectors which in turn decrease perfusion of other organs, particularly the kidney.

AVP Antagonists in Severe Symptomatic Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, restlessness, confusion and disorientation.9 Life threatening symptoms are those of impending brain herniation and include seizures, decreased levels consciousness, and obtundation. Hypertonic saline remains the treatment of choice in those patients who are clinically determined to be severely symptomatic.10 Vaptan therapy does result in a brisk and relatively prolonged water diuresis. Indeed, in the first 6 hours following the parenteral administration of conivaptan there is a several mEq/L increase in the serum Na+ concentration.5 Whether this time course of correction is sufficient to abort fatal hyponatremic encephalopathy is simply not known. Such patients to date have been excluded from clinical studies since randomization in a placebo‐controlled trial would be clearly unethical in such subjects, and not using hypertonic saline might also be considered unsafe

Although not well studied, one could coadminister conivaptan along with hypertonic saline and anticipate a more rapid initial rise in the serum Na+. Once stabilized, the hypertonic saline could be discontinued and the remainder of correction be accomplished by use of the receptor antagonist alone. This strategy would help to minimize the likelihood of volume overload due to the use of hypertonic saline but would require frequent monitoring of the serum Na+ to ensure the rate of correction was within accepted guidelines.

AVP Antagonists in Patients With Mild to Moderate Symptoms of Hyponatremia

Hospitalized patients with mild to moderate symptoms of hyponatremia can be considered ideal candidates for the use of V2 receptor antagonists. Both conivaptan and tolvaptan can be expected to increase the serum Na+ concentration to a greater extent and more predictably than fluid restriction alone. The superiority of these drugs would be particularly evident in those who require ongoing fluid administration for any number of reasons such as parenteral administration of antibiotics or proton pump inhibitors.11

In hyponatremic patients with decompensated heart failure there may be changes in mental status in which it is difficult to separate out the contribution of hyponatremia from the decrease in cerebral perfusion. Vaptan therapy offers a predictable way to improve the serum Na+ contribution over an acceptable period of time thereby removing one of the variables. Even if hyponatremia is not the direct cause of symptoms, it may lower the threshold for mental status changes resulting from poor cerebral perfusion. A similar argument for vaptan therapy can be made in hyponatremic patients with cirrhosis. In this population mental status changes can be due to the hyponatremia, the underlying liver disease, or both. If no improvement occurred following the correction of the hyponatremia, therapy could then be focused on the underlying liver disease. In fact, in any delirious patient with hyponatremia, correction of the underlying electrolyte disorder can help to clarify the degree to which the hyponatremia is exacerbating the altered mental status.

There are no head to head studies comparing conivaptan and tolvaptan in patients with mild to moderate symptoms of hyponatremia. Presumably both drugs would work with similar efficacy. Conivaptan therapy can be complicated by phlebitis when administered through a peripheral vein due to the irritative effects of polypropylene glycol which serves as a diluent for the drug.12 For this reason the drug is often given through a central vein. The theoretical risk of conivaptan's V1a receptor blocking effects in patients with cirrhosis has already been discussed. Tolvaptan is given orally starting at 15 mg daily. Depending on the response the dose can be increased to 30 mg and ultimately to 60 mg daily. Few patients in the SALT 1 and 2 trials required the 60 mg dose.

Measurement of the urine osmolality may be useful in predicting how responsive a patient will be to the administration of a vaptan. In this regard, urine osmolality can be thought as a biomarker of the effect of AVP on the renal collecting duct.13 The higher the urine osmolality, presumably the higher the serum AVP level or the greater the effect of AVP is on the tubule. By contrast, those with a lower urine osmolality presumably have less AVP effect on the tubule and therefore will manifest a less robust response to a V2 receptor blocker.

There are circumstances unique to the hospitalized patient where one may consider use of a V2 receptor antagonist. Consider a patient admitted with pneumonia complicated by hyponatremia (Na+ = 128 mEq/L) due to syndrome of inappropriate antidiuretic hormone secretion (SIADH) who is treated with intravenous antibiotics, stabilized, and is now being considered for discharge. Despite fluid restriction the serum Na+ concentration is currently 126 mEq/L and the admitting physician is reluctant to discharge the patient for fear the Na+ may fall further and result in symptomatic hyponatremia. The hospital discharge is postponed and fluid restriction is intensified. Two days later the serum Na+ concentration has risen to 131 mEq/L and the patient is discharged. While one can question the wisdom of postponing the original planned discharge based on the lab value alone, this type of scenario is common in clinical practice. Fluid restriction is poorly tolerated, difficult to enforce, and often unpredictable in response. The administration of a single dose of a V2 receptor antagonist is a reliable way to increase the serum Na+ concentration over a 24 hour period and in this case could have potentially shortened the hospital stay.

Consider another patient admitted with a hip fracture who is found to have a serum Na+ concentration of 126 meq/L attributed to ongoing use of an antidepressant agent. The patient is otherwise medically stable but the anesthesiologist is unwilling to accept the patient for surgery until the serum Na+ is at least >130 mEq/L. Fluid restriction is prescribed and only 2 to 3 days later has the serum Na+ reached a value of 131 mEq/L. Once again, administration of a vaptan would have almost certainly increased the Na+ concentration to the desired threshold over a 24 hour period, allowing the patient to undergo the surgical procedure without experiencing the undue delay.

In each of these scenarios short term use of a vaptan would provide for a more rapid and predictable increase in the serum Na+ concentration and potentially decrease the length of hospital stay in comparison to fluid restriction alone. In addition to being less predictable and often poorly tolerated, fluid restriction is often ineffective due to the obligatory fluid administration hospitalized patients require for other therapies or nutrition. Demeclocycline and lithium have been used to antagonize the effects of AVP on the tubule but these drugs take several days before any demonstrable increase in renal water excretion is seen. Demeclocycline antagonizes the effect of AVP through nephrotoxic effects on the tubular cell whereas vaptan's are competitive inhibitors of the V2 receptor and are not associated with nephrotoxicity. Lithium interferes in the intracellular signaling pathways by which AVP causes insertion of water channels into the apical membrane. The doses required to illicit an increase in renal water excretion are near those which can result in lithium levels sufficient to cause neurotoxicity. The primary side effect of vaptan therapy are those one would predict from inducing an aquaretic effect and include thirst, increased urinary frequency, and increased urinary volume.

The decision to use a vaptan on a more prolonged basis is made on a case by case basis. When the underlying cause of increased AVP is deemed to be chronic and irreversible then therapy can be extended into the outpatient setting. Consideration for more chronic therapy would be appropriate for patients with SIADH due to underlying cancer, or those with severe chronic congestive heart failure or advanced cirrhosis. If the cause of the increased AVP is transient, then 1 to 2 doses of a vaptan while in the hospital may be all that is required. Transient causes of increased AVP would include drug related causes, acute pneumonia, hypoxia associated with respiratory failure, and acute decompensated heart failure. Once again vaptan therapy should not be used when the cause of increased AVP is due to total body salt depletion.

AVP Antagonists in Asymptomatic Hyponatremic Patients

One remaining question concerning the use of vaptan therapy is whether they are helpful in hyponatremic patients who are asymptomatic. There are several observations which raise the possibility that patients deemed to be otherwise asymptomatic do in fact have subtle abnormalities attributable to the low Na+ concentration and therefore could benefit if the hyponatremia was corrected. First, some patients with a serum Na+ in the range of 120 mEq/L to 129 mEq/L have subtle neurologic changes that improve when the serum Na+ concentration is increased.14 These include scores on tests of mental and social functioning. In the SALT 1 and 2 trials a general health survey filled out by the patients showed tolvaptan therapy was associated with improvements in the mental health component of the instrument assessing parameters such as vitality, social functioning, emotionally limited accomplishments, calmness, and sadness.6 Response time is delayed and the error rate is increased in response to various stimuli while patients are hyponatremic, suggesting hyponatremia is associated with reversible impairment in attention. It is certainly reasonable to speculate that correction of hyponatremia in an elderly patient with SIADH might improve that individual's quality of life by allowing them to better enjoy any number of activities such as reading or completing a crossword puzzle. The presence of concomitant neuropsychiatric disturbance (delirium and dementia in particular) may also be relevant, since even mild hyponatremia may increase vulnerability to further alterations in mental status in patients who are impaired at baseline.

Second, asymptomatic hyponatremic patients exhibit subtle disturbances in gait that improve following correction of the serum Na+ concentration.14, 15 In this regard, case control studies have shown an association between hyponatremia and risk of falls and fractures particularly in the elderly population.15, 16 Chronic hyponatremia has been shown to cause a reduction in bone mass in an experimental model of SIADH and hyponatremia is associated with osteoporosis in cross‐sectional human data.17 Fall‐related injury is associated with substantial adverse psychological and physical outcomes and is a cause of both death and disability in this population. It would be of considerable socioeconomic benefit if treatment of asymptomatic hyponatremia were able to reduce the risk of this complication.

Lastly, hyponatremia is associated with increased morbidity and mortality but generally the low Na+ concentration is merely thought to be marker of the severity of the underlying disease and not a direct contributor to the adverse outcome.18, 19 For example in decompensated heart failure there is an inverse relationship between the degree of hyponatremia and the extent of neurohormonal activation. Adverse effects resulting from persistent activation of the renin‐angiotensin‐aldosterone system and sympathetic nerve activity are thought to be the mechanism underlying the increase in mortality in hyponatremic patients with heart failure. Nevertheless, it has been suggested that hyponatremia itself, the associated hypotonicity, and/or elevated levels of AVP might exert adverse effects on the cardiovascular system or other organ systems and therefore play a contributory role in patient morbidity and mortality. While it remains speculative as to whether correction of the hyponatremia per se will improve patient outcomes, the V2 receptor antagonists offer an opportunity to test this uncertainty in patients with euvolemic and hypervolemic hyponatremia.0, 0

Location and Function of AVP Receptor Subtypes
Subtype Location Function
  • Abbreviations: ACTH, adrenocorticotropin stimulating hormone; AVP, arginine vasopressin.

V1A Vascular smooth muscle to include splanchnic bed Vasoconstriction
V1B Anterior pituitary Release of ACTH
V2 Basolateral surface of renal collecting duct, vascular endothelium, vascular smooth muscle Insertion of aquaporin channel into luminal membrane, release of von Willebrand factor, vasodilation
AVP Receptor Antagonists Currently Approved by FDA
Parameter Tolvaptan Conivaptan
  • Abbreviations: AVP: arginine vasopressin; CYP3A4, cytochrome P450 3A4; FDA, Food and Drug Administration.

Trade name Samsca Vaprisol
Administration Oral Intravenous
Dose 1560 mg daily 20 mg loading dose followed by 2040 mg continuous infusion
Receptor V2 V1A and V2
Protein binding (%) 99 98.5
Half life 68 hours 38 hours
Metabolism Hepatic (CYP3A4) Hepatic (CYP3A4)
Elimination Feces Feces
References
  1. Palmer BF,Alpern RJ.Integrated Response to Abnormalities in Tonicity. In: Seldin DW, Giebisch G, eds.Clinical Disturbances of Water Metabolism.New York, New York:Raven Press Ltd;1993:273295.
  2. Palmer BF,Gates JR,Lader M.Causes and management of hyponatremia.Ann Pharmacother.2003;37:16941702.
  3. Nedvetsky P,Tamma G,Beulshausen S,Valenti G,Rosenthal W,Klussmann E.Regulation of aquaporin‐2 trafficking.Handb Exp Pharmacol.2009;190:133157.
  4. Chen S,Jalandhara N,Batlle D.Evaluation and management of hyponatremia: an emerging role for vasopressin receptor antagonists.Nat Clin Pract Nephrol.2007;3:8295.
  5. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
  6. Schrier R,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:2099112.
  7. Koshimizu T,Nasa Y,Tanoue A, et al.V1a vasopressin receptors maintain normal blood pressure by regulating circulating blood volume and baroreflex sensitivity.Proc Natl Acad Sci.2006;103:78077812.
  8. Palmer B.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  9. Adrogue H,Madias N.Hyponatremia.N Engl J Med.2000;342:15811589.
  10. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  11. Palmer BF.Hyponatremia in the intensive care unit.Semin Nephrol.2009;29:257270.
  12. Ghali J,Farah J,Daifallah S,Zabalawi H,Zmily H.Conivaptan and its role in the treatment of hyponatremia.Drug Des Devel Ther.2009;3:253268.
  13. Verbalis J.Vaptans for the treatment of hyponatremia: how who when and why.Nephrol Self Assess Program.2007;6:199209.
  14. Decaux G.The syndrome of inappropriate secretion of antidiuretic hormone (SIADH).Semin Nephrol.2009;29(3):239256.
  15. Kengne F,Andres C,Sattar L,Melot C,Decaux C.Mild hyponatremia and risk of fracture in the ambulatory elderly.Q J Med.2008;101:583588.
  16. Kinsella S,Moran S,Sullivan M,Molloy M,Eustace J.Hyponatremia independent of osteoporosis is associated with fracture occurrence.Clin J Am Soc Nephrol.2010 (in press).
  17. Verbalis J,Barsony J,Sugimura Y, et al.Hyponatremia‐induced osteoporosis.J Bone Miner Res.2009;999:137.
  18. Wald R,Jaber B,Price L,Upadhyay A,Madias, N.Impact of hospital‐associated hyponatremia on selected outcomes.Arch Intern Med.2010;170(3):294302.
  19. Waikar S,Mount D,Curhan G, et al.Mortality after hospitalization with mild, moderate, and severe hyponatremia.Am J Med.2009;122:857865.
References
  1. Palmer BF,Alpern RJ.Integrated Response to Abnormalities in Tonicity. In: Seldin DW, Giebisch G, eds.Clinical Disturbances of Water Metabolism.New York, New York:Raven Press Ltd;1993:273295.
  2. Palmer BF,Gates JR,Lader M.Causes and management of hyponatremia.Ann Pharmacother.2003;37:16941702.
  3. Nedvetsky P,Tamma G,Beulshausen S,Valenti G,Rosenthal W,Klussmann E.Regulation of aquaporin‐2 trafficking.Handb Exp Pharmacol.2009;190:133157.
  4. Chen S,Jalandhara N,Batlle D.Evaluation and management of hyponatremia: an emerging role for vasopressin receptor antagonists.Nat Clin Pract Nephrol.2007;3:8295.
  5. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
  6. Schrier R,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:2099112.
  7. Koshimizu T,Nasa Y,Tanoue A, et al.V1a vasopressin receptors maintain normal blood pressure by regulating circulating blood volume and baroreflex sensitivity.Proc Natl Acad Sci.2006;103:78077812.
  8. Palmer B.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  9. Adrogue H,Madias N.Hyponatremia.N Engl J Med.2000;342:15811589.
  10. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  11. Palmer BF.Hyponatremia in the intensive care unit.Semin Nephrol.2009;29:257270.
  12. Ghali J,Farah J,Daifallah S,Zabalawi H,Zmily H.Conivaptan and its role in the treatment of hyponatremia.Drug Des Devel Ther.2009;3:253268.
  13. Verbalis J.Vaptans for the treatment of hyponatremia: how who when and why.Nephrol Self Assess Program.2007;6:199209.
  14. Decaux G.The syndrome of inappropriate secretion of antidiuretic hormone (SIADH).Semin Nephrol.2009;29(3):239256.
  15. Kengne F,Andres C,Sattar L,Melot C,Decaux C.Mild hyponatremia and risk of fracture in the ambulatory elderly.Q J Med.2008;101:583588.
  16. Kinsella S,Moran S,Sullivan M,Molloy M,Eustace J.Hyponatremia independent of osteoporosis is associated with fracture occurrence.Clin J Am Soc Nephrol.2010 (in press).
  17. Verbalis J,Barsony J,Sugimura Y, et al.Hyponatremia‐induced osteoporosis.J Bone Miner Res.2009;999:137.
  18. Wald R,Jaber B,Price L,Upadhyay A,Madias, N.Impact of hospital‐associated hyponatremia on selected outcomes.Arch Intern Med.2010;170(3):294302.
  19. Waikar S,Mount D,Curhan G, et al.Mortality after hospitalization with mild, moderate, and severe hyponatremia.Am J Med.2009;122:857865.
Issue
Journal of Hospital Medicine - 5(3)
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S27-S32
Page Number
S27-S32
Article Type
Display Headline
New horizons in the pharmacologic approach to hyponatremia: The V2 receptor antagonists
Display Headline
New horizons in the pharmacologic approach to hyponatremia: The V2 receptor antagonists
Legacy Keywords
arginine vasopressin, AVP receptor antagonists, conivaptan, tolvaptan, hyponatremia
Legacy Keywords
arginine vasopressin, AVP receptor antagonists, conivaptan, tolvaptan, hyponatremia
Sections
Article Source
Copyright © 2010 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

Managing Hyponatremia in Cirrhosis

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Managing hyponatremia in cirrhosis

The serum sodium (Na) level is the major determinant of serum osmolality. In normal physiologic states is tightly regulated between 135 mEq/L to 145 mEq/L despite variable intake of water and solute through the interaction of osmoreceptors in the hypothalamus where arginine vasopressin (AVP) is synthesized and then released by the posterior pituitary and the binding of AVP with V2 AVP receptors on the basolateral surface of the principal cells within the collecting duct of the kidney. Binding of AVP to the V2 receptors promotes the translocation and fusion of cytoplasmic vesicles which carry the water channel protein aquaporin 2 (AQP2) to the apical membrane of the cell and, in this manner, increases water permeability and absorption.1, 2, 3

Patients with hyponatremia, defined by a serum Na level <135 mEq/L, can be broadly classified by their volume status into those who are euvolemic, hypervolemic, and hypovolemic (Table 1). In patients with euvolemic hyponatremia such as those with Syndrome of Inappropriate Antidiuretic Hormone (SIADH), total body Na is nearly normal, but total body water is increased. In patients with hypervolemic hyponatremia, both total body Na and water are increased, but water to a much greater degree. These patients typically have increased extracellular fluid such as edema and/or ascites. The most common conditions associated with this condition are cirrhosis, congestive heart failure (CHF), and renal failure. In contrast, hypovolemic hyponatremia is associated with a reduction in both total body Na and water, but Na to a greater degree. This condition is encountered in patients with excessive fluid losses such as those with over‐diuresis, excessive gastrointestinal losses, burns, and pancreatitis.4

Classification of Hyponatremia: Sodium and Water Changes in the 3 Different Types of Hyponatremia
Depletional Hyponatreima Dilutional Hyponatremia
Euvolumic Hypervolumic
Total body water
Total body Na normal
Common etiologies SIADH cirrhosis/CHF vomiting, diarrhea

Hyponatremia is the most common electrolyte abnormality seen in general hospital patients.5 In a database of over 120,000 patients, a serum sodium level of <136mEq/L was observed in 28.2%.6 Hyponatremia is associated with selected medical conditions (especially cirrhosis and CHF), the extremes of age, and those receiving selected medications, including several that are commonly administered to cirrhotic patients (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors).7, 8 Hyponatremia is associated with increased total costs per hospital admission.5, 9 In an analysis of the effect of hyponatremia on length of stay in a retrospective cohort study of hospitalized patients derived from a large administrative database of 198,281 discharges from 39 US hospitals, mean length of stay was significantly greater among patients with hyponatremia than those with normal Na levels (8.6 8.0 vs. 7.2 8.2 days). After adjusting for confounders that may be associated with more severe disease and hyponatremia (age, gender, race, geographic region, teaching status of the hospital, admission source, principal payer, comorbidity index score and primary diagnosis), the presence of hyponatremia contributed an increase in length of stay of 1.0 day. Patients with hyponatremia are more frequently admitted to the intensive care unit (ICU) and require mechanical ventilation. In patients with CHF, the presence of hyponatremia at discharge is associated with increased risk for early mortality and rehospitalization.10

Although frequently asymptomatic, hyponatremia may be associated with a range of findings, from subtle and non‐specific complaints, including headache, fatigue, confusion, malaise, to severe and life‐threatening manifestations with lethargy, seizures, brainstem herniation, respiratory arrest and death.11 The most important complications are neurologic consequences related to cerebral edema. However, there is increased morbidity even in hyponatremic patients considered to be asymptomatic. Patients with low serum sodium have attention deficients, and falls are common. In a study of 122 patients who were considered to have chronic asymptomatic hyponatremia, the incidence of falls was significantly higher at 21.3% compared to only 5.3% in a control population.12

In hyponatremia, water enters into the cells to attain osmotic balance, resulting in cellular swelling.4 To avoid cerebral edema, the brain is capable of adapting to hyponatremia by regulating its volume to avoid swelling, especially when hyponatremia is chronic. In acute hyponatremia, astrocytes and neurons adapt through osmoregulatory mechanisms by extruding intracellular electrolytes such as potassium.13 Chronically, adaption occurs through the loss of low‐molecular weight organic compounds termed organic osmolytes including myoinsoitol, glutamine, choline and taurine. As a result, both the severity and the rate of its development are critical factors in determining the neurologic manifestation of hyponatremia in a given patient.14

Dilutional Hyponatremia and Cirrhosis

Patients with hyponatremia who are either euvolemic or hypervolemic are considered to have dilutional hyponatremia (DH). Management of these patients is distinct from those who are hypovolemic in whom appropriate therapy consists of the administration of normal saline. The remainder of this article addresses the pathogenesis, management and treatment of cirrhotic patients with DH.

Pathogenesis

The development of hyponatremia in cirrhosis is intimately related to the pathophysiology of portal hypertension and the non‐osmotic release of AVP3, 15 (Figure 1). In the early phases of cirrhosis, portal hypertension is the result of an increase in intrahepatic resistance. With the development of porto‐systemic collaterals, a hyperdynamic splanchnic circulation develops as a result of splanchnic arterial vasodilatation and increased vascular capacity. Nitric oxide, an endothelial derived relaxing factor, is the critical mediator of this process, and upregulation of its expression is pivotal in the pathogenesis of portal hypertension.

Figure 1
Proposed mechanism of hypersecretion and renal and systemic effects of vasopressin in cirrhosis with ascites. Gines P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management. Hepatology. 2008;48:1002–1010. Copyright 2008 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.

Multiple factors are related to the development of DH in cirrhosis. A reduction of effective central blood volume due to the development of porto‐venous collaterals and arterial splanchnic vasodilation, leading to baroreceptor‐mediated nonosmotic release of AVP, is considered the initiating and most important factor. Patients with cirrhosis and DH have higher plasma and urine vasopressin levels, higher plasma renin activity, and decreased plasma levels of atrial natriuretic factor than those with normal serum sodium concentrations, findings consistent with the presence of a decreased effective plasma volume.16 Arterial underfilling is sensed by baroreceptors located in the left ventricle, aortic arch, carotid sinus and renal afferent arterioles. Decreased activation leads to neurohumoral compensatory responses which include non‐osmotic release of vasopressin from the neurohypophysis and increased levels. Impaired catabolism of AVP that has been correlated with the severity of liver dysfunction may further contribute to increased levels.17 Initially, the increased AVP maintains arterial circulatory integrity by inducing splanchnic, peripheral and renal arterial vasoconstriction through its action on the V1a receptors and expansion of blood volume through renal water retention by its action on the V2 receptors located on the collecting ducts.

The initial adaptive response which leads to increased central blood volume can chronically result in detrimental effects, including the development of fluid overload with ascites, edema, and hyponatremia.16, 18 Additional factors that contribute to hyponatremia include decreased glomerular filtration rate (GFR) and/or increased proximal reabsorption of sodium (that reduce the distal delivery of filtrate and the potential for water reabsorption) and decreased cardiac function that further impairs effective central blood volume.19 In addition, urinary levels of AQP2 are increased in cirrhotic patients, especially those with decompensated disease with higher Child‐Pugh scores and ascites, and provide another potential mechanism to increase water reabsorption.20

Prevalence and Prognostic Significance

Hyponatremia in cirrhosis is a common finding. In a survey of 997 cirrhotic patients with ascites from 28 centers in Europe, North and South America, the prevalence of serum sodium concentration 135, 130, 125, 120 meq/L were 49.4%, 21.6%, 5.7%, and 1.2%, respectively.21 In a retrospective analysis of 188 inpatients, the prevalence of DH of 135, 130, and 125 were 20.8%, 14.9%, and 12.2%, respectively.22 The development of hyponatremia is a manifestation of increasing portal hypertension. In a natural history study of 263 patients hospitalized for first episode of significant ascites, 74 patients developed DH (Na level < 130 mEq/L), including 11 patients in whom it appeared during the first episode and 63 cases during follow‐up (mean period of 40 3 months) with a 5‐year incidence of 37.1%.23

The presence of hyponatremia carries significant adverse prognostic significance. It is strongly associated with severity of liver function impairment as assessed by Child‐Pugh and model for end‐stage liver disease (MELD) scores.22 Even mild hyponatremia is associated with severe complications such as massive ascites, severe hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), and hepatic hydrothorax, and the severity of hyponatremia is directly related to the severity of these complications.21, 22 (Figure 2). In a natural history study of patients presenting with large volume ascites, 1‐year survival after its development was reduced to only 25.6%.230

Figure 2
Percentage of patients with complications of cirrhosis classified according to serum sodium concentration. Angeli P, Wong F, Watson H, et al. Hyponatremia in cirrhosis: results of a patient population survey. Hepatology. 2006;44:1535–1542. Copyright 2006 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.
Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Hyponatremia is an especially poor prognostic sign for a hospitalized cirrhotic patient. In a retrospective analysis of 156 cirrhotic patients, hyponatremiapresent in 57 (29.8%) of admissionswas associated with increased hospital mortality (26.3% vs. 8.9% among those with normal Na levels), and the mortality rate was even higher (48%) among the 25 patients who developed severe hyponatremia during the hospital stay.24 In hospitalized patients, hyponatremia is predictive of the development of acute renal failure which is associated with substantially increased mortality (73% vs. 13%).25 Similarly, a low serum sodium level in critically ill cirrhotic patients admitted to the ICU is associated with complications, in‐hospital mortality, and poor short‐term prognosis.26

Whether hyponatremia should impact liver transplant prioritization remains an area of controversy. The United Network for Organ Sharing (UNOS) contracted by the Organ Procurement and Transplant Network (OPTN) to optimize the efficient use of deceased organs through fair and timely allocation, currently uses the MELD score, a formula that calculates the risk of death within three months from the bilirubin, creatinine, and International Normalized Ratio (INR) levels. Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction in patients with advanced cirrhosis and adds to MELD in predicting waitlist mortality.2729 In patients with a MELD score of <21, only low serum sodium and persistent ascites are independent predictors of mortality.28 To account for the importance of hyponatremia on survival, both modification of the MELD score in which the Na level is incorporated (MELD‐Na model) and the MELD to serum sodium ratio (MESO) have been developed. Adding hyponatremia to the MELD score is a better predictor of death than MELD alone, particularly in patients with low MELD scores.27, 2931 The OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has discussed updating the liver allocation system to include the Na level. However, it was concluded that implementation of MELD‐Na would change the allocation status of only 4% of candidates. Further, based on the concerns about the ability to manipulate serum sodium levels and the utility of employing resources to change the system for a relatively small number of patients, it was decided to defer incorporating the Na level pending further analysis (Report of the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee To the Board of Directors, Los Angeles, California, September 17‐18, 2007). At this time, the use of Na is a regional decision.32 However, the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has recently solicited feedback from the transplant community about including Na in allocation for review at a forum in April 2010.

Precipitating Factors

The most important factor related to development of hyponatremia in cirrhosis is increasing severity of portal hypertension that is associated with impaired central blood volume as a result of progressive splanchnic vasodilatation. In a study in which 170 patients with decompensated alcoholic cirrhosis were prospectively followed for 33.9 27.9 months, the initial hepatic venous pressure gradient (HVPG) was an independent predictive factor for the 20 patients who developed hyponatremia.22

Cirrhotic patients with ascites with hyponatremia have increased AVP secretion, higher levels of plasma renin activity, and higher serum concentrations of aldosterone and norepinephrine compared to those with normal Na levels.33 Diuretic therapy is associated with the development of DH by inducing volume depletion and arterial underfilling, further activating the renin‐angiotensin system (RAS) and increasing the non‐osmotic release of AVP.34 Although diuretics block the salt retention associated with the RAS activation, the water‐retaining effects of AVP persist, and DH develops. The process is further exacerbated by a low sodium intake and a frequent uncontrollable thirst. As a result, diuretic therapy is commonly associated with the development of hyponatremia in patients with ascites.24, 35 Similarly, paracentesis (particularly when performed without albumin) is often associated with an increase in blood urea nitrogen and marked elevations in plasma renin activity and plasma aldosterone concentration, which may exacerbate this physiology, leading to further reduction in serum sodium concentration.36 Tense ascites can contribute to DH by increasing baroreceptor mediated AVP release by increasing intrathoracic pressure.37 Finally, non‐steroidal anti‐inflammatory drugs (NSAIDs) can cause DH by inhibiting the synthesis of renal prostaglandins (which normally function to antagonize the tubular action of AVP and are important in the maintenance of appropriate renal tubular transport of fluid and electrolytes in states of renal hypoperfusion).38

Medical Impact of Hyponatremia: Marker of Severe Disease or Direct Pathophysiologic Role?

Hyponatremia is associated with severe ascites, impaired renal function, hepatic encephalopathy, SBP, and hepatorenal syndrome.3, 20 Because hyponatremia is frequently present in advanced liver failure, it is unclear whether it is only a marker of advanced disease or whether it plays a direct pathophysiologic role, or both. Until recently, it has not been possible to address this issue due to the inability to easily and rapidly correct the hyponatremia. However, there is increasing evidence that hyponatremia has direct impact on the severity of hepatic encephalopathy (see Hepatic Encephalopathy section). The recent introduction of tolvaptan for the treatment of hyponatremia in cirrhosis (discussed below) will allow this question to be directly answered.

Fluid Management and Diuresis

The typical cirrhotic patient with DH is characterized by expanded extracellular fluid with ascites and edema. The profound vasodilation of the splanchnic arterial circulation is associated with decreased effective arterial blood volume, leading to the non‐osmotic release of AVP. Diuretic therapy can further exacerbate this process. In addition, the increased water permeability induced by AVP results in reduced urine volume and fluid retention. As a result, hyponatremia directly adversely affects severity of fluid overload and limits and/or precludes diuretic treatment.

Hepatorenal Syndrome

Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction and is frequently a precursor to overt hepatorenal syndrome.27 Hyponatremia is predictive of the development of acute renal failure during hospitalization, and in‐hospital development of acute renal failure portends a high mortality.25 In patients admitted with SBP, the presence of hyponatremia is significantly associated with higher mortality and renal failure.39

Hepatic Encephalopathy

The neurologic manifestations of cerebral edema associated with hyponatremia closely mirror those of hepatic encephalopathy. In fact, a recently proposed pathogenic mechanism for hepatic encephalopathy is the development of low‐grade cerebral edema associated with astrocyte swelling in response to ammonia and other precipitating factors.40 DH is associated with a further reduction in brain organic osmolytes that probably reflects a compensatory osmoregulatory mechanism against cell swelling triggered by a combination of high intracellular glutamine and low extracellular osmolality.41 As a result, it has been proposed that hyponatremia contributes to the development of hepatic encephalopathy through the development or exacerbation of low‐grade cerebral edema. In this manner, low serum sodium acts as a second hit to the swelling produced by increased intracellular glutamine created by ammonia metabolism.42

Clinically, hyponatremia is a major risk factor for hepatic encephalopathy. Serum sodium and ammonia levels are the major factors that predict electroencephalographic abnormalities in cirrhotics who do not have hepatic encephalopathy.43 In a prospective study of 61 patients, hyponatremia was associated with a low brain concentration of organic osmolytes as assessed by proton magnetic resonance spectroscopy (1H‐MRS) and magnetic resonance imaging, and both conditions were major risk factors for the development of overt hepatic encephalopathy.44 Finally, hyponatremia is a risk factor for hepatic encephalopathy in patients undergoing TIPS.45

Adverse Effect on Outcome After Liver Transplantation

Hyponatremia before liver transplantation is associated with adverse post‐transplant outcomes. Among patients undergoing liver transplantation, the presence of hyponatremia is associated with abnormal cardiac response in patients after reperfusion.46 Pre‐transplant hyponatremia is associated with longer ICU and hospital stay, higher rates of delirium and neurologic disorders, acute renal failure, acute cellular rejection, infection, and in one study a reduced 3‐month survival compared to normonatremic recipients.32, 47, 48 In 1 retrospective study that compared post‐transplant outcomes of patients with corrected vs. uncorrected pre‐transplant hyponatremia, patients with pre‐operative correction of hyponatremia had a lower risk of prolonged post‐transplant hospitalization than those with uncorrected hyponatremia.32 However, both hyponatremic groups had more complicated post‐transplant courses compared to those without a history of hyponatremia. However, given the small sample size, retrospective design, and the potential for confounding, the impact of correction of pre‐transplant hyponatremia remains to be determined.

Management

Most patients with mild hypervolemic hyponatremia are asymptomatic. The initial recommended approach is fluid restriction and an Na‐restricted diet. For those with severe or progressive hyponatremia, diuretics should be minimized or discontinued to avoid intravascular volume depletion.49 For patients with tense ascites and severe DH, therapeutic paracentesis with plasma expanders is safe.33 Unfortunately, fluid restriction is limited in efficacy and often poorly tolerated. The use of hypertonic saline is generally not recommended unless severe neurologic symptoms are present as it leads to increased ascites and edema. When administered, it is important to avoid a rapid correction of the hyponatremia to prevent the development of central pontine myelinolysis and the osmotic demyelination syndrome.

Due to the pivotal role of AVP in the pathogenesis of DH, antagonism of its action has long been proposed to be the most rational approach, but until recently, effective and specific antagonism of AVP has remained elusive. Approaches that have been attempted include interference with its secretion and actions. Intravenous albumin has been reported to improve hyponatremia in patients with cirrhosis, ascites, and hyponatremia, presumably by decreasing AVP release by plasma volume expansion.50 An attempt at inhibition of central AVP release with the use of a kappa‐opioid receptor agonist, niravoline, was limited by loss of efficacy and potential adverse effects.51 Use of demeclocycline and lithium (which induce renal resistance to AVP and lead to a modest increase in urine volume with decreased urine osmolality and a corresponding rise in serum sodium) is limited by nephrotoxicity and hepatotoxicity.7, 52 Because of the important role played by prostaglandins in the maintenance of renal hemodynamics and water excretion in cirrhosis, oral misoprostol has also been evaluated but determined to be ineffective in inducing significant changes in free water clearance in patients with functional renal failure and/or DH.53

The recent introduction of vaptans, vasopressin receptor antagonists that block the physiologic action of vasopressin, represents a revolutionary and highly effective approach to the treatment of hyponatremia. Vaptans are antagonists of the V2 receptors of AVP in the principal cells of the collecting ducts. In healthy subjects, vaptans cause a dose‐dependent increase in urine volume and produce a dilute urine without causing natriuresis. To date, 2 AVP antagonists, conivaptan and tolvaptan, have been Food and Drug Administration (FDA)‐approved for the treatment of DH. Conivaptan, the first to be approved in 2005, is a mixed vasopressin V1a and V2 receptor antagonist that is administered intravenously for up to 4 days. In a randomized placebo‐controlled study of patients with euvolemic or hypervolemic hyponatremia, intravenous conivaptan treatment increased serum Na levels by >6 mEq/l or to a serum Na >135 mEq/l in 69 to 88.5% of subjects compared to 20.7% of those receiving placebo (Zeltser D, Rosansky S, Van Rensburg H, et al. Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia. American J Nephrology 2007;27:447457). In a pilot study involving 24 patients with end‐stage liver disease, an infusion of conivaptan over 1 to 4 days was associated with an increase of serum sodium by >5 mmol/L in 60% of patients not receiving diuretics and 67% of patients on concomitant diuretic therapy by the end of treatment (O'Leary and Davis, 2009). Despite a concern about the potential for conivaptan to increase portal hypertension due to inhibition of splanchnic V1a receptors, the brief treatment appeared to be well tolerated without significant changes in systolic blood pressure, serum creatinine, variceal bleeding or worsening of ascites during the infusion period. However, approval for only 4 days of therapy and requirement for intravenous use eliminate any potential for chronic use.54

Tolvaptan is an orally available, selective V2 receptor antagonist whose efficacy was assessed in two multicenter, prospective, randomized, placebo‐controlled trials, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2).55 In these trials, clinically stable patients with DH (Na <135meq/l) associated with cirrhosis (22.4% in SALT‐1, 30.5% in SALT‐2), CHF or SIADH were randomized in a hospital setting to receive tolvaptan 15mg daily or placebo. Repeat Na levels were obtained at 8 hours, 2, 3, and 4 days and then weekly at days 11, 18, 25 and 30 after which study drug was discontinued and follow‐up Na level was determined 7 days later. The dose was adjusted to 30 mg and then 60 mg in an attempt to achieve a Na level >135 in those in whom hyponatremia persisted. During the initial day of the titration phase, fluid restriction was not maintained, and the patients were encouraged to respond to thirst with increased water ingestion.

Tolvaptan use was associated with a prompt increase in Na level as early as 8 hours after administration of the first dose. Serum Na increased more among those receiving tolvaptan than among those receiving placebo during the first 4 days and throughout the study period regardless of baseline Na level but returned to baseline within 1 week after discontinuation (Figure 3). The main side effects were increased thirst, dry mouth and increased urination. Importantly, an increased incidence of renal failure was not observed. Based on these results, FDA approval for tolvaptan in patients with hyponatremia was obtained in May 2009 for patients with DH‐associated with cirrhosis, CHF or SIADH for patients with Na levels <125 or symptomatic patients with Na levels between 125 and 135 that have not responded to fluid restriction.

Management of the Hospitalized Cirrhotic Patient With Hyponatremia: Recommendations

Hyponatremia in hospitalized cirrhotic patients is a marker for severe disease and high risk of hospital mortality.24 As a result, prompt evaluation and treatment is imperative. The availability of tolvaptan potentially revolutionizes the manner in which these patients are treated. In the SALT trials, only clinically stable patients were enrolled. In this last section, a guideline for the evaluation and treatment of acutely ill, hospitalized cirrhotic patients with DH is presented.

Evaluation

Determination of volume status is paramount but frequently problematic in the hospitalized cirrhotic patient. Due to the vasodilated state present in severe portal hypertension that is characterized by a relative hypotension and resting tachycardia, the usual hemodynamic parameters of blood pressure and heart rate can be difficult to interpret. Although significant extravascular volume in the form of ascites and edema may be present, patients may be intravascularly depleted due to previous diuretic use and extra‐renal losses due to impaired oral intake, vomiting, lactulose‐induced diarrhea, and gastrointestinal bleeding. Infection is a commonly associated condition, and endotoxin mediated splanchnic vasodilatation, especially in the setting of SBP, can adversely effective central blood volume status in the presence of severe ascites. Also, due to the Na avidity of the kidney and previous diuretic use, renal electrolytes can be difficult to interpret.

For patients in whom there is strong clinical concern about intravascular depletion (history of impaired oral intake, excessive vomiting and/or diarrhea, rapid weight loss, small volume ascites with history of large volume, azotemia), administration of limited intravenous normal saline (0.5‐1 L) should be considered. Patients with severe neurological symptoms should receive normal saline or hypertonic saline. Unless severe neurologic symptoms associated with profound hyponatremia is present, however, intravenous normal saline should not be administered for the hyponatremia alone. Administration of salt poor albumin (25%), especially for those with marked fluid overload and ascites, is an effective means to expand the central blood volume without exacerbating ascites and edema. After evaluation for and/or treatment of hypovolemia, all patients should receive a Na restricted diet (<2000 mg daily) and placed on fluid restriction (see below for liberalization of fluid restriction upon initiation of tolvaptan therapy).

Diagnostic paracentesis should be performed for those with ascites to rule out the presence of SBP, and antibiotics administered to those with evidence of infection. High dose intravenous salt poor albumin should also be administered, especially to those at high risk of renal failure as determined by the presence of azotemia (Cr > 1.0 mg/dL) or severe liver insufficiency (TBili > 4.0 mg/dL).56, 57 Finally, all medications should be reviewed, and those associated with hyponatremia (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors) discontinued if possible.

Tolvaptan for DH

Patient Selection

Appropriate patient selection for tolvaptan therapy is extremely important (Table 2). In the SALT trials, only clinically stable patients were enrolled. The presence of hyponatremia in a recently hospitalized cirrhotic patient, however, frequently indicates severe disease with a high risk of acute renal failure and hospital death. In the SALT trials, many received concomitant diuretic therapy. Because of the importance of avoiding tolvaptan administration to hypovolemic patients, discontinuation of diuretic therapy prior the initiation of tolvaptan therapy and/or reevaluation after limited volume expansion should be considered.

Patient Selection for Tolvaptan Therapy for Hospitalized Patients With Cirrhosis and Hyponatremia
Hospital setting
Euvolumia or hypervolumia
Absence of recent weight loss, decrease in ascites, edema
Absence of excessive vomiting, diarrhea
Consider discontinuation of diuretic therapy prior to initiation of tolvaptan
Consider evaluation after limited volume expansion, especially with salt poor albumin prior to initiation of tolvaptan
Presence of clinically significant hyponatremia: 125mEq/L or less severe but symptomatic hyponatremia (125 to 134 mEq/L) that has resisted fluid restriction
Absence of severe neurologic symptoms attributable to hyponatremia
No co‐administration with intravenous saline
Ability to respond to thirst
No co‐administration with strong CYP 3A inhibitors (ketoconazole)
Absence of kidney failure with anuria

Tolvaptan is indicated for cirrhotic patients with DH in whom the serum sodium is <125 mEq/L and in those with less severe but symptomatic hyponatremia (125‐134 mEq/L) that has resisted fluid restriction. Although the definition of symptomatic was not specifically defined, possible considerations include symptoms of mild hepatic encephalopathy or inability to tolerate dieresis due to the presence of hyponatremia. According to FDA guidelines, tolvaptan therapy must be initiated and re‐initiated in a hospital setting. Patients with severe neurologic symptoms attributable to hyponatremia in whom rapid treatment is critical should not receive tolvaptan but should rather be treated with normal saline. Similarly, patients should not receive combination therapy with tolvaptan and normal saline due to potential for a too‐rapid correction of hyponatremia and the development of central pontine myelinolysis. If saline had been administered for treatment of possible hypovolemia, it should be discontinued and persistent hyponatremia confirmed before starting tolvaptan. Other factors that need to be considered before initiating tolvaptan include the ability of the patient to respond to thirst with increased water ingestion and recognition that the patient will experience increased urine volume and frequency, requiring easy access to toilet. Patients should not be fluid restricted during the first day of tolvaptan therapy, but should be instructed to respond to their thirst with increased water ingestion. As a result, caution should be exercised in administering tolvaptan to a confused, restrained, unresponsive and/or bed‐bound patient who is not able to respond appropriately to thirst or increased urination.

In the SALT trials, the incidence of hyperkalemia (5%) was similar in the tolvaptan and placebo treated patients.55 However, further analysis of all multiple‐dose, placebo‐controlled trials, demonstrated that the aggregate incidence of hyperkalemia was slightly higher for tolvaptan‐treated subjects compared with placebo‐treated subjects (Otsuka). Because treatment with tolvaptan is associated with an acute reduction of the extracellular fluid volume which could result in increased serum potassium through hemoconcentration, it is recommended that serum potassium levels be monitored after initiation of tolvaptan treatment in patients with a serum potassium > 5 mEq/L as well as those who are receiving drugs known to increase serum potassium levels such as angiotensin converting enzyme inhibitors, angiotensin receptor blockers, or potassium sparing diuretics (Samsca Package Insert, Otsuka). Because tolvaptan is metabolized by the cytochrome P 3A system, patients receiving strong inhibitors such as ketoconazole should not receive tolvaptan. Anuric patients will not respond to tolvaptan. Finally, it is extremely important to administer tolvaptan only to patients with true hyponatremia and not to those with pseudohyponatremia in whom the plasma osmolality is normal but the measured serum sodium concentration artificially low due to marked elevations of other substances, such as can be seen in severe hyperglycemia, marked hyperlipidemia, or hyperproteinemia (as in multiple myeloma).

Tolvaptan Administration

The initial dose of tolvaptan is 15 mg daily. After receiving tolvaptan, many patients will develop an increased sense of thirst and need to urinate. As a result, patients should not be fluid restricted during the first day of therapy, and it is important to monitor the hemodynamics and Na level closely after initiating therapy with a repeat Na level at approximately 8 hours after the first dose. As a result, it should probably be administered early in the day and not at bedtime. The dose should be increased to 30 mg, then 60 mg in patients who do not respond by at least 5 mEq/L over the previous 24 hours and remain hyponatremic. In those with an excessive response (more than 8 meq/L during the first 8 hours or 12 meq/L on any subsequent day), the patient should be encourage to either drink more water, or the dose should be held or reduced. After the appropriate dose has been identified, the patient may be discharged and continued on tolvaptan long‐term.

With the advent of this exciting therapy, practical issues will need to be addressed, most important of which is its cost at $250 per day (Otsuka). In addition, the current recommendation to initiate tolvaptan only in a hospital further limits its widespread use. Most important, long‐term clinical benefit will need to be demonstrated. Although the SALT trials only involved treatment for up to 1 month, a multicenter, open‐label extension study for a mean duration of 701 days demonstrated that prolonged administration of tolvaptan maintains an increased serum sodium level.58 However, at this time, tolvaptan can only be considered as one of the promising drugs whose long‐term cost‐effectiveness is yet to be proven. Proof will require showing that correction of the hyponatremia leads to improved clinical outcomes, such as a reduction in length of stay or frequency of hospitalization, decreased renal failure, improved hepatic encephalopathy, deceased mortality, and improved post‐transplant outcomes.

Unanswered Questions

The vaptans provide an important opportunity to clarify the role that hyponatremia plays in the pathogenesis of cirrhosis. In the past, DH in a cirrhotic patient represented a sign of advanced disease. With the availability of safe and effective therapy, we can now determine whether it also plays an important role in the pathophysiology of end‐stage liver disease and whether its treatment will have a beneficial effect on patient outcomes.

Specific clinical questions that will inevitably be addressed over the next few years to determine whether DH is only a marker for advanced disease or whether it plays a direct but modifiable role in the pathophysiology of cirrhosis will include:

  • Role of vaptans in the management of ascites: In a 14‐day randomized, trial of a satavaptan, another selective vasopressin V(2) receptor antagonist, vs. placebo with spironolactone, combination therapy was associated with improved control of ascites and improvements in serum sodium levels in hyponatremic patients with ascites.59 If future similar studies demonstrate more prolonged benefits, this would constitute an important advance in the treatment of ascites in cirrhosis.

  • Effect on renal function: Prolonged use of tolvaptan leads to a compensatory increase in endogenous levels of AVP and, potentially, increased stimulation of V1a receptors, which might be helpful in the setting of portal hypertension. In patients with hepatorenal syndrome, vasopressin stimulation of splanchnic V1a receptors leads to improved renal function, presumably by decreasing splanchnic blood flow and improving central blood volume.60 As a result, tolvaptan may indirectly improve kidney function in patients with advanced cirrhosis and refractory ascites. Whether long‐term tolvaptan therapy will help to prevent hepatorenal syndrome through this mechanism remains to be determined but is an exciting possibility.61

  • Effect on hepatic encephalopathy: Hepatic encephalopathy is associated with poor quality of life in patients with cirrhosis. Although hepatic encephalopathy was not directly assessed in the SALT trials, the mean mental component summary of the Short Form General Health Survey, a quality of life measure, improved in cirrhotic patients receiving tolvaptan to a greater degree that those receiving placebo.62 A possible explanation for this finding is a beneficial effect of tolvaptan on hepatic encephalopathy. Confirmation of this hypothesis, however, will require prospective studies in which hepatic encephalopathy is directly assessed.

  • Effect on medical economics: Based on retrospective reviews, hyponatremia has an adverse impact on length of stay and outcomes following liver transplantation. It will be important to demonstrate in prospective studies that correction of hyponatremia with tolvaptan reduces length of stay, complications, and costs.

References
  1. Nielsen S,Marples D,Frokiaer J, et al.The aquaporin family of water channels in kidney: an update on physiology and pathophysiology of aquaporin‐2.Kidney Int.1996;49:17181723.
  2. Gines P,Berl T,Bernardi M, et al.Hyponatremia in cirrhosis: from pathogenesis to treatment.Hepatology.1998;28:851864.
  3. Gines P,Guevara M.Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management.Hepatology.2008;48:10021010.
  4. Adrogue HJ,Madias NE.Hyponatremia.N Engl J Med.2000;342:15811589.
  5. Callahan MA,Do HT,Caplan DW,Yoon‐Flannery K.Economic impact of hyponatremia in hospitalized patients: a retrospective cohort study.Postgrad Med.2009;121:186191.
  6. Hawkins RC.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chim Acta.2003;337:169172.
  7. Adrogue HJ.Consequences of inadequate management of hyponatremia.Am J Nephrol.2005;25:240249.
  8. Liamis G,Milionis H,Elisaf M.A review of drug‐induced hyponatremia.Am J Kidney Dis.2008;52:144153.
  9. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  10. Upadhyay A,Jaber BL,Madias NE.Incidence and prevalence of hyponatremia.Am J Med.2006;119(7 Supple 1):S30S35.
  11. Bagshaw SM,Townsend DR,McDermid RC.Disorders of sodium and water balance in hospitalized patients.Can J Anesth.2009;56:151167.
  12. Renneboog B,Musch W,Vandemergel X, et al.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119;71.e1‐8.
  13. Sterns RH,Silver SM.Brain volume regulation in response to hypo‐osmolality and its correction.Am J Med.2006;119 (7 Suppl 1):S12S16.
  14. Arieff AI,Llach F,Massry SG.Neurological manifestations and morbidity of hyponatremia: correlation with brain water and electrolytes.Medicine.1976;55:121129.
  15. Iwakiri Y,Groszmann RJ.The hyperdynamic circulation of chronic liver diseases: from the patient to the molecule.Hepatology.2006;43:S121S131.
  16. Akriviadis EA,Ervin MG,Cominelli F, et al.Hyponatremia of cirrhosis: role of vasopressin and decreased “effective” plasma volume.Scand J Gastroenterol.1997;32:829834.
  17. Solis‐Herruzo JA,Gonzalez‐Gamarra A,Castellano G,Muñoz‐Yagüe MT.Metabolic clearance rate of arginine vasopressin in patients with cirrhosis.Hepatology.1992;16:974979.
  18. Schrier RW.Water and sodium retention in edematous disorders: role of vasopressin and aldosterone.Am J Med.2006;119:S47S53.
  19. Ruiz‐del‐Arbol L,Monescillo A,Arocena C, et al.Circulatory function and hepatorenal syndrome in cirrhosis.Hepatology.2005;42:439447.
  20. Chung SH,Jun DW,Kim KT, et al.Aquaporin‐2 urinary excretion in cirrhosis: relationship to vasopressin and nitric oxide.Dig Dis Sci.2010;55(4):11351141.
  21. Angeli P,Wong F,Watson H, et al.Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44:15351542.
  22. Kim WR et al.Hyponatremia and mortality among patients on the liver‐transplant list.N Engl J Med.2009;359:10181026.
  23. Planas R,Montoliu S,Balleste B, et al.Natural history of patients hospitalized for management of cirrhotic ascites.Clin Gastroenterol Hepatol.2006;4:13851394.
  24. Borroni G,Maggi A,Sangiovanni A, et al.Clinical relevance of hyponatremia for the hospital outcome of cirrhotic patieints.Dig Liver Dis.2000;32:605610.
  25. Wu CC,Yeung LK,Tsai WS, et al.Incidence and factors predictive of acute renal failure in patients with advanced liver cirrhosis.Clin Nephrol.2006;65:2833.
  26. Jenq CC,Tsai MH,Tian YC, et al.Serum sodium predicts prognosis in critically ill cirrhotic patients.J Clin Gastroenterol2010;44(3):220226.
  27. Ruf AE,Kremers WK,Chavez LL, et al.Addition of serum sodium into the MELD score predicts waiting list mortality better than MELD alone.Liver Transpl.2005;11:336343.
  28. Heuman DM,Abou‐Assi SG,Habib A, et al.Persistent ascites and low serum sodium identify patients with cirrhosis and low MELD scores who are at high risk for early death.Hepatology.2004;40:802810.
  29. Lv XH,Liu HB,Wang Y, et al.,Validation of model for end‐stage liver disease score to serum sodium ratio index as a prognostic predictor in patients with cirrhosis.J Gastroenterol Hepatol.2009;24:15471553.
  30. Kim MY,Liu HB,Wang Y, et al.Hepatic venous pressure gradient can predict the development of hepatocellular carcinoma and hyponatremia in decompensated alcoholic cirrhosis.Eur J Gastroenterol Hepatol.2009;21:12411246.
  31. Biggins SW,Rodriguez HJ,Bacchetti P, et al.Serum sodium predicts mortality in patients listed for liver transplantation.Hepatology.2005;41:3239.
  32. Hackworth WA,Heuman DM,Sanyal AJ, et al.Effect of hyponatremia on outcomes following orthotopic liver transplantation.Liver Int.2009;29:10711077.
  33. Vila MC,Coll S,Sola R, et al.Total paracentesis in cirrhotic patients with tense ascites and dilutional hyponatremia.Am J Gastroenterol.1999;94:22192223.
  34. Porcel A,Diaz F,Rendon P, et al.Dilutional hyponatremia in patients with cirrhosis and ascites.Arch Intern Med.2002;162:323328.
  35. Somberg JC,Molnar J.Therapeutic approaches to the treatment of edema and ascites: the use of diuretics.Am J Ther.2009;16:98101.
  36. Gines P,Tito L,Arroyo V, et al.Randomized comparative study of therapeutic paracentesis with and without intravenous albumin in cirrhosis.Gastroenterol.1988;94:14931502.
  37. Solis‐Herruzo JA,Moreno D,Gonzalez A, et al.Effect of intrathoracic pressure on plasma arginine vasopressin levels.Gastroenterology.1991;101:607617.
  38. Wen SF.Nephrotoxicities of nonsteroidal anti‐inflammatory drugs.J Formos Med Assoc.1997;96:157171.
  39. Terg R,Gadano A,Cartier M, et al.Serum creatinine and bilirubin predict renal failure and mortality in patients with spontaneous bacterial peritonitis: a retrospective study.Liver Int.2009;29:415419.
  40. Haussinger D,Schliess F.Pathogenic mechanisms of hepatic encephalopathy.Gut2008;57:11561165.
  41. Restuccia T,Gomez‐Anson B,Guevara M, et al.Effects of dilutional hyponatremia on brain organic osmolytes and water content in patients with cirrhosis.Hepatology.2004;39:16131622.
  42. Hausinger D.Low grade cerebral edema and the pathogenesis of hepatic encephalopathy in cirrhosis.Hepatology.2006;43:11871190.
  43. Amodio P,Del Piccolo F,Petteno E, et al.Prevalence and prognostic value of quantified electroencephalogram (EEG) alterations in cirrhotic patients.J Hepatol.2001;35:3745.
  44. Guevara M,Baccaro ME,Torre A, et al.Hyponatremia is a risk factor of hepatic encephalopathy in patients with cirrhosis: a propective study with time‐dependent analysis.Am J Gastroenterol.2009;104:13821389.
  45. Jalan R,Elton RA,Redhead DN, et al.Analysis of prognostic variables in the prediction of mortality, shunt failure, variceal rebleeding and encephalopathy following the transjugular intrahepatic portosystecim stent‐shunt for variceal haemorrhage.J Hepatol.1995;2:123128.
  46. Ripoll C,Catalina MV,Yotti R, et al.Cardiac dysfunction during liver transplantation: incidence and preoperative predictors.Transplantation.2008;85:17661772.
  47. Yun BC,Kim WR,Benson JT, et al.,Impact of pretransplant hyponatremia on outcome following liver transplantation.Hepatology.2009;49:16101615.
  48. Londono MC,Guevara M,Rimola A, et al.Hyponatremia impairs early posttransplantation outcome in patients with cirrhosis undergoing liver transplantation.Gastroenterology.2006;130:11351143.
  49. Martin‐Llahi M,Guevara M,Gines P.Hyponatremia in cirrhosis: clinical features and management.Gastroenterol Clin Biol.2006;30:11441151.
  50. McCormick PA,Mistry P,Kaye G, et al.Intravenous albumin infusion is an effective therapy for hyponatremia in cirrhotic patients with ascites.Gut.1990;31:204207.
  51. Bosch‐Marce M,Poo JL,Jiménez W, et al.Comparison of two aquaretic drugs (niravoline and OPC‐31260) in cirrhotic rats with ascites and water retention.J Pharmacol Exp Ther.1999;289:194201.
  52. Miller PD,Linas SL,Schrier RW.Plasma demeclocycline levels and nephrotoxicity. Correlation in hyponatremic cirrhotic patients.JAMA.1980;243:25132515.
  53. Gines A,Salmeron JM,Gines P, et al.Oral misoprostol or intravenous prostaglandin E2 do not improve renal function in patients with cirrhosis and ascites with hyponatremia and renal failure.J Hepatol.1993;17:220226.
  54. Fernandez‐Varo G,Ros J,Cejudo‐Martin P, et al.Effect of the V1a/V2‐AVP receptor antagonist, Conivaptan, on renal water metabolism and systemic hemodynamics in rats with cirrhosis and ascites.J Hepatol.2003;38:755761.
  55. Schrier RW,Gheorghiade M,Berl T, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  56. Sort P,Navasa M,Arroyo V, et al.Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis.N Engl J Med.1999;341:403409.
  57. Sigal SH,Stanca CM,Fernandez J, et al.Restricted use of albumin for spontaneous bacterial peritonitis.Gut.2007;56:597599.
  58. Berl T,Quittnat‐Pelletier F,Verbalis JG, et al.Oral tolvaptan is safe and effective in chronic hypyonatremia.J Am Soc Nephrol.2010;21(4):705712.
  59. Gines P,Wong F,Watson H, et al.Effects of satavaptan, a selective vasopressin V(2) receptor antagonist, on ascites and serum sodium in cirrhosis with hyponatremia: a randomized trial.Hepatology.2008;48:204213.
  60. Sanyal AJ,Boyer T,Garcia‐Tsao G, et al.A randomized, prospective, double‐blind, placebo‐controlled trial of terlipressin for type 1 hepatorenal syndrome.Gastroenterology.2008;134:13601368.
  61. Dixon MB,Lien YH.Tolvapten and its potential in the treatment of hyponatremia.Ther Clin Risk Manag.2008;4:114911455.
  62. Cardenas A,Gines P,Marotta P, et al.The effects of vasopressin V2 receptor antagonist in the management of patients with cirrhosis and hyponatremia. Safety and efficacy of oral tolvaptan in the SALT trials.Hepatology.2009;50S:467A.
  63. Decaux G,Mols P,Cauchie P., et al.Treatment of hyponatremic cirrhosis with ascites resistant to diuretics by urea.Nephron.1986;44:337343.
  64. Gadano A,Moreau R,Pessione F, et al.Aquaretic effects of niravoline, a kappa‐opioid agonist, in patients with cirrhosis.J Hepatol.2000;32:3842.
  65. Kim JH,Lee JS, et al.The association between the serum sodium level and the severity of complications in liver cirrhosis.Korean J Intern Med.2009;24:106112.
  66. Such J,Hillebrand DJ,Guamer C, et al.Nitric oxide in ascitic fluid is an independent predictor of the development of renal impairment in patients with cirrhosis and spontaneous bacterial peritonitis.Eur J Gastroenterol Hepatol.2004;16:571577.
Article PDF
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S8-S17
Legacy Keywords
hyponatremia, common electrolyte disorder, cirrhosis, portal hypertension, cirrhosis, liver failure
Sections
Article PDF
Article PDF

The serum sodium (Na) level is the major determinant of serum osmolality. In normal physiologic states is tightly regulated between 135 mEq/L to 145 mEq/L despite variable intake of water and solute through the interaction of osmoreceptors in the hypothalamus where arginine vasopressin (AVP) is synthesized and then released by the posterior pituitary and the binding of AVP with V2 AVP receptors on the basolateral surface of the principal cells within the collecting duct of the kidney. Binding of AVP to the V2 receptors promotes the translocation and fusion of cytoplasmic vesicles which carry the water channel protein aquaporin 2 (AQP2) to the apical membrane of the cell and, in this manner, increases water permeability and absorption.1, 2, 3

Patients with hyponatremia, defined by a serum Na level <135 mEq/L, can be broadly classified by their volume status into those who are euvolemic, hypervolemic, and hypovolemic (Table 1). In patients with euvolemic hyponatremia such as those with Syndrome of Inappropriate Antidiuretic Hormone (SIADH), total body Na is nearly normal, but total body water is increased. In patients with hypervolemic hyponatremia, both total body Na and water are increased, but water to a much greater degree. These patients typically have increased extracellular fluid such as edema and/or ascites. The most common conditions associated with this condition are cirrhosis, congestive heart failure (CHF), and renal failure. In contrast, hypovolemic hyponatremia is associated with a reduction in both total body Na and water, but Na to a greater degree. This condition is encountered in patients with excessive fluid losses such as those with over‐diuresis, excessive gastrointestinal losses, burns, and pancreatitis.4

Classification of Hyponatremia: Sodium and Water Changes in the 3 Different Types of Hyponatremia
Depletional Hyponatreima Dilutional Hyponatremia
Euvolumic Hypervolumic
Total body water
Total body Na normal
Common etiologies SIADH cirrhosis/CHF vomiting, diarrhea

Hyponatremia is the most common electrolyte abnormality seen in general hospital patients.5 In a database of over 120,000 patients, a serum sodium level of <136mEq/L was observed in 28.2%.6 Hyponatremia is associated with selected medical conditions (especially cirrhosis and CHF), the extremes of age, and those receiving selected medications, including several that are commonly administered to cirrhotic patients (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors).7, 8 Hyponatremia is associated with increased total costs per hospital admission.5, 9 In an analysis of the effect of hyponatremia on length of stay in a retrospective cohort study of hospitalized patients derived from a large administrative database of 198,281 discharges from 39 US hospitals, mean length of stay was significantly greater among patients with hyponatremia than those with normal Na levels (8.6 8.0 vs. 7.2 8.2 days). After adjusting for confounders that may be associated with more severe disease and hyponatremia (age, gender, race, geographic region, teaching status of the hospital, admission source, principal payer, comorbidity index score and primary diagnosis), the presence of hyponatremia contributed an increase in length of stay of 1.0 day. Patients with hyponatremia are more frequently admitted to the intensive care unit (ICU) and require mechanical ventilation. In patients with CHF, the presence of hyponatremia at discharge is associated with increased risk for early mortality and rehospitalization.10

Although frequently asymptomatic, hyponatremia may be associated with a range of findings, from subtle and non‐specific complaints, including headache, fatigue, confusion, malaise, to severe and life‐threatening manifestations with lethargy, seizures, brainstem herniation, respiratory arrest and death.11 The most important complications are neurologic consequences related to cerebral edema. However, there is increased morbidity even in hyponatremic patients considered to be asymptomatic. Patients with low serum sodium have attention deficients, and falls are common. In a study of 122 patients who were considered to have chronic asymptomatic hyponatremia, the incidence of falls was significantly higher at 21.3% compared to only 5.3% in a control population.12

In hyponatremia, water enters into the cells to attain osmotic balance, resulting in cellular swelling.4 To avoid cerebral edema, the brain is capable of adapting to hyponatremia by regulating its volume to avoid swelling, especially when hyponatremia is chronic. In acute hyponatremia, astrocytes and neurons adapt through osmoregulatory mechanisms by extruding intracellular electrolytes such as potassium.13 Chronically, adaption occurs through the loss of low‐molecular weight organic compounds termed organic osmolytes including myoinsoitol, glutamine, choline and taurine. As a result, both the severity and the rate of its development are critical factors in determining the neurologic manifestation of hyponatremia in a given patient.14

Dilutional Hyponatremia and Cirrhosis

Patients with hyponatremia who are either euvolemic or hypervolemic are considered to have dilutional hyponatremia (DH). Management of these patients is distinct from those who are hypovolemic in whom appropriate therapy consists of the administration of normal saline. The remainder of this article addresses the pathogenesis, management and treatment of cirrhotic patients with DH.

Pathogenesis

The development of hyponatremia in cirrhosis is intimately related to the pathophysiology of portal hypertension and the non‐osmotic release of AVP3, 15 (Figure 1). In the early phases of cirrhosis, portal hypertension is the result of an increase in intrahepatic resistance. With the development of porto‐systemic collaterals, a hyperdynamic splanchnic circulation develops as a result of splanchnic arterial vasodilatation and increased vascular capacity. Nitric oxide, an endothelial derived relaxing factor, is the critical mediator of this process, and upregulation of its expression is pivotal in the pathogenesis of portal hypertension.

Figure 1
Proposed mechanism of hypersecretion and renal and systemic effects of vasopressin in cirrhosis with ascites. Gines P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management. Hepatology. 2008;48:1002–1010. Copyright 2008 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.

Multiple factors are related to the development of DH in cirrhosis. A reduction of effective central blood volume due to the development of porto‐venous collaterals and arterial splanchnic vasodilation, leading to baroreceptor‐mediated nonosmotic release of AVP, is considered the initiating and most important factor. Patients with cirrhosis and DH have higher plasma and urine vasopressin levels, higher plasma renin activity, and decreased plasma levels of atrial natriuretic factor than those with normal serum sodium concentrations, findings consistent with the presence of a decreased effective plasma volume.16 Arterial underfilling is sensed by baroreceptors located in the left ventricle, aortic arch, carotid sinus and renal afferent arterioles. Decreased activation leads to neurohumoral compensatory responses which include non‐osmotic release of vasopressin from the neurohypophysis and increased levels. Impaired catabolism of AVP that has been correlated with the severity of liver dysfunction may further contribute to increased levels.17 Initially, the increased AVP maintains arterial circulatory integrity by inducing splanchnic, peripheral and renal arterial vasoconstriction through its action on the V1a receptors and expansion of blood volume through renal water retention by its action on the V2 receptors located on the collecting ducts.

The initial adaptive response which leads to increased central blood volume can chronically result in detrimental effects, including the development of fluid overload with ascites, edema, and hyponatremia.16, 18 Additional factors that contribute to hyponatremia include decreased glomerular filtration rate (GFR) and/or increased proximal reabsorption of sodium (that reduce the distal delivery of filtrate and the potential for water reabsorption) and decreased cardiac function that further impairs effective central blood volume.19 In addition, urinary levels of AQP2 are increased in cirrhotic patients, especially those with decompensated disease with higher Child‐Pugh scores and ascites, and provide another potential mechanism to increase water reabsorption.20

Prevalence and Prognostic Significance

Hyponatremia in cirrhosis is a common finding. In a survey of 997 cirrhotic patients with ascites from 28 centers in Europe, North and South America, the prevalence of serum sodium concentration 135, 130, 125, 120 meq/L were 49.4%, 21.6%, 5.7%, and 1.2%, respectively.21 In a retrospective analysis of 188 inpatients, the prevalence of DH of 135, 130, and 125 were 20.8%, 14.9%, and 12.2%, respectively.22 The development of hyponatremia is a manifestation of increasing portal hypertension. In a natural history study of 263 patients hospitalized for first episode of significant ascites, 74 patients developed DH (Na level < 130 mEq/L), including 11 patients in whom it appeared during the first episode and 63 cases during follow‐up (mean period of 40 3 months) with a 5‐year incidence of 37.1%.23

The presence of hyponatremia carries significant adverse prognostic significance. It is strongly associated with severity of liver function impairment as assessed by Child‐Pugh and model for end‐stage liver disease (MELD) scores.22 Even mild hyponatremia is associated with severe complications such as massive ascites, severe hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), and hepatic hydrothorax, and the severity of hyponatremia is directly related to the severity of these complications.21, 22 (Figure 2). In a natural history study of patients presenting with large volume ascites, 1‐year survival after its development was reduced to only 25.6%.230

Figure 2
Percentage of patients with complications of cirrhosis classified according to serum sodium concentration. Angeli P, Wong F, Watson H, et al. Hyponatremia in cirrhosis: results of a patient population survey. Hepatology. 2006;44:1535–1542. Copyright 2006 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.
Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Hyponatremia is an especially poor prognostic sign for a hospitalized cirrhotic patient. In a retrospective analysis of 156 cirrhotic patients, hyponatremiapresent in 57 (29.8%) of admissionswas associated with increased hospital mortality (26.3% vs. 8.9% among those with normal Na levels), and the mortality rate was even higher (48%) among the 25 patients who developed severe hyponatremia during the hospital stay.24 In hospitalized patients, hyponatremia is predictive of the development of acute renal failure which is associated with substantially increased mortality (73% vs. 13%).25 Similarly, a low serum sodium level in critically ill cirrhotic patients admitted to the ICU is associated with complications, in‐hospital mortality, and poor short‐term prognosis.26

Whether hyponatremia should impact liver transplant prioritization remains an area of controversy. The United Network for Organ Sharing (UNOS) contracted by the Organ Procurement and Transplant Network (OPTN) to optimize the efficient use of deceased organs through fair and timely allocation, currently uses the MELD score, a formula that calculates the risk of death within three months from the bilirubin, creatinine, and International Normalized Ratio (INR) levels. Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction in patients with advanced cirrhosis and adds to MELD in predicting waitlist mortality.2729 In patients with a MELD score of <21, only low serum sodium and persistent ascites are independent predictors of mortality.28 To account for the importance of hyponatremia on survival, both modification of the MELD score in which the Na level is incorporated (MELD‐Na model) and the MELD to serum sodium ratio (MESO) have been developed. Adding hyponatremia to the MELD score is a better predictor of death than MELD alone, particularly in patients with low MELD scores.27, 2931 The OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has discussed updating the liver allocation system to include the Na level. However, it was concluded that implementation of MELD‐Na would change the allocation status of only 4% of candidates. Further, based on the concerns about the ability to manipulate serum sodium levels and the utility of employing resources to change the system for a relatively small number of patients, it was decided to defer incorporating the Na level pending further analysis (Report of the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee To the Board of Directors, Los Angeles, California, September 17‐18, 2007). At this time, the use of Na is a regional decision.32 However, the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has recently solicited feedback from the transplant community about including Na in allocation for review at a forum in April 2010.

Precipitating Factors

The most important factor related to development of hyponatremia in cirrhosis is increasing severity of portal hypertension that is associated with impaired central blood volume as a result of progressive splanchnic vasodilatation. In a study in which 170 patients with decompensated alcoholic cirrhosis were prospectively followed for 33.9 27.9 months, the initial hepatic venous pressure gradient (HVPG) was an independent predictive factor for the 20 patients who developed hyponatremia.22

Cirrhotic patients with ascites with hyponatremia have increased AVP secretion, higher levels of plasma renin activity, and higher serum concentrations of aldosterone and norepinephrine compared to those with normal Na levels.33 Diuretic therapy is associated with the development of DH by inducing volume depletion and arterial underfilling, further activating the renin‐angiotensin system (RAS) and increasing the non‐osmotic release of AVP.34 Although diuretics block the salt retention associated with the RAS activation, the water‐retaining effects of AVP persist, and DH develops. The process is further exacerbated by a low sodium intake and a frequent uncontrollable thirst. As a result, diuretic therapy is commonly associated with the development of hyponatremia in patients with ascites.24, 35 Similarly, paracentesis (particularly when performed without albumin) is often associated with an increase in blood urea nitrogen and marked elevations in plasma renin activity and plasma aldosterone concentration, which may exacerbate this physiology, leading to further reduction in serum sodium concentration.36 Tense ascites can contribute to DH by increasing baroreceptor mediated AVP release by increasing intrathoracic pressure.37 Finally, non‐steroidal anti‐inflammatory drugs (NSAIDs) can cause DH by inhibiting the synthesis of renal prostaglandins (which normally function to antagonize the tubular action of AVP and are important in the maintenance of appropriate renal tubular transport of fluid and electrolytes in states of renal hypoperfusion).38

Medical Impact of Hyponatremia: Marker of Severe Disease or Direct Pathophysiologic Role?

Hyponatremia is associated with severe ascites, impaired renal function, hepatic encephalopathy, SBP, and hepatorenal syndrome.3, 20 Because hyponatremia is frequently present in advanced liver failure, it is unclear whether it is only a marker of advanced disease or whether it plays a direct pathophysiologic role, or both. Until recently, it has not been possible to address this issue due to the inability to easily and rapidly correct the hyponatremia. However, there is increasing evidence that hyponatremia has direct impact on the severity of hepatic encephalopathy (see Hepatic Encephalopathy section). The recent introduction of tolvaptan for the treatment of hyponatremia in cirrhosis (discussed below) will allow this question to be directly answered.

Fluid Management and Diuresis

The typical cirrhotic patient with DH is characterized by expanded extracellular fluid with ascites and edema. The profound vasodilation of the splanchnic arterial circulation is associated with decreased effective arterial blood volume, leading to the non‐osmotic release of AVP. Diuretic therapy can further exacerbate this process. In addition, the increased water permeability induced by AVP results in reduced urine volume and fluid retention. As a result, hyponatremia directly adversely affects severity of fluid overload and limits and/or precludes diuretic treatment.

Hepatorenal Syndrome

Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction and is frequently a precursor to overt hepatorenal syndrome.27 Hyponatremia is predictive of the development of acute renal failure during hospitalization, and in‐hospital development of acute renal failure portends a high mortality.25 In patients admitted with SBP, the presence of hyponatremia is significantly associated with higher mortality and renal failure.39

Hepatic Encephalopathy

The neurologic manifestations of cerebral edema associated with hyponatremia closely mirror those of hepatic encephalopathy. In fact, a recently proposed pathogenic mechanism for hepatic encephalopathy is the development of low‐grade cerebral edema associated with astrocyte swelling in response to ammonia and other precipitating factors.40 DH is associated with a further reduction in brain organic osmolytes that probably reflects a compensatory osmoregulatory mechanism against cell swelling triggered by a combination of high intracellular glutamine and low extracellular osmolality.41 As a result, it has been proposed that hyponatremia contributes to the development of hepatic encephalopathy through the development or exacerbation of low‐grade cerebral edema. In this manner, low serum sodium acts as a second hit to the swelling produced by increased intracellular glutamine created by ammonia metabolism.42

Clinically, hyponatremia is a major risk factor for hepatic encephalopathy. Serum sodium and ammonia levels are the major factors that predict electroencephalographic abnormalities in cirrhotics who do not have hepatic encephalopathy.43 In a prospective study of 61 patients, hyponatremia was associated with a low brain concentration of organic osmolytes as assessed by proton magnetic resonance spectroscopy (1H‐MRS) and magnetic resonance imaging, and both conditions were major risk factors for the development of overt hepatic encephalopathy.44 Finally, hyponatremia is a risk factor for hepatic encephalopathy in patients undergoing TIPS.45

Adverse Effect on Outcome After Liver Transplantation

Hyponatremia before liver transplantation is associated with adverse post‐transplant outcomes. Among patients undergoing liver transplantation, the presence of hyponatremia is associated with abnormal cardiac response in patients after reperfusion.46 Pre‐transplant hyponatremia is associated with longer ICU and hospital stay, higher rates of delirium and neurologic disorders, acute renal failure, acute cellular rejection, infection, and in one study a reduced 3‐month survival compared to normonatremic recipients.32, 47, 48 In 1 retrospective study that compared post‐transplant outcomes of patients with corrected vs. uncorrected pre‐transplant hyponatremia, patients with pre‐operative correction of hyponatremia had a lower risk of prolonged post‐transplant hospitalization than those with uncorrected hyponatremia.32 However, both hyponatremic groups had more complicated post‐transplant courses compared to those without a history of hyponatremia. However, given the small sample size, retrospective design, and the potential for confounding, the impact of correction of pre‐transplant hyponatremia remains to be determined.

Management

Most patients with mild hypervolemic hyponatremia are asymptomatic. The initial recommended approach is fluid restriction and an Na‐restricted diet. For those with severe or progressive hyponatremia, diuretics should be minimized or discontinued to avoid intravascular volume depletion.49 For patients with tense ascites and severe DH, therapeutic paracentesis with plasma expanders is safe.33 Unfortunately, fluid restriction is limited in efficacy and often poorly tolerated. The use of hypertonic saline is generally not recommended unless severe neurologic symptoms are present as it leads to increased ascites and edema. When administered, it is important to avoid a rapid correction of the hyponatremia to prevent the development of central pontine myelinolysis and the osmotic demyelination syndrome.

Due to the pivotal role of AVP in the pathogenesis of DH, antagonism of its action has long been proposed to be the most rational approach, but until recently, effective and specific antagonism of AVP has remained elusive. Approaches that have been attempted include interference with its secretion and actions. Intravenous albumin has been reported to improve hyponatremia in patients with cirrhosis, ascites, and hyponatremia, presumably by decreasing AVP release by plasma volume expansion.50 An attempt at inhibition of central AVP release with the use of a kappa‐opioid receptor agonist, niravoline, was limited by loss of efficacy and potential adverse effects.51 Use of demeclocycline and lithium (which induce renal resistance to AVP and lead to a modest increase in urine volume with decreased urine osmolality and a corresponding rise in serum sodium) is limited by nephrotoxicity and hepatotoxicity.7, 52 Because of the important role played by prostaglandins in the maintenance of renal hemodynamics and water excretion in cirrhosis, oral misoprostol has also been evaluated but determined to be ineffective in inducing significant changes in free water clearance in patients with functional renal failure and/or DH.53

The recent introduction of vaptans, vasopressin receptor antagonists that block the physiologic action of vasopressin, represents a revolutionary and highly effective approach to the treatment of hyponatremia. Vaptans are antagonists of the V2 receptors of AVP in the principal cells of the collecting ducts. In healthy subjects, vaptans cause a dose‐dependent increase in urine volume and produce a dilute urine without causing natriuresis. To date, 2 AVP antagonists, conivaptan and tolvaptan, have been Food and Drug Administration (FDA)‐approved for the treatment of DH. Conivaptan, the first to be approved in 2005, is a mixed vasopressin V1a and V2 receptor antagonist that is administered intravenously for up to 4 days. In a randomized placebo‐controlled study of patients with euvolemic or hypervolemic hyponatremia, intravenous conivaptan treatment increased serum Na levels by >6 mEq/l or to a serum Na >135 mEq/l in 69 to 88.5% of subjects compared to 20.7% of those receiving placebo (Zeltser D, Rosansky S, Van Rensburg H, et al. Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia. American J Nephrology 2007;27:447457). In a pilot study involving 24 patients with end‐stage liver disease, an infusion of conivaptan over 1 to 4 days was associated with an increase of serum sodium by >5 mmol/L in 60% of patients not receiving diuretics and 67% of patients on concomitant diuretic therapy by the end of treatment (O'Leary and Davis, 2009). Despite a concern about the potential for conivaptan to increase portal hypertension due to inhibition of splanchnic V1a receptors, the brief treatment appeared to be well tolerated without significant changes in systolic blood pressure, serum creatinine, variceal bleeding or worsening of ascites during the infusion period. However, approval for only 4 days of therapy and requirement for intravenous use eliminate any potential for chronic use.54

Tolvaptan is an orally available, selective V2 receptor antagonist whose efficacy was assessed in two multicenter, prospective, randomized, placebo‐controlled trials, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2).55 In these trials, clinically stable patients with DH (Na <135meq/l) associated with cirrhosis (22.4% in SALT‐1, 30.5% in SALT‐2), CHF or SIADH were randomized in a hospital setting to receive tolvaptan 15mg daily or placebo. Repeat Na levels were obtained at 8 hours, 2, 3, and 4 days and then weekly at days 11, 18, 25 and 30 after which study drug was discontinued and follow‐up Na level was determined 7 days later. The dose was adjusted to 30 mg and then 60 mg in an attempt to achieve a Na level >135 in those in whom hyponatremia persisted. During the initial day of the titration phase, fluid restriction was not maintained, and the patients were encouraged to respond to thirst with increased water ingestion.

Tolvaptan use was associated with a prompt increase in Na level as early as 8 hours after administration of the first dose. Serum Na increased more among those receiving tolvaptan than among those receiving placebo during the first 4 days and throughout the study period regardless of baseline Na level but returned to baseline within 1 week after discontinuation (Figure 3). The main side effects were increased thirst, dry mouth and increased urination. Importantly, an increased incidence of renal failure was not observed. Based on these results, FDA approval for tolvaptan in patients with hyponatremia was obtained in May 2009 for patients with DH‐associated with cirrhosis, CHF or SIADH for patients with Na levels <125 or symptomatic patients with Na levels between 125 and 135 that have not responded to fluid restriction.

Management of the Hospitalized Cirrhotic Patient With Hyponatremia: Recommendations

Hyponatremia in hospitalized cirrhotic patients is a marker for severe disease and high risk of hospital mortality.24 As a result, prompt evaluation and treatment is imperative. The availability of tolvaptan potentially revolutionizes the manner in which these patients are treated. In the SALT trials, only clinically stable patients were enrolled. In this last section, a guideline for the evaluation and treatment of acutely ill, hospitalized cirrhotic patients with DH is presented.

Evaluation

Determination of volume status is paramount but frequently problematic in the hospitalized cirrhotic patient. Due to the vasodilated state present in severe portal hypertension that is characterized by a relative hypotension and resting tachycardia, the usual hemodynamic parameters of blood pressure and heart rate can be difficult to interpret. Although significant extravascular volume in the form of ascites and edema may be present, patients may be intravascularly depleted due to previous diuretic use and extra‐renal losses due to impaired oral intake, vomiting, lactulose‐induced diarrhea, and gastrointestinal bleeding. Infection is a commonly associated condition, and endotoxin mediated splanchnic vasodilatation, especially in the setting of SBP, can adversely effective central blood volume status in the presence of severe ascites. Also, due to the Na avidity of the kidney and previous diuretic use, renal electrolytes can be difficult to interpret.

For patients in whom there is strong clinical concern about intravascular depletion (history of impaired oral intake, excessive vomiting and/or diarrhea, rapid weight loss, small volume ascites with history of large volume, azotemia), administration of limited intravenous normal saline (0.5‐1 L) should be considered. Patients with severe neurological symptoms should receive normal saline or hypertonic saline. Unless severe neurologic symptoms associated with profound hyponatremia is present, however, intravenous normal saline should not be administered for the hyponatremia alone. Administration of salt poor albumin (25%), especially for those with marked fluid overload and ascites, is an effective means to expand the central blood volume without exacerbating ascites and edema. After evaluation for and/or treatment of hypovolemia, all patients should receive a Na restricted diet (<2000 mg daily) and placed on fluid restriction (see below for liberalization of fluid restriction upon initiation of tolvaptan therapy).

Diagnostic paracentesis should be performed for those with ascites to rule out the presence of SBP, and antibiotics administered to those with evidence of infection. High dose intravenous salt poor albumin should also be administered, especially to those at high risk of renal failure as determined by the presence of azotemia (Cr > 1.0 mg/dL) or severe liver insufficiency (TBili > 4.0 mg/dL).56, 57 Finally, all medications should be reviewed, and those associated with hyponatremia (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors) discontinued if possible.

Tolvaptan for DH

Patient Selection

Appropriate patient selection for tolvaptan therapy is extremely important (Table 2). In the SALT trials, only clinically stable patients were enrolled. The presence of hyponatremia in a recently hospitalized cirrhotic patient, however, frequently indicates severe disease with a high risk of acute renal failure and hospital death. In the SALT trials, many received concomitant diuretic therapy. Because of the importance of avoiding tolvaptan administration to hypovolemic patients, discontinuation of diuretic therapy prior the initiation of tolvaptan therapy and/or reevaluation after limited volume expansion should be considered.

Patient Selection for Tolvaptan Therapy for Hospitalized Patients With Cirrhosis and Hyponatremia
Hospital setting
Euvolumia or hypervolumia
Absence of recent weight loss, decrease in ascites, edema
Absence of excessive vomiting, diarrhea
Consider discontinuation of diuretic therapy prior to initiation of tolvaptan
Consider evaluation after limited volume expansion, especially with salt poor albumin prior to initiation of tolvaptan
Presence of clinically significant hyponatremia: 125mEq/L or less severe but symptomatic hyponatremia (125 to 134 mEq/L) that has resisted fluid restriction
Absence of severe neurologic symptoms attributable to hyponatremia
No co‐administration with intravenous saline
Ability to respond to thirst
No co‐administration with strong CYP 3A inhibitors (ketoconazole)
Absence of kidney failure with anuria

Tolvaptan is indicated for cirrhotic patients with DH in whom the serum sodium is <125 mEq/L and in those with less severe but symptomatic hyponatremia (125‐134 mEq/L) that has resisted fluid restriction. Although the definition of symptomatic was not specifically defined, possible considerations include symptoms of mild hepatic encephalopathy or inability to tolerate dieresis due to the presence of hyponatremia. According to FDA guidelines, tolvaptan therapy must be initiated and re‐initiated in a hospital setting. Patients with severe neurologic symptoms attributable to hyponatremia in whom rapid treatment is critical should not receive tolvaptan but should rather be treated with normal saline. Similarly, patients should not receive combination therapy with tolvaptan and normal saline due to potential for a too‐rapid correction of hyponatremia and the development of central pontine myelinolysis. If saline had been administered for treatment of possible hypovolemia, it should be discontinued and persistent hyponatremia confirmed before starting tolvaptan. Other factors that need to be considered before initiating tolvaptan include the ability of the patient to respond to thirst with increased water ingestion and recognition that the patient will experience increased urine volume and frequency, requiring easy access to toilet. Patients should not be fluid restricted during the first day of tolvaptan therapy, but should be instructed to respond to their thirst with increased water ingestion. As a result, caution should be exercised in administering tolvaptan to a confused, restrained, unresponsive and/or bed‐bound patient who is not able to respond appropriately to thirst or increased urination.

In the SALT trials, the incidence of hyperkalemia (5%) was similar in the tolvaptan and placebo treated patients.55 However, further analysis of all multiple‐dose, placebo‐controlled trials, demonstrated that the aggregate incidence of hyperkalemia was slightly higher for tolvaptan‐treated subjects compared with placebo‐treated subjects (Otsuka). Because treatment with tolvaptan is associated with an acute reduction of the extracellular fluid volume which could result in increased serum potassium through hemoconcentration, it is recommended that serum potassium levels be monitored after initiation of tolvaptan treatment in patients with a serum potassium > 5 mEq/L as well as those who are receiving drugs known to increase serum potassium levels such as angiotensin converting enzyme inhibitors, angiotensin receptor blockers, or potassium sparing diuretics (Samsca Package Insert, Otsuka). Because tolvaptan is metabolized by the cytochrome P 3A system, patients receiving strong inhibitors such as ketoconazole should not receive tolvaptan. Anuric patients will not respond to tolvaptan. Finally, it is extremely important to administer tolvaptan only to patients with true hyponatremia and not to those with pseudohyponatremia in whom the plasma osmolality is normal but the measured serum sodium concentration artificially low due to marked elevations of other substances, such as can be seen in severe hyperglycemia, marked hyperlipidemia, or hyperproteinemia (as in multiple myeloma).

Tolvaptan Administration

The initial dose of tolvaptan is 15 mg daily. After receiving tolvaptan, many patients will develop an increased sense of thirst and need to urinate. As a result, patients should not be fluid restricted during the first day of therapy, and it is important to monitor the hemodynamics and Na level closely after initiating therapy with a repeat Na level at approximately 8 hours after the first dose. As a result, it should probably be administered early in the day and not at bedtime. The dose should be increased to 30 mg, then 60 mg in patients who do not respond by at least 5 mEq/L over the previous 24 hours and remain hyponatremic. In those with an excessive response (more than 8 meq/L during the first 8 hours or 12 meq/L on any subsequent day), the patient should be encourage to either drink more water, or the dose should be held or reduced. After the appropriate dose has been identified, the patient may be discharged and continued on tolvaptan long‐term.

With the advent of this exciting therapy, practical issues will need to be addressed, most important of which is its cost at $250 per day (Otsuka). In addition, the current recommendation to initiate tolvaptan only in a hospital further limits its widespread use. Most important, long‐term clinical benefit will need to be demonstrated. Although the SALT trials only involved treatment for up to 1 month, a multicenter, open‐label extension study for a mean duration of 701 days demonstrated that prolonged administration of tolvaptan maintains an increased serum sodium level.58 However, at this time, tolvaptan can only be considered as one of the promising drugs whose long‐term cost‐effectiveness is yet to be proven. Proof will require showing that correction of the hyponatremia leads to improved clinical outcomes, such as a reduction in length of stay or frequency of hospitalization, decreased renal failure, improved hepatic encephalopathy, deceased mortality, and improved post‐transplant outcomes.

Unanswered Questions

The vaptans provide an important opportunity to clarify the role that hyponatremia plays in the pathogenesis of cirrhosis. In the past, DH in a cirrhotic patient represented a sign of advanced disease. With the availability of safe and effective therapy, we can now determine whether it also plays an important role in the pathophysiology of end‐stage liver disease and whether its treatment will have a beneficial effect on patient outcomes.

Specific clinical questions that will inevitably be addressed over the next few years to determine whether DH is only a marker for advanced disease or whether it plays a direct but modifiable role in the pathophysiology of cirrhosis will include:

  • Role of vaptans in the management of ascites: In a 14‐day randomized, trial of a satavaptan, another selective vasopressin V(2) receptor antagonist, vs. placebo with spironolactone, combination therapy was associated with improved control of ascites and improvements in serum sodium levels in hyponatremic patients with ascites.59 If future similar studies demonstrate more prolonged benefits, this would constitute an important advance in the treatment of ascites in cirrhosis.

  • Effect on renal function: Prolonged use of tolvaptan leads to a compensatory increase in endogenous levels of AVP and, potentially, increased stimulation of V1a receptors, which might be helpful in the setting of portal hypertension. In patients with hepatorenal syndrome, vasopressin stimulation of splanchnic V1a receptors leads to improved renal function, presumably by decreasing splanchnic blood flow and improving central blood volume.60 As a result, tolvaptan may indirectly improve kidney function in patients with advanced cirrhosis and refractory ascites. Whether long‐term tolvaptan therapy will help to prevent hepatorenal syndrome through this mechanism remains to be determined but is an exciting possibility.61

  • Effect on hepatic encephalopathy: Hepatic encephalopathy is associated with poor quality of life in patients with cirrhosis. Although hepatic encephalopathy was not directly assessed in the SALT trials, the mean mental component summary of the Short Form General Health Survey, a quality of life measure, improved in cirrhotic patients receiving tolvaptan to a greater degree that those receiving placebo.62 A possible explanation for this finding is a beneficial effect of tolvaptan on hepatic encephalopathy. Confirmation of this hypothesis, however, will require prospective studies in which hepatic encephalopathy is directly assessed.

  • Effect on medical economics: Based on retrospective reviews, hyponatremia has an adverse impact on length of stay and outcomes following liver transplantation. It will be important to demonstrate in prospective studies that correction of hyponatremia with tolvaptan reduces length of stay, complications, and costs.

The serum sodium (Na) level is the major determinant of serum osmolality. In normal physiologic states is tightly regulated between 135 mEq/L to 145 mEq/L despite variable intake of water and solute through the interaction of osmoreceptors in the hypothalamus where arginine vasopressin (AVP) is synthesized and then released by the posterior pituitary and the binding of AVP with V2 AVP receptors on the basolateral surface of the principal cells within the collecting duct of the kidney. Binding of AVP to the V2 receptors promotes the translocation and fusion of cytoplasmic vesicles which carry the water channel protein aquaporin 2 (AQP2) to the apical membrane of the cell and, in this manner, increases water permeability and absorption.1, 2, 3

Patients with hyponatremia, defined by a serum Na level <135 mEq/L, can be broadly classified by their volume status into those who are euvolemic, hypervolemic, and hypovolemic (Table 1). In patients with euvolemic hyponatremia such as those with Syndrome of Inappropriate Antidiuretic Hormone (SIADH), total body Na is nearly normal, but total body water is increased. In patients with hypervolemic hyponatremia, both total body Na and water are increased, but water to a much greater degree. These patients typically have increased extracellular fluid such as edema and/or ascites. The most common conditions associated with this condition are cirrhosis, congestive heart failure (CHF), and renal failure. In contrast, hypovolemic hyponatremia is associated with a reduction in both total body Na and water, but Na to a greater degree. This condition is encountered in patients with excessive fluid losses such as those with over‐diuresis, excessive gastrointestinal losses, burns, and pancreatitis.4

Classification of Hyponatremia: Sodium and Water Changes in the 3 Different Types of Hyponatremia
Depletional Hyponatreima Dilutional Hyponatremia
Euvolumic Hypervolumic
Total body water
Total body Na normal
Common etiologies SIADH cirrhosis/CHF vomiting, diarrhea

Hyponatremia is the most common electrolyte abnormality seen in general hospital patients.5 In a database of over 120,000 patients, a serum sodium level of <136mEq/L was observed in 28.2%.6 Hyponatremia is associated with selected medical conditions (especially cirrhosis and CHF), the extremes of age, and those receiving selected medications, including several that are commonly administered to cirrhotic patients (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors).7, 8 Hyponatremia is associated with increased total costs per hospital admission.5, 9 In an analysis of the effect of hyponatremia on length of stay in a retrospective cohort study of hospitalized patients derived from a large administrative database of 198,281 discharges from 39 US hospitals, mean length of stay was significantly greater among patients with hyponatremia than those with normal Na levels (8.6 8.0 vs. 7.2 8.2 days). After adjusting for confounders that may be associated with more severe disease and hyponatremia (age, gender, race, geographic region, teaching status of the hospital, admission source, principal payer, comorbidity index score and primary diagnosis), the presence of hyponatremia contributed an increase in length of stay of 1.0 day. Patients with hyponatremia are more frequently admitted to the intensive care unit (ICU) and require mechanical ventilation. In patients with CHF, the presence of hyponatremia at discharge is associated with increased risk for early mortality and rehospitalization.10

Although frequently asymptomatic, hyponatremia may be associated with a range of findings, from subtle and non‐specific complaints, including headache, fatigue, confusion, malaise, to severe and life‐threatening manifestations with lethargy, seizures, brainstem herniation, respiratory arrest and death.11 The most important complications are neurologic consequences related to cerebral edema. However, there is increased morbidity even in hyponatremic patients considered to be asymptomatic. Patients with low serum sodium have attention deficients, and falls are common. In a study of 122 patients who were considered to have chronic asymptomatic hyponatremia, the incidence of falls was significantly higher at 21.3% compared to only 5.3% in a control population.12

In hyponatremia, water enters into the cells to attain osmotic balance, resulting in cellular swelling.4 To avoid cerebral edema, the brain is capable of adapting to hyponatremia by regulating its volume to avoid swelling, especially when hyponatremia is chronic. In acute hyponatremia, astrocytes and neurons adapt through osmoregulatory mechanisms by extruding intracellular electrolytes such as potassium.13 Chronically, adaption occurs through the loss of low‐molecular weight organic compounds termed organic osmolytes including myoinsoitol, glutamine, choline and taurine. As a result, both the severity and the rate of its development are critical factors in determining the neurologic manifestation of hyponatremia in a given patient.14

Dilutional Hyponatremia and Cirrhosis

Patients with hyponatremia who are either euvolemic or hypervolemic are considered to have dilutional hyponatremia (DH). Management of these patients is distinct from those who are hypovolemic in whom appropriate therapy consists of the administration of normal saline. The remainder of this article addresses the pathogenesis, management and treatment of cirrhotic patients with DH.

Pathogenesis

The development of hyponatremia in cirrhosis is intimately related to the pathophysiology of portal hypertension and the non‐osmotic release of AVP3, 15 (Figure 1). In the early phases of cirrhosis, portal hypertension is the result of an increase in intrahepatic resistance. With the development of porto‐systemic collaterals, a hyperdynamic splanchnic circulation develops as a result of splanchnic arterial vasodilatation and increased vascular capacity. Nitric oxide, an endothelial derived relaxing factor, is the critical mediator of this process, and upregulation of its expression is pivotal in the pathogenesis of portal hypertension.

Figure 1
Proposed mechanism of hypersecretion and renal and systemic effects of vasopressin in cirrhosis with ascites. Gines P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management. Hepatology. 2008;48:1002–1010. Copyright 2008 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.

Multiple factors are related to the development of DH in cirrhosis. A reduction of effective central blood volume due to the development of porto‐venous collaterals and arterial splanchnic vasodilation, leading to baroreceptor‐mediated nonosmotic release of AVP, is considered the initiating and most important factor. Patients with cirrhosis and DH have higher plasma and urine vasopressin levels, higher plasma renin activity, and decreased plasma levels of atrial natriuretic factor than those with normal serum sodium concentrations, findings consistent with the presence of a decreased effective plasma volume.16 Arterial underfilling is sensed by baroreceptors located in the left ventricle, aortic arch, carotid sinus and renal afferent arterioles. Decreased activation leads to neurohumoral compensatory responses which include non‐osmotic release of vasopressin from the neurohypophysis and increased levels. Impaired catabolism of AVP that has been correlated with the severity of liver dysfunction may further contribute to increased levels.17 Initially, the increased AVP maintains arterial circulatory integrity by inducing splanchnic, peripheral and renal arterial vasoconstriction through its action on the V1a receptors and expansion of blood volume through renal water retention by its action on the V2 receptors located on the collecting ducts.

The initial adaptive response which leads to increased central blood volume can chronically result in detrimental effects, including the development of fluid overload with ascites, edema, and hyponatremia.16, 18 Additional factors that contribute to hyponatremia include decreased glomerular filtration rate (GFR) and/or increased proximal reabsorption of sodium (that reduce the distal delivery of filtrate and the potential for water reabsorption) and decreased cardiac function that further impairs effective central blood volume.19 In addition, urinary levels of AQP2 are increased in cirrhotic patients, especially those with decompensated disease with higher Child‐Pugh scores and ascites, and provide another potential mechanism to increase water reabsorption.20

Prevalence and Prognostic Significance

Hyponatremia in cirrhosis is a common finding. In a survey of 997 cirrhotic patients with ascites from 28 centers in Europe, North and South America, the prevalence of serum sodium concentration 135, 130, 125, 120 meq/L were 49.4%, 21.6%, 5.7%, and 1.2%, respectively.21 In a retrospective analysis of 188 inpatients, the prevalence of DH of 135, 130, and 125 were 20.8%, 14.9%, and 12.2%, respectively.22 The development of hyponatremia is a manifestation of increasing portal hypertension. In a natural history study of 263 patients hospitalized for first episode of significant ascites, 74 patients developed DH (Na level < 130 mEq/L), including 11 patients in whom it appeared during the first episode and 63 cases during follow‐up (mean period of 40 3 months) with a 5‐year incidence of 37.1%.23

The presence of hyponatremia carries significant adverse prognostic significance. It is strongly associated with severity of liver function impairment as assessed by Child‐Pugh and model for end‐stage liver disease (MELD) scores.22 Even mild hyponatremia is associated with severe complications such as massive ascites, severe hepatic encephalopathy, spontaneous bacterial peritonitis (SBP), and hepatic hydrothorax, and the severity of hyponatremia is directly related to the severity of these complications.21, 22 (Figure 2). In a natural history study of patients presenting with large volume ascites, 1‐year survival after its development was reduced to only 25.6%.230

Figure 2
Percentage of patients with complications of cirrhosis classified according to serum sodium concentration. Angeli P, Wong F, Watson H, et al. Hyponatremia in cirrhosis: results of a patient population survey. Hepatology. 2006;44:1535–1542. Copyright 2006 John Wiley & Sons, Inc. Reprinted with permission of John Wiley & Sons, Inc.
Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Hyponatremia is an especially poor prognostic sign for a hospitalized cirrhotic patient. In a retrospective analysis of 156 cirrhotic patients, hyponatremiapresent in 57 (29.8%) of admissionswas associated with increased hospital mortality (26.3% vs. 8.9% among those with normal Na levels), and the mortality rate was even higher (48%) among the 25 patients who developed severe hyponatremia during the hospital stay.24 In hospitalized patients, hyponatremia is predictive of the development of acute renal failure which is associated with substantially increased mortality (73% vs. 13%).25 Similarly, a low serum sodium level in critically ill cirrhotic patients admitted to the ICU is associated with complications, in‐hospital mortality, and poor short‐term prognosis.26

Whether hyponatremia should impact liver transplant prioritization remains an area of controversy. The United Network for Organ Sharing (UNOS) contracted by the Organ Procurement and Transplant Network (OPTN) to optimize the efficient use of deceased organs through fair and timely allocation, currently uses the MELD score, a formula that calculates the risk of death within three months from the bilirubin, creatinine, and International Normalized Ratio (INR) levels. Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction in patients with advanced cirrhosis and adds to MELD in predicting waitlist mortality.2729 In patients with a MELD score of <21, only low serum sodium and persistent ascites are independent predictors of mortality.28 To account for the importance of hyponatremia on survival, both modification of the MELD score in which the Na level is incorporated (MELD‐Na model) and the MELD to serum sodium ratio (MESO) have been developed. Adding hyponatremia to the MELD score is a better predictor of death than MELD alone, particularly in patients with low MELD scores.27, 2931 The OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has discussed updating the liver allocation system to include the Na level. However, it was concluded that implementation of MELD‐Na would change the allocation status of only 4% of candidates. Further, based on the concerns about the ability to manipulate serum sodium levels and the utility of employing resources to change the system for a relatively small number of patients, it was decided to defer incorporating the Na level pending further analysis (Report of the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee To the Board of Directors, Los Angeles, California, September 17‐18, 2007). At this time, the use of Na is a regional decision.32 However, the OPTN/UNOS Liver and Intestinal Organ Transplantation Committee has recently solicited feedback from the transplant community about including Na in allocation for review at a forum in April 2010.

Precipitating Factors

The most important factor related to development of hyponatremia in cirrhosis is increasing severity of portal hypertension that is associated with impaired central blood volume as a result of progressive splanchnic vasodilatation. In a study in which 170 patients with decompensated alcoholic cirrhosis were prospectively followed for 33.9 27.9 months, the initial hepatic venous pressure gradient (HVPG) was an independent predictive factor for the 20 patients who developed hyponatremia.22

Cirrhotic patients with ascites with hyponatremia have increased AVP secretion, higher levels of plasma renin activity, and higher serum concentrations of aldosterone and norepinephrine compared to those with normal Na levels.33 Diuretic therapy is associated with the development of DH by inducing volume depletion and arterial underfilling, further activating the renin‐angiotensin system (RAS) and increasing the non‐osmotic release of AVP.34 Although diuretics block the salt retention associated with the RAS activation, the water‐retaining effects of AVP persist, and DH develops. The process is further exacerbated by a low sodium intake and a frequent uncontrollable thirst. As a result, diuretic therapy is commonly associated with the development of hyponatremia in patients with ascites.24, 35 Similarly, paracentesis (particularly when performed without albumin) is often associated with an increase in blood urea nitrogen and marked elevations in plasma renin activity and plasma aldosterone concentration, which may exacerbate this physiology, leading to further reduction in serum sodium concentration.36 Tense ascites can contribute to DH by increasing baroreceptor mediated AVP release by increasing intrathoracic pressure.37 Finally, non‐steroidal anti‐inflammatory drugs (NSAIDs) can cause DH by inhibiting the synthesis of renal prostaglandins (which normally function to antagonize the tubular action of AVP and are important in the maintenance of appropriate renal tubular transport of fluid and electrolytes in states of renal hypoperfusion).38

Medical Impact of Hyponatremia: Marker of Severe Disease or Direct Pathophysiologic Role?

Hyponatremia is associated with severe ascites, impaired renal function, hepatic encephalopathy, SBP, and hepatorenal syndrome.3, 20 Because hyponatremia is frequently present in advanced liver failure, it is unclear whether it is only a marker of advanced disease or whether it plays a direct pathophysiologic role, or both. Until recently, it has not been possible to address this issue due to the inability to easily and rapidly correct the hyponatremia. However, there is increasing evidence that hyponatremia has direct impact on the severity of hepatic encephalopathy (see Hepatic Encephalopathy section). The recent introduction of tolvaptan for the treatment of hyponatremia in cirrhosis (discussed below) will allow this question to be directly answered.

Fluid Management and Diuresis

The typical cirrhotic patient with DH is characterized by expanded extracellular fluid with ascites and edema. The profound vasodilation of the splanchnic arterial circulation is associated with decreased effective arterial blood volume, leading to the non‐osmotic release of AVP. Diuretic therapy can further exacerbate this process. In addition, the increased water permeability induced by AVP results in reduced urine volume and fluid retention. As a result, hyponatremia directly adversely affects severity of fluid overload and limits and/or precludes diuretic treatment.

Hepatorenal Syndrome

Hyponatremia is an earlier and more sensitive marker than serum creatinine to detect renal impairment and/or circulatory dysfunction and is frequently a precursor to overt hepatorenal syndrome.27 Hyponatremia is predictive of the development of acute renal failure during hospitalization, and in‐hospital development of acute renal failure portends a high mortality.25 In patients admitted with SBP, the presence of hyponatremia is significantly associated with higher mortality and renal failure.39

Hepatic Encephalopathy

The neurologic manifestations of cerebral edema associated with hyponatremia closely mirror those of hepatic encephalopathy. In fact, a recently proposed pathogenic mechanism for hepatic encephalopathy is the development of low‐grade cerebral edema associated with astrocyte swelling in response to ammonia and other precipitating factors.40 DH is associated with a further reduction in brain organic osmolytes that probably reflects a compensatory osmoregulatory mechanism against cell swelling triggered by a combination of high intracellular glutamine and low extracellular osmolality.41 As a result, it has been proposed that hyponatremia contributes to the development of hepatic encephalopathy through the development or exacerbation of low‐grade cerebral edema. In this manner, low serum sodium acts as a second hit to the swelling produced by increased intracellular glutamine created by ammonia metabolism.42

Clinically, hyponatremia is a major risk factor for hepatic encephalopathy. Serum sodium and ammonia levels are the major factors that predict electroencephalographic abnormalities in cirrhotics who do not have hepatic encephalopathy.43 In a prospective study of 61 patients, hyponatremia was associated with a low brain concentration of organic osmolytes as assessed by proton magnetic resonance spectroscopy (1H‐MRS) and magnetic resonance imaging, and both conditions were major risk factors for the development of overt hepatic encephalopathy.44 Finally, hyponatremia is a risk factor for hepatic encephalopathy in patients undergoing TIPS.45

Adverse Effect on Outcome After Liver Transplantation

Hyponatremia before liver transplantation is associated with adverse post‐transplant outcomes. Among patients undergoing liver transplantation, the presence of hyponatremia is associated with abnormal cardiac response in patients after reperfusion.46 Pre‐transplant hyponatremia is associated with longer ICU and hospital stay, higher rates of delirium and neurologic disorders, acute renal failure, acute cellular rejection, infection, and in one study a reduced 3‐month survival compared to normonatremic recipients.32, 47, 48 In 1 retrospective study that compared post‐transplant outcomes of patients with corrected vs. uncorrected pre‐transplant hyponatremia, patients with pre‐operative correction of hyponatremia had a lower risk of prolonged post‐transplant hospitalization than those with uncorrected hyponatremia.32 However, both hyponatremic groups had more complicated post‐transplant courses compared to those without a history of hyponatremia. However, given the small sample size, retrospective design, and the potential for confounding, the impact of correction of pre‐transplant hyponatremia remains to be determined.

Management

Most patients with mild hypervolemic hyponatremia are asymptomatic. The initial recommended approach is fluid restriction and an Na‐restricted diet. For those with severe or progressive hyponatremia, diuretics should be minimized or discontinued to avoid intravascular volume depletion.49 For patients with tense ascites and severe DH, therapeutic paracentesis with plasma expanders is safe.33 Unfortunately, fluid restriction is limited in efficacy and often poorly tolerated. The use of hypertonic saline is generally not recommended unless severe neurologic symptoms are present as it leads to increased ascites and edema. When administered, it is important to avoid a rapid correction of the hyponatremia to prevent the development of central pontine myelinolysis and the osmotic demyelination syndrome.

Due to the pivotal role of AVP in the pathogenesis of DH, antagonism of its action has long been proposed to be the most rational approach, but until recently, effective and specific antagonism of AVP has remained elusive. Approaches that have been attempted include interference with its secretion and actions. Intravenous albumin has been reported to improve hyponatremia in patients with cirrhosis, ascites, and hyponatremia, presumably by decreasing AVP release by plasma volume expansion.50 An attempt at inhibition of central AVP release with the use of a kappa‐opioid receptor agonist, niravoline, was limited by loss of efficacy and potential adverse effects.51 Use of demeclocycline and lithium (which induce renal resistance to AVP and lead to a modest increase in urine volume with decreased urine osmolality and a corresponding rise in serum sodium) is limited by nephrotoxicity and hepatotoxicity.7, 52 Because of the important role played by prostaglandins in the maintenance of renal hemodynamics and water excretion in cirrhosis, oral misoprostol has also been evaluated but determined to be ineffective in inducing significant changes in free water clearance in patients with functional renal failure and/or DH.53

The recent introduction of vaptans, vasopressin receptor antagonists that block the physiologic action of vasopressin, represents a revolutionary and highly effective approach to the treatment of hyponatremia. Vaptans are antagonists of the V2 receptors of AVP in the principal cells of the collecting ducts. In healthy subjects, vaptans cause a dose‐dependent increase in urine volume and produce a dilute urine without causing natriuresis. To date, 2 AVP antagonists, conivaptan and tolvaptan, have been Food and Drug Administration (FDA)‐approved for the treatment of DH. Conivaptan, the first to be approved in 2005, is a mixed vasopressin V1a and V2 receptor antagonist that is administered intravenously for up to 4 days. In a randomized placebo‐controlled study of patients with euvolemic or hypervolemic hyponatremia, intravenous conivaptan treatment increased serum Na levels by >6 mEq/l or to a serum Na >135 mEq/l in 69 to 88.5% of subjects compared to 20.7% of those receiving placebo (Zeltser D, Rosansky S, Van Rensburg H, et al. Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia. American J Nephrology 2007;27:447457). In a pilot study involving 24 patients with end‐stage liver disease, an infusion of conivaptan over 1 to 4 days was associated with an increase of serum sodium by >5 mmol/L in 60% of patients not receiving diuretics and 67% of patients on concomitant diuretic therapy by the end of treatment (O'Leary and Davis, 2009). Despite a concern about the potential for conivaptan to increase portal hypertension due to inhibition of splanchnic V1a receptors, the brief treatment appeared to be well tolerated without significant changes in systolic blood pressure, serum creatinine, variceal bleeding or worsening of ascites during the infusion period. However, approval for only 4 days of therapy and requirement for intravenous use eliminate any potential for chronic use.54

Tolvaptan is an orally available, selective V2 receptor antagonist whose efficacy was assessed in two multicenter, prospective, randomized, placebo‐controlled trials, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2).55 In these trials, clinically stable patients with DH (Na <135meq/l) associated with cirrhosis (22.4% in SALT‐1, 30.5% in SALT‐2), CHF or SIADH were randomized in a hospital setting to receive tolvaptan 15mg daily or placebo. Repeat Na levels were obtained at 8 hours, 2, 3, and 4 days and then weekly at days 11, 18, 25 and 30 after which study drug was discontinued and follow‐up Na level was determined 7 days later. The dose was adjusted to 30 mg and then 60 mg in an attempt to achieve a Na level >135 in those in whom hyponatremia persisted. During the initial day of the titration phase, fluid restriction was not maintained, and the patients were encouraged to respond to thirst with increased water ingestion.

Tolvaptan use was associated with a prompt increase in Na level as early as 8 hours after administration of the first dose. Serum Na increased more among those receiving tolvaptan than among those receiving placebo during the first 4 days and throughout the study period regardless of baseline Na level but returned to baseline within 1 week after discontinuation (Figure 3). The main side effects were increased thirst, dry mouth and increased urination. Importantly, an increased incidence of renal failure was not observed. Based on these results, FDA approval for tolvaptan in patients with hyponatremia was obtained in May 2009 for patients with DH‐associated with cirrhosis, CHF or SIADH for patients with Na levels <125 or symptomatic patients with Na levels between 125 and 135 that have not responded to fluid restriction.

Management of the Hospitalized Cirrhotic Patient With Hyponatremia: Recommendations

Hyponatremia in hospitalized cirrhotic patients is a marker for severe disease and high risk of hospital mortality.24 As a result, prompt evaluation and treatment is imperative. The availability of tolvaptan potentially revolutionizes the manner in which these patients are treated. In the SALT trials, only clinically stable patients were enrolled. In this last section, a guideline for the evaluation and treatment of acutely ill, hospitalized cirrhotic patients with DH is presented.

Evaluation

Determination of volume status is paramount but frequently problematic in the hospitalized cirrhotic patient. Due to the vasodilated state present in severe portal hypertension that is characterized by a relative hypotension and resting tachycardia, the usual hemodynamic parameters of blood pressure and heart rate can be difficult to interpret. Although significant extravascular volume in the form of ascites and edema may be present, patients may be intravascularly depleted due to previous diuretic use and extra‐renal losses due to impaired oral intake, vomiting, lactulose‐induced diarrhea, and gastrointestinal bleeding. Infection is a commonly associated condition, and endotoxin mediated splanchnic vasodilatation, especially in the setting of SBP, can adversely effective central blood volume status in the presence of severe ascites. Also, due to the Na avidity of the kidney and previous diuretic use, renal electrolytes can be difficult to interpret.

For patients in whom there is strong clinical concern about intravascular depletion (history of impaired oral intake, excessive vomiting and/or diarrhea, rapid weight loss, small volume ascites with history of large volume, azotemia), administration of limited intravenous normal saline (0.5‐1 L) should be considered. Patients with severe neurological symptoms should receive normal saline or hypertonic saline. Unless severe neurologic symptoms associated with profound hyponatremia is present, however, intravenous normal saline should not be administered for the hyponatremia alone. Administration of salt poor albumin (25%), especially for those with marked fluid overload and ascites, is an effective means to expand the central blood volume without exacerbating ascites and edema. After evaluation for and/or treatment of hypovolemia, all patients should receive a Na restricted diet (<2000 mg daily) and placed on fluid restriction (see below for liberalization of fluid restriction upon initiation of tolvaptan therapy).

Diagnostic paracentesis should be performed for those with ascites to rule out the presence of SBP, and antibiotics administered to those with evidence of infection. High dose intravenous salt poor albumin should also be administered, especially to those at high risk of renal failure as determined by the presence of azotemia (Cr > 1.0 mg/dL) or severe liver insufficiency (TBili > 4.0 mg/dL).56, 57 Finally, all medications should be reviewed, and those associated with hyponatremia (diuretics, selective serotonin reuptake inhibitors, opiates, proton‐pump inhibitors) discontinued if possible.

Tolvaptan for DH

Patient Selection

Appropriate patient selection for tolvaptan therapy is extremely important (Table 2). In the SALT trials, only clinically stable patients were enrolled. The presence of hyponatremia in a recently hospitalized cirrhotic patient, however, frequently indicates severe disease with a high risk of acute renal failure and hospital death. In the SALT trials, many received concomitant diuretic therapy. Because of the importance of avoiding tolvaptan administration to hypovolemic patients, discontinuation of diuretic therapy prior the initiation of tolvaptan therapy and/or reevaluation after limited volume expansion should be considered.

Patient Selection for Tolvaptan Therapy for Hospitalized Patients With Cirrhosis and Hyponatremia
Hospital setting
Euvolumia or hypervolumia
Absence of recent weight loss, decrease in ascites, edema
Absence of excessive vomiting, diarrhea
Consider discontinuation of diuretic therapy prior to initiation of tolvaptan
Consider evaluation after limited volume expansion, especially with salt poor albumin prior to initiation of tolvaptan
Presence of clinically significant hyponatremia: 125mEq/L or less severe but symptomatic hyponatremia (125 to 134 mEq/L) that has resisted fluid restriction
Absence of severe neurologic symptoms attributable to hyponatremia
No co‐administration with intravenous saline
Ability to respond to thirst
No co‐administration with strong CYP 3A inhibitors (ketoconazole)
Absence of kidney failure with anuria

Tolvaptan is indicated for cirrhotic patients with DH in whom the serum sodium is <125 mEq/L and in those with less severe but symptomatic hyponatremia (125‐134 mEq/L) that has resisted fluid restriction. Although the definition of symptomatic was not specifically defined, possible considerations include symptoms of mild hepatic encephalopathy or inability to tolerate dieresis due to the presence of hyponatremia. According to FDA guidelines, tolvaptan therapy must be initiated and re‐initiated in a hospital setting. Patients with severe neurologic symptoms attributable to hyponatremia in whom rapid treatment is critical should not receive tolvaptan but should rather be treated with normal saline. Similarly, patients should not receive combination therapy with tolvaptan and normal saline due to potential for a too‐rapid correction of hyponatremia and the development of central pontine myelinolysis. If saline had been administered for treatment of possible hypovolemia, it should be discontinued and persistent hyponatremia confirmed before starting tolvaptan. Other factors that need to be considered before initiating tolvaptan include the ability of the patient to respond to thirst with increased water ingestion and recognition that the patient will experience increased urine volume and frequency, requiring easy access to toilet. Patients should not be fluid restricted during the first day of tolvaptan therapy, but should be instructed to respond to their thirst with increased water ingestion. As a result, caution should be exercised in administering tolvaptan to a confused, restrained, unresponsive and/or bed‐bound patient who is not able to respond appropriately to thirst or increased urination.

In the SALT trials, the incidence of hyperkalemia (5%) was similar in the tolvaptan and placebo treated patients.55 However, further analysis of all multiple‐dose, placebo‐controlled trials, demonstrated that the aggregate incidence of hyperkalemia was slightly higher for tolvaptan‐treated subjects compared with placebo‐treated subjects (Otsuka). Because treatment with tolvaptan is associated with an acute reduction of the extracellular fluid volume which could result in increased serum potassium through hemoconcentration, it is recommended that serum potassium levels be monitored after initiation of tolvaptan treatment in patients with a serum potassium > 5 mEq/L as well as those who are receiving drugs known to increase serum potassium levels such as angiotensin converting enzyme inhibitors, angiotensin receptor blockers, or potassium sparing diuretics (Samsca Package Insert, Otsuka). Because tolvaptan is metabolized by the cytochrome P 3A system, patients receiving strong inhibitors such as ketoconazole should not receive tolvaptan. Anuric patients will not respond to tolvaptan. Finally, it is extremely important to administer tolvaptan only to patients with true hyponatremia and not to those with pseudohyponatremia in whom the plasma osmolality is normal but the measured serum sodium concentration artificially low due to marked elevations of other substances, such as can be seen in severe hyperglycemia, marked hyperlipidemia, or hyperproteinemia (as in multiple myeloma).

Tolvaptan Administration

The initial dose of tolvaptan is 15 mg daily. After receiving tolvaptan, many patients will develop an increased sense of thirst and need to urinate. As a result, patients should not be fluid restricted during the first day of therapy, and it is important to monitor the hemodynamics and Na level closely after initiating therapy with a repeat Na level at approximately 8 hours after the first dose. As a result, it should probably be administered early in the day and not at bedtime. The dose should be increased to 30 mg, then 60 mg in patients who do not respond by at least 5 mEq/L over the previous 24 hours and remain hyponatremic. In those with an excessive response (more than 8 meq/L during the first 8 hours or 12 meq/L on any subsequent day), the patient should be encourage to either drink more water, or the dose should be held or reduced. After the appropriate dose has been identified, the patient may be discharged and continued on tolvaptan long‐term.

With the advent of this exciting therapy, practical issues will need to be addressed, most important of which is its cost at $250 per day (Otsuka). In addition, the current recommendation to initiate tolvaptan only in a hospital further limits its widespread use. Most important, long‐term clinical benefit will need to be demonstrated. Although the SALT trials only involved treatment for up to 1 month, a multicenter, open‐label extension study for a mean duration of 701 days demonstrated that prolonged administration of tolvaptan maintains an increased serum sodium level.58 However, at this time, tolvaptan can only be considered as one of the promising drugs whose long‐term cost‐effectiveness is yet to be proven. Proof will require showing that correction of the hyponatremia leads to improved clinical outcomes, such as a reduction in length of stay or frequency of hospitalization, decreased renal failure, improved hepatic encephalopathy, deceased mortality, and improved post‐transplant outcomes.

Unanswered Questions

The vaptans provide an important opportunity to clarify the role that hyponatremia plays in the pathogenesis of cirrhosis. In the past, DH in a cirrhotic patient represented a sign of advanced disease. With the availability of safe and effective therapy, we can now determine whether it also plays an important role in the pathophysiology of end‐stage liver disease and whether its treatment will have a beneficial effect on patient outcomes.

Specific clinical questions that will inevitably be addressed over the next few years to determine whether DH is only a marker for advanced disease or whether it plays a direct but modifiable role in the pathophysiology of cirrhosis will include:

  • Role of vaptans in the management of ascites: In a 14‐day randomized, trial of a satavaptan, another selective vasopressin V(2) receptor antagonist, vs. placebo with spironolactone, combination therapy was associated with improved control of ascites and improvements in serum sodium levels in hyponatremic patients with ascites.59 If future similar studies demonstrate more prolonged benefits, this would constitute an important advance in the treatment of ascites in cirrhosis.

  • Effect on renal function: Prolonged use of tolvaptan leads to a compensatory increase in endogenous levels of AVP and, potentially, increased stimulation of V1a receptors, which might be helpful in the setting of portal hypertension. In patients with hepatorenal syndrome, vasopressin stimulation of splanchnic V1a receptors leads to improved renal function, presumably by decreasing splanchnic blood flow and improving central blood volume.60 As a result, tolvaptan may indirectly improve kidney function in patients with advanced cirrhosis and refractory ascites. Whether long‐term tolvaptan therapy will help to prevent hepatorenal syndrome through this mechanism remains to be determined but is an exciting possibility.61

  • Effect on hepatic encephalopathy: Hepatic encephalopathy is associated with poor quality of life in patients with cirrhosis. Although hepatic encephalopathy was not directly assessed in the SALT trials, the mean mental component summary of the Short Form General Health Survey, a quality of life measure, improved in cirrhotic patients receiving tolvaptan to a greater degree that those receiving placebo.62 A possible explanation for this finding is a beneficial effect of tolvaptan on hepatic encephalopathy. Confirmation of this hypothesis, however, will require prospective studies in which hepatic encephalopathy is directly assessed.

  • Effect on medical economics: Based on retrospective reviews, hyponatremia has an adverse impact on length of stay and outcomes following liver transplantation. It will be important to demonstrate in prospective studies that correction of hyponatremia with tolvaptan reduces length of stay, complications, and costs.

References
  1. Nielsen S,Marples D,Frokiaer J, et al.The aquaporin family of water channels in kidney: an update on physiology and pathophysiology of aquaporin‐2.Kidney Int.1996;49:17181723.
  2. Gines P,Berl T,Bernardi M, et al.Hyponatremia in cirrhosis: from pathogenesis to treatment.Hepatology.1998;28:851864.
  3. Gines P,Guevara M.Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management.Hepatology.2008;48:10021010.
  4. Adrogue HJ,Madias NE.Hyponatremia.N Engl J Med.2000;342:15811589.
  5. Callahan MA,Do HT,Caplan DW,Yoon‐Flannery K.Economic impact of hyponatremia in hospitalized patients: a retrospective cohort study.Postgrad Med.2009;121:186191.
  6. Hawkins RC.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chim Acta.2003;337:169172.
  7. Adrogue HJ.Consequences of inadequate management of hyponatremia.Am J Nephrol.2005;25:240249.
  8. Liamis G,Milionis H,Elisaf M.A review of drug‐induced hyponatremia.Am J Kidney Dis.2008;52:144153.
  9. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  10. Upadhyay A,Jaber BL,Madias NE.Incidence and prevalence of hyponatremia.Am J Med.2006;119(7 Supple 1):S30S35.
  11. Bagshaw SM,Townsend DR,McDermid RC.Disorders of sodium and water balance in hospitalized patients.Can J Anesth.2009;56:151167.
  12. Renneboog B,Musch W,Vandemergel X, et al.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119;71.e1‐8.
  13. Sterns RH,Silver SM.Brain volume regulation in response to hypo‐osmolality and its correction.Am J Med.2006;119 (7 Suppl 1):S12S16.
  14. Arieff AI,Llach F,Massry SG.Neurological manifestations and morbidity of hyponatremia: correlation with brain water and electrolytes.Medicine.1976;55:121129.
  15. Iwakiri Y,Groszmann RJ.The hyperdynamic circulation of chronic liver diseases: from the patient to the molecule.Hepatology.2006;43:S121S131.
  16. Akriviadis EA,Ervin MG,Cominelli F, et al.Hyponatremia of cirrhosis: role of vasopressin and decreased “effective” plasma volume.Scand J Gastroenterol.1997;32:829834.
  17. Solis‐Herruzo JA,Gonzalez‐Gamarra A,Castellano G,Muñoz‐Yagüe MT.Metabolic clearance rate of arginine vasopressin in patients with cirrhosis.Hepatology.1992;16:974979.
  18. Schrier RW.Water and sodium retention in edematous disorders: role of vasopressin and aldosterone.Am J Med.2006;119:S47S53.
  19. Ruiz‐del‐Arbol L,Monescillo A,Arocena C, et al.Circulatory function and hepatorenal syndrome in cirrhosis.Hepatology.2005;42:439447.
  20. Chung SH,Jun DW,Kim KT, et al.Aquaporin‐2 urinary excretion in cirrhosis: relationship to vasopressin and nitric oxide.Dig Dis Sci.2010;55(4):11351141.
  21. Angeli P,Wong F,Watson H, et al.Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44:15351542.
  22. Kim WR et al.Hyponatremia and mortality among patients on the liver‐transplant list.N Engl J Med.2009;359:10181026.
  23. Planas R,Montoliu S,Balleste B, et al.Natural history of patients hospitalized for management of cirrhotic ascites.Clin Gastroenterol Hepatol.2006;4:13851394.
  24. Borroni G,Maggi A,Sangiovanni A, et al.Clinical relevance of hyponatremia for the hospital outcome of cirrhotic patieints.Dig Liver Dis.2000;32:605610.
  25. Wu CC,Yeung LK,Tsai WS, et al.Incidence and factors predictive of acute renal failure in patients with advanced liver cirrhosis.Clin Nephrol.2006;65:2833.
  26. Jenq CC,Tsai MH,Tian YC, et al.Serum sodium predicts prognosis in critically ill cirrhotic patients.J Clin Gastroenterol2010;44(3):220226.
  27. Ruf AE,Kremers WK,Chavez LL, et al.Addition of serum sodium into the MELD score predicts waiting list mortality better than MELD alone.Liver Transpl.2005;11:336343.
  28. Heuman DM,Abou‐Assi SG,Habib A, et al.Persistent ascites and low serum sodium identify patients with cirrhosis and low MELD scores who are at high risk for early death.Hepatology.2004;40:802810.
  29. Lv XH,Liu HB,Wang Y, et al.,Validation of model for end‐stage liver disease score to serum sodium ratio index as a prognostic predictor in patients with cirrhosis.J Gastroenterol Hepatol.2009;24:15471553.
  30. Kim MY,Liu HB,Wang Y, et al.Hepatic venous pressure gradient can predict the development of hepatocellular carcinoma and hyponatremia in decompensated alcoholic cirrhosis.Eur J Gastroenterol Hepatol.2009;21:12411246.
  31. Biggins SW,Rodriguez HJ,Bacchetti P, et al.Serum sodium predicts mortality in patients listed for liver transplantation.Hepatology.2005;41:3239.
  32. Hackworth WA,Heuman DM,Sanyal AJ, et al.Effect of hyponatremia on outcomes following orthotopic liver transplantation.Liver Int.2009;29:10711077.
  33. Vila MC,Coll S,Sola R, et al.Total paracentesis in cirrhotic patients with tense ascites and dilutional hyponatremia.Am J Gastroenterol.1999;94:22192223.
  34. Porcel A,Diaz F,Rendon P, et al.Dilutional hyponatremia in patients with cirrhosis and ascites.Arch Intern Med.2002;162:323328.
  35. Somberg JC,Molnar J.Therapeutic approaches to the treatment of edema and ascites: the use of diuretics.Am J Ther.2009;16:98101.
  36. Gines P,Tito L,Arroyo V, et al.Randomized comparative study of therapeutic paracentesis with and without intravenous albumin in cirrhosis.Gastroenterol.1988;94:14931502.
  37. Solis‐Herruzo JA,Moreno D,Gonzalez A, et al.Effect of intrathoracic pressure on plasma arginine vasopressin levels.Gastroenterology.1991;101:607617.
  38. Wen SF.Nephrotoxicities of nonsteroidal anti‐inflammatory drugs.J Formos Med Assoc.1997;96:157171.
  39. Terg R,Gadano A,Cartier M, et al.Serum creatinine and bilirubin predict renal failure and mortality in patients with spontaneous bacterial peritonitis: a retrospective study.Liver Int.2009;29:415419.
  40. Haussinger D,Schliess F.Pathogenic mechanisms of hepatic encephalopathy.Gut2008;57:11561165.
  41. Restuccia T,Gomez‐Anson B,Guevara M, et al.Effects of dilutional hyponatremia on brain organic osmolytes and water content in patients with cirrhosis.Hepatology.2004;39:16131622.
  42. Hausinger D.Low grade cerebral edema and the pathogenesis of hepatic encephalopathy in cirrhosis.Hepatology.2006;43:11871190.
  43. Amodio P,Del Piccolo F,Petteno E, et al.Prevalence and prognostic value of quantified electroencephalogram (EEG) alterations in cirrhotic patients.J Hepatol.2001;35:3745.
  44. Guevara M,Baccaro ME,Torre A, et al.Hyponatremia is a risk factor of hepatic encephalopathy in patients with cirrhosis: a propective study with time‐dependent analysis.Am J Gastroenterol.2009;104:13821389.
  45. Jalan R,Elton RA,Redhead DN, et al.Analysis of prognostic variables in the prediction of mortality, shunt failure, variceal rebleeding and encephalopathy following the transjugular intrahepatic portosystecim stent‐shunt for variceal haemorrhage.J Hepatol.1995;2:123128.
  46. Ripoll C,Catalina MV,Yotti R, et al.Cardiac dysfunction during liver transplantation: incidence and preoperative predictors.Transplantation.2008;85:17661772.
  47. Yun BC,Kim WR,Benson JT, et al.,Impact of pretransplant hyponatremia on outcome following liver transplantation.Hepatology.2009;49:16101615.
  48. Londono MC,Guevara M,Rimola A, et al.Hyponatremia impairs early posttransplantation outcome in patients with cirrhosis undergoing liver transplantation.Gastroenterology.2006;130:11351143.
  49. Martin‐Llahi M,Guevara M,Gines P.Hyponatremia in cirrhosis: clinical features and management.Gastroenterol Clin Biol.2006;30:11441151.
  50. McCormick PA,Mistry P,Kaye G, et al.Intravenous albumin infusion is an effective therapy for hyponatremia in cirrhotic patients with ascites.Gut.1990;31:204207.
  51. Bosch‐Marce M,Poo JL,Jiménez W, et al.Comparison of two aquaretic drugs (niravoline and OPC‐31260) in cirrhotic rats with ascites and water retention.J Pharmacol Exp Ther.1999;289:194201.
  52. Miller PD,Linas SL,Schrier RW.Plasma demeclocycline levels and nephrotoxicity. Correlation in hyponatremic cirrhotic patients.JAMA.1980;243:25132515.
  53. Gines A,Salmeron JM,Gines P, et al.Oral misoprostol or intravenous prostaglandin E2 do not improve renal function in patients with cirrhosis and ascites with hyponatremia and renal failure.J Hepatol.1993;17:220226.
  54. Fernandez‐Varo G,Ros J,Cejudo‐Martin P, et al.Effect of the V1a/V2‐AVP receptor antagonist, Conivaptan, on renal water metabolism and systemic hemodynamics in rats with cirrhosis and ascites.J Hepatol.2003;38:755761.
  55. Schrier RW,Gheorghiade M,Berl T, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  56. Sort P,Navasa M,Arroyo V, et al.Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis.N Engl J Med.1999;341:403409.
  57. Sigal SH,Stanca CM,Fernandez J, et al.Restricted use of albumin for spontaneous bacterial peritonitis.Gut.2007;56:597599.
  58. Berl T,Quittnat‐Pelletier F,Verbalis JG, et al.Oral tolvaptan is safe and effective in chronic hypyonatremia.J Am Soc Nephrol.2010;21(4):705712.
  59. Gines P,Wong F,Watson H, et al.Effects of satavaptan, a selective vasopressin V(2) receptor antagonist, on ascites and serum sodium in cirrhosis with hyponatremia: a randomized trial.Hepatology.2008;48:204213.
  60. Sanyal AJ,Boyer T,Garcia‐Tsao G, et al.A randomized, prospective, double‐blind, placebo‐controlled trial of terlipressin for type 1 hepatorenal syndrome.Gastroenterology.2008;134:13601368.
  61. Dixon MB,Lien YH.Tolvapten and its potential in the treatment of hyponatremia.Ther Clin Risk Manag.2008;4:114911455.
  62. Cardenas A,Gines P,Marotta P, et al.The effects of vasopressin V2 receptor antagonist in the management of patients with cirrhosis and hyponatremia. Safety and efficacy of oral tolvaptan in the SALT trials.Hepatology.2009;50S:467A.
  63. Decaux G,Mols P,Cauchie P., et al.Treatment of hyponatremic cirrhosis with ascites resistant to diuretics by urea.Nephron.1986;44:337343.
  64. Gadano A,Moreau R,Pessione F, et al.Aquaretic effects of niravoline, a kappa‐opioid agonist, in patients with cirrhosis.J Hepatol.2000;32:3842.
  65. Kim JH,Lee JS, et al.The association between the serum sodium level and the severity of complications in liver cirrhosis.Korean J Intern Med.2009;24:106112.
  66. Such J,Hillebrand DJ,Guamer C, et al.Nitric oxide in ascitic fluid is an independent predictor of the development of renal impairment in patients with cirrhosis and spontaneous bacterial peritonitis.Eur J Gastroenterol Hepatol.2004;16:571577.
References
  1. Nielsen S,Marples D,Frokiaer J, et al.The aquaporin family of water channels in kidney: an update on physiology and pathophysiology of aquaporin‐2.Kidney Int.1996;49:17181723.
  2. Gines P,Berl T,Bernardi M, et al.Hyponatremia in cirrhosis: from pathogenesis to treatment.Hepatology.1998;28:851864.
  3. Gines P,Guevara M.Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management.Hepatology.2008;48:10021010.
  4. Adrogue HJ,Madias NE.Hyponatremia.N Engl J Med.2000;342:15811589.
  5. Callahan MA,Do HT,Caplan DW,Yoon‐Flannery K.Economic impact of hyponatremia in hospitalized patients: a retrospective cohort study.Postgrad Med.2009;121:186191.
  6. Hawkins RC.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chim Acta.2003;337:169172.
  7. Adrogue HJ.Consequences of inadequate management of hyponatremia.Am J Nephrol.2005;25:240249.
  8. Liamis G,Milionis H,Elisaf M.A review of drug‐induced hyponatremia.Am J Kidney Dis.2008;52:144153.
  9. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  10. Upadhyay A,Jaber BL,Madias NE.Incidence and prevalence of hyponatremia.Am J Med.2006;119(7 Supple 1):S30S35.
  11. Bagshaw SM,Townsend DR,McDermid RC.Disorders of sodium and water balance in hospitalized patients.Can J Anesth.2009;56:151167.
  12. Renneboog B,Musch W,Vandemergel X, et al.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119;71.e1‐8.
  13. Sterns RH,Silver SM.Brain volume regulation in response to hypo‐osmolality and its correction.Am J Med.2006;119 (7 Suppl 1):S12S16.
  14. Arieff AI,Llach F,Massry SG.Neurological manifestations and morbidity of hyponatremia: correlation with brain water and electrolytes.Medicine.1976;55:121129.
  15. Iwakiri Y,Groszmann RJ.The hyperdynamic circulation of chronic liver diseases: from the patient to the molecule.Hepatology.2006;43:S121S131.
  16. Akriviadis EA,Ervin MG,Cominelli F, et al.Hyponatremia of cirrhosis: role of vasopressin and decreased “effective” plasma volume.Scand J Gastroenterol.1997;32:829834.
  17. Solis‐Herruzo JA,Gonzalez‐Gamarra A,Castellano G,Muñoz‐Yagüe MT.Metabolic clearance rate of arginine vasopressin in patients with cirrhosis.Hepatology.1992;16:974979.
  18. Schrier RW.Water and sodium retention in edematous disorders: role of vasopressin and aldosterone.Am J Med.2006;119:S47S53.
  19. Ruiz‐del‐Arbol L,Monescillo A,Arocena C, et al.Circulatory function and hepatorenal syndrome in cirrhosis.Hepatology.2005;42:439447.
  20. Chung SH,Jun DW,Kim KT, et al.Aquaporin‐2 urinary excretion in cirrhosis: relationship to vasopressin and nitric oxide.Dig Dis Sci.2010;55(4):11351141.
  21. Angeli P,Wong F,Watson H, et al.Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44:15351542.
  22. Kim WR et al.Hyponatremia and mortality among patients on the liver‐transplant list.N Engl J Med.2009;359:10181026.
  23. Planas R,Montoliu S,Balleste B, et al.Natural history of patients hospitalized for management of cirrhotic ascites.Clin Gastroenterol Hepatol.2006;4:13851394.
  24. Borroni G,Maggi A,Sangiovanni A, et al.Clinical relevance of hyponatremia for the hospital outcome of cirrhotic patieints.Dig Liver Dis.2000;32:605610.
  25. Wu CC,Yeung LK,Tsai WS, et al.Incidence and factors predictive of acute renal failure in patients with advanced liver cirrhosis.Clin Nephrol.2006;65:2833.
  26. Jenq CC,Tsai MH,Tian YC, et al.Serum sodium predicts prognosis in critically ill cirrhotic patients.J Clin Gastroenterol2010;44(3):220226.
  27. Ruf AE,Kremers WK,Chavez LL, et al.Addition of serum sodium into the MELD score predicts waiting list mortality better than MELD alone.Liver Transpl.2005;11:336343.
  28. Heuman DM,Abou‐Assi SG,Habib A, et al.Persistent ascites and low serum sodium identify patients with cirrhosis and low MELD scores who are at high risk for early death.Hepatology.2004;40:802810.
  29. Lv XH,Liu HB,Wang Y, et al.,Validation of model for end‐stage liver disease score to serum sodium ratio index as a prognostic predictor in patients with cirrhosis.J Gastroenterol Hepatol.2009;24:15471553.
  30. Kim MY,Liu HB,Wang Y, et al.Hepatic venous pressure gradient can predict the development of hepatocellular carcinoma and hyponatremia in decompensated alcoholic cirrhosis.Eur J Gastroenterol Hepatol.2009;21:12411246.
  31. Biggins SW,Rodriguez HJ,Bacchetti P, et al.Serum sodium predicts mortality in patients listed for liver transplantation.Hepatology.2005;41:3239.
  32. Hackworth WA,Heuman DM,Sanyal AJ, et al.Effect of hyponatremia on outcomes following orthotopic liver transplantation.Liver Int.2009;29:10711077.
  33. Vila MC,Coll S,Sola R, et al.Total paracentesis in cirrhotic patients with tense ascites and dilutional hyponatremia.Am J Gastroenterol.1999;94:22192223.
  34. Porcel A,Diaz F,Rendon P, et al.Dilutional hyponatremia in patients with cirrhosis and ascites.Arch Intern Med.2002;162:323328.
  35. Somberg JC,Molnar J.Therapeutic approaches to the treatment of edema and ascites: the use of diuretics.Am J Ther.2009;16:98101.
  36. Gines P,Tito L,Arroyo V, et al.Randomized comparative study of therapeutic paracentesis with and without intravenous albumin in cirrhosis.Gastroenterol.1988;94:14931502.
  37. Solis‐Herruzo JA,Moreno D,Gonzalez A, et al.Effect of intrathoracic pressure on plasma arginine vasopressin levels.Gastroenterology.1991;101:607617.
  38. Wen SF.Nephrotoxicities of nonsteroidal anti‐inflammatory drugs.J Formos Med Assoc.1997;96:157171.
  39. Terg R,Gadano A,Cartier M, et al.Serum creatinine and bilirubin predict renal failure and mortality in patients with spontaneous bacterial peritonitis: a retrospective study.Liver Int.2009;29:415419.
  40. Haussinger D,Schliess F.Pathogenic mechanisms of hepatic encephalopathy.Gut2008;57:11561165.
  41. Restuccia T,Gomez‐Anson B,Guevara M, et al.Effects of dilutional hyponatremia on brain organic osmolytes and water content in patients with cirrhosis.Hepatology.2004;39:16131622.
  42. Hausinger D.Low grade cerebral edema and the pathogenesis of hepatic encephalopathy in cirrhosis.Hepatology.2006;43:11871190.
  43. Amodio P,Del Piccolo F,Petteno E, et al.Prevalence and prognostic value of quantified electroencephalogram (EEG) alterations in cirrhotic patients.J Hepatol.2001;35:3745.
  44. Guevara M,Baccaro ME,Torre A, et al.Hyponatremia is a risk factor of hepatic encephalopathy in patients with cirrhosis: a propective study with time‐dependent analysis.Am J Gastroenterol.2009;104:13821389.
  45. Jalan R,Elton RA,Redhead DN, et al.Analysis of prognostic variables in the prediction of mortality, shunt failure, variceal rebleeding and encephalopathy following the transjugular intrahepatic portosystecim stent‐shunt for variceal haemorrhage.J Hepatol.1995;2:123128.
  46. Ripoll C,Catalina MV,Yotti R, et al.Cardiac dysfunction during liver transplantation: incidence and preoperative predictors.Transplantation.2008;85:17661772.
  47. Yun BC,Kim WR,Benson JT, et al.,Impact of pretransplant hyponatremia on outcome following liver transplantation.Hepatology.2009;49:16101615.
  48. Londono MC,Guevara M,Rimola A, et al.Hyponatremia impairs early posttransplantation outcome in patients with cirrhosis undergoing liver transplantation.Gastroenterology.2006;130:11351143.
  49. Martin‐Llahi M,Guevara M,Gines P.Hyponatremia in cirrhosis: clinical features and management.Gastroenterol Clin Biol.2006;30:11441151.
  50. McCormick PA,Mistry P,Kaye G, et al.Intravenous albumin infusion is an effective therapy for hyponatremia in cirrhotic patients with ascites.Gut.1990;31:204207.
  51. Bosch‐Marce M,Poo JL,Jiménez W, et al.Comparison of two aquaretic drugs (niravoline and OPC‐31260) in cirrhotic rats with ascites and water retention.J Pharmacol Exp Ther.1999;289:194201.
  52. Miller PD,Linas SL,Schrier RW.Plasma demeclocycline levels and nephrotoxicity. Correlation in hyponatremic cirrhotic patients.JAMA.1980;243:25132515.
  53. Gines A,Salmeron JM,Gines P, et al.Oral misoprostol or intravenous prostaglandin E2 do not improve renal function in patients with cirrhosis and ascites with hyponatremia and renal failure.J Hepatol.1993;17:220226.
  54. Fernandez‐Varo G,Ros J,Cejudo‐Martin P, et al.Effect of the V1a/V2‐AVP receptor antagonist, Conivaptan, on renal water metabolism and systemic hemodynamics in rats with cirrhosis and ascites.J Hepatol.2003;38:755761.
  55. Schrier RW,Gheorghiade M,Berl T, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  56. Sort P,Navasa M,Arroyo V, et al.Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis.N Engl J Med.1999;341:403409.
  57. Sigal SH,Stanca CM,Fernandez J, et al.Restricted use of albumin for spontaneous bacterial peritonitis.Gut.2007;56:597599.
  58. Berl T,Quittnat‐Pelletier F,Verbalis JG, et al.Oral tolvaptan is safe and effective in chronic hypyonatremia.J Am Soc Nephrol.2010;21(4):705712.
  59. Gines P,Wong F,Watson H, et al.Effects of satavaptan, a selective vasopressin V(2) receptor antagonist, on ascites and serum sodium in cirrhosis with hyponatremia: a randomized trial.Hepatology.2008;48:204213.
  60. Sanyal AJ,Boyer T,Garcia‐Tsao G, et al.A randomized, prospective, double‐blind, placebo‐controlled trial of terlipressin for type 1 hepatorenal syndrome.Gastroenterology.2008;134:13601368.
  61. Dixon MB,Lien YH.Tolvapten and its potential in the treatment of hyponatremia.Ther Clin Risk Manag.2008;4:114911455.
  62. Cardenas A,Gines P,Marotta P, et al.The effects of vasopressin V2 receptor antagonist in the management of patients with cirrhosis and hyponatremia. Safety and efficacy of oral tolvaptan in the SALT trials.Hepatology.2009;50S:467A.
  63. Decaux G,Mols P,Cauchie P., et al.Treatment of hyponatremic cirrhosis with ascites resistant to diuretics by urea.Nephron.1986;44:337343.
  64. Gadano A,Moreau R,Pessione F, et al.Aquaretic effects of niravoline, a kappa‐opioid agonist, in patients with cirrhosis.J Hepatol.2000;32:3842.
  65. Kim JH,Lee JS, et al.The association between the serum sodium level and the severity of complications in liver cirrhosis.Korean J Intern Med.2009;24:106112.
  66. Such J,Hillebrand DJ,Guamer C, et al.Nitric oxide in ascitic fluid is an independent predictor of the development of renal impairment in patients with cirrhosis and spontaneous bacterial peritonitis.Eur J Gastroenterol Hepatol.2004;16:571577.
Issue
Journal of Hospital Medicine - 5(3)
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S8-S17
Page Number
S8-S17
Article Type
Display Headline
Managing hyponatremia in cirrhosis
Display Headline
Managing hyponatremia in cirrhosis
Legacy Keywords
hyponatremia, common electrolyte disorder, cirrhosis, portal hypertension, cirrhosis, liver failure
Legacy Keywords
hyponatremia, common electrolyte disorder, cirrhosis, portal hypertension, cirrhosis, liver failure
Sections
Article Source
Copyright © 2010 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
530 First Avenue, HCC 3D, New York, NY 10016
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

Clinical Approach and Treatment of the Hyponatremic Patient

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Diagnostic approach and management of inpatient hyponatremia

Hyponatremia is one of the most common electrolyte abnormalities encountered in clinical practice. The frequency of the disorder varies according to definition and clinical setting but has been reported to be present in 28% of patients upon hospital admission and in 7% of patients attending an ambulatory community clinic.1 Increasing age, medications, various disease states, and administration of hypotonic fluids are among the known risk factors for the disorder.

The mortality rate in hyponatremic patients is approximately 3 times that of normonatremic hospitalized patients.25 Outcomes are particularly poor in those patients whose serum sodium (Na+) falls during a hospitalization. In 1 prospective study the mortality rate in patients with a normal serum Na+ concentration was 0.2% in comparison to a mortality rate of 11.2% and 25% in patients with a serum Na+ concentration <130 mEq/L and <120 mEq/L, respectively.2 In a recent retrospective cohort study of 10,899 hospitalized patients, the incidence of hyponatremia (<135 mmol/L) at admission was 5.5%.5 As compared to those with normonatremia, these patients were more likely to require intensive care and mechanical ventilation within 48 hours of hospitalization. In addition, hospital mortality, mean length of stay, and costs were significantly greater among patients with hyponatremia than those without.

The association with hyponatremia and adverse outcomes could be the direct result of hyponatremia, the comormidities that lead to the electrolyte derangement, or both. Whatever the mechanism, hyponatremia should not be viewed as an innocuous condition. Rather, clinicians should view this disorder with urgency and institute measures to prevent any further decline in the serum Na+ concentration and initiate appropriate therapy for its correction. This review will first briefly summarize the pathogenesis of hyponatremia and then discuss various disease states encountered in the hospital setting in which hyponatremia is frequently present.

Pathogenesis of Hyponatremia

Hyponatremia is generally associated with a hypoosmolar state and is a marker for a disturbance in water balance. Stated differently, all hyponatremia is dilutional. The approach to the patient with hyponatremia is outlined in Figure 1.

Figure 1
Approach to the hyponatremic patient (EABV, effective arterial blood volume, SIADH, syndrome of inappropriate antidiuretic hormone secretion).

Is the Hyponatremia Representative of a Hypoosmolar State?

There are 3 causes of hyponatremia in which it is not associated with a hypoosmolar state. The first of these is pseudohyponatremia which involves an abnormal measurement of the serum Na+. This occurs in patients with hyperglobulinemia or hypertriglyceridemia in whom plasma water relative to plasma solids is decreased in blood, leading to less Na+ in a given volume of blood. In general, this problem is becoming less prevalent as many laboratories are using Na+ electrodes without diluting the blood such that the Na+ measurement becomes independent of plasma water and nonaqueous contents.

A second cause of hyponatremia in the absence of a hypoosmolar state involves true hyponatremia but with elevations in the concentration of another osmole. Clinical examples include hyperglycemia as seen in uncontrolled diabetes or rarely hypertonic infusion of mannitol used in the treatment of cerebral edema. The accumulation of these effective osmoles creates an osmotic force causing water to move from the intracellular to the extracellular space thus diluting the serum Na+. For every 100 mg/dL rise in glucose or mannitol, the serum Na+ will quickly fall by 1.6 mEq/L. This increase in tonicity also stimulates thirst and arginine vasopressin (AVP) secretion, both of which contribute to further water retention. As a result the plasma osmolality and serum Na+ concentration will continue to fall. Once the plasma osmolality normalizes the serum Na+ will have decreased by 2.8 mEq/L for every 100 mg/dL rise in glucose.

The third cause of hyponatremia in the absence of a hypoosmolar state is the addition of an isosmotic (or near isosmotic) non‐Na+ containing fluid to the extracellular space. This situation typically occurs during a transurethral resection of the prostate or during laprascopic surgery when large amounts of a nonconducting flushing solution containing glycine or sorbitol are absorbed systemically.

Is the Kidney's Ability to Dilute the Urine Intact?

The presence of hypotonic hyponatremia implies that water intake exceeds the ability of the kidney to excrete water. In unusual circumstances, this can occur when the kidneys ability to excrete free water is intact. However, because a normal kidney can excrete 18 L of water per day, the presence of hyponatremia with normal renal water excretion implies the patient is drinking >20 L water/day. This condition is referred to as primary polydipsia. These patients should have a urine osmolality <100 mOsm/L. While primary polydipsia is a common condition which leads to polyuria and polydipsia, it is uncommon as a sole cause of hyponatremia.

Hyponatremia in association with a maximally dilute urine can also result from more moderate fluid intake combined with extremely limited solute intake, a condition often referred to as beer potomania syndrome. In normal subjects, daily solute excretion is usually in the range of 800 mOsm to 1000 mOsm per day. Since the limit of urinary dilution is approximately 50 mOsm/L, then maximum urine output can be calculated to be approximately 16 L to 20 L per day (1000 mOsm/L/50 mOsm/L = 20). Individuals who drink beer or who are ingesting a fad diet have very limited protein intake so that daily solute excretion may be as low as 200 mOsm per day. In this setting, water intake >4 L/day (200 mOsm/50 mOsm/L = 4 L) will exceed renal water excretion and cause hyponatremia. Urine osmolality will be maximally dilute in such a patient.

In the absence of primary polydipsia, hyponatremia is associated with decreased renal water excretion and a urine that is inappropriately concentrated. It is important to note that in the presence of hyponatremia urine should be maximally dilute and a urine osmolality higher than this (>100 mOsm/L) is inappropriate. An inappropriately concentrated urine implies a defect in renal water excretion.

Excretion of water by the kidney is dependent on three factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be normal so that the tubular fluid will be diluted. Lastly, AVP levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is probably most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. In many conditions, decreased delivery of filtrate to the tip of the loop of Henle also contributes.

What is the Volume Status of the Patient?

In patients with hypotonic hyponatremia with an inappropriately concentrated urine, one needs to define whether effective arterial volume is decreased. Most causes of hyponatremia result from a decrease in effective arterial volume which leads to both baroreceptor‐mediated stimulation of AVP secretion and decreased distal delivery of filtrate to the tip of the loop of Henle. If effective arterial volume is low, extracellular fluid (ECF) volume can be low in the volume‐depleted patient (hypovolemic hyponatremia) or can be high in the edematous patient (hypervolemic hyponatremia). If effective arterial volume is normal, one is dealing with the euvolemic causes of hyponatremia (isovolemic hyponatremia).

The clinical determination of effective arterial volume is usually straightforward. On physical examination the best index of effective arterial volume is the presence or absence of an orthostatic change in pulse and blood pressure. Urinary electrolytes are also extremely useful in the assessment of effective arterial volume. Patients with a low effective arterial volume will tend to have a low urinary Na+ and Cl concentration and low fractional excretions of Na+ and Cl in the urine. Patients with euvolemic hyponatremia will be in balance and will excrete Na+ and Cl at rates that reflect dietary intake of Na+ and Cl. Generally urinary Na+ and Cl is >20 mEq/L and fractional excretions of these electrolytes are >1%.

Plasma composition can also be used to assess effective arterial volume. The blood urea nitrogen (BUN) is particularly sensitive to effective arterial volume. In patients with a normal serum creatinine concentration, a high BUN suggests a low effective arterial volume and a low BUN suggests a high effective arterial volume. The plasma uric acid can also be used as a sensitive index of effective arterial volume. In comparing patients with the syndrome of inappropriate secretion of antidiuretic hormone (SIADH) and other causes of hyponatremia, patients with low effective arterial volume tend to have an elevated serum uric acid. The serum urate is low in patients with SIADH. This is due to the fact that these patients are volume expanded although it is clinically difficult to detect the degree of volume expansion.

Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)

In patients who are determined to be clinically euvolemic, a concentrated urine and high AVP levels are inappropriate. The most common cause of this condition is SIADH. This syndrome is generally associated with diseases of the central nervous system, pulmonary diseases, and neoplasms. These conditions lead to secretion of AVP which is inappropriate both from the standpoint of plasma osmolality and effective arterial volume. A number of other etiologies cause a condition of hypoosmolality associated with euvolemia and can mimic the syndrome of inappropriate AVP secretion. These include isolated glucocorticoid deficiency (normal mineralocorticoid activity), hypothyroidism, pain, nausea, acute psychosis, and a variety of drugs.

Clinical Conditions Associated With Hyponatremia in the Hospital Setting

Post‐Operative Hyponatremia

The postoperative patient is particularly prone to developing hyponatremia. AVP levels are increased for several days following surgical procedures due to baroreceptor and nonbaroreceptor‐mediated mechanisms. These patients typically have subtle or overt decreases in effective arterial blood volume due to prolonged preoperative fasting combined with intraoperative and postoperative blood loss and third spacing of fluid. In addition to these factors which unload baroreceptors, postoperative pain, stress, anxiety, nausea, and administration of morphine can further stimulate the release of AVP. In some instances nonsteroidal antiinflmmatory drugs are given which have the effect of augmenting the hydroosmotic actions of AVP.6 In this setting of compromised ability to excrete a water load, administration of hypotonic fluid can precipitate acute iatrogenic hyponatremia.

Postoperative hyponatremia has been a major problem in pediatric populations due to the widespread practice of using hypotonic fluids for maintenance therapy. This approach is based on guidelines developed 50 years ago which were based on calculations linking energy expenditures to water and electrolyte losses.7 More recently, several groups have argued that isotonic rather than hypotonic fluids should be the routine maintenance fluid in such patients.8, 9 The pediatric community has been slow to embrace this approach out of the concern that excessive administration of Na+ would increase the risk of hypernatremia. A systematic meta‐analysis of studies comparing isotonic and hypotonic fluids in hospitalized children found the odds of developing hyponatremia following hypotonic solutions was 17.2 times greater than with isotonic fluids.10 The concern that isotonic maintenance fluids carry a risk of hypernatremia was not supported in the review. Some studies actually reported a decrease in serum Na+ concentration, presumably due to the desalination phenomenon in which hypertonic urine is excreted in volume expanded subjects with persistent vasopressin secretion.

Endocrine Disorders

Glucocorticoid Deficiency

Patients with glucocorticoid deficiency develop hyponatremia. It is important to separate this condition from that of mineralocorticoid deficiency and combined mineralocorticoid‐glucocorticoid deficiency. In patients with mineralocorticoid deficiency, ECF volume and effective arterial volume are low. This leads to baroreceptor stimulation of AVP secretion and to decreased distal delivery of filtrate to the diluting segments of the nephron. In isolated glucocorticoid deficiency, the patients are euvolemic. In these patients, for any given level of low plasma osmolality, vasopressin levels are inappropriately elevated. The administration of hydrocortisone restores the relationship between vasopressin and plasma osmolality to normal.

While it is possible to develop isolated glucocorticoid deficiency with adrenal disease, most adrenal diseases cause loss of mineralocorticoid and glucocorticoid function. Glucocorticoid deficiency in the absence of mineralocorticoid deficiency is usually due to pituitary disease. In fact, severe hyponatremia may be the initial clue to the presence of previously unrecognized hypopituitarism. Insufficient adrenal secretion of glucocorticoids may also be a complication following the long term use of exogenous glucocorticoids.

In addition to high AVP levels, AVP‐independent mechanisms lead to increases in urinary osmolality in glucocorticoid deficiency. The nature of this AVP‐independent effect is likely multifactorial. First, glucocorticoid deficiency is associated with a diminished cardiac output and an impaired systemic vascular response to hypotension. These changes will lead to a slight decline in glomerular filtration rate and increased volume absorption in the proximal tubule and thin descending limb. As a result, distal delivery of filtrate to the diluting segment will be abnormally low in glucocorticoid deficiency. Second, mineralocorticoid and glucocorticoid deficiency have both been shown to result in increased expression of aquaporin 2 water channels in the collecting duct.11 This later effect will further limit maximal urinary dilution and contribute to net water retention.

These AVP‐independent mechanisms are consistent with the clinical observation that patients with diabetes insipidus appear to improve clinically when they develop coexistent anterior pituitary insufficiency, and treatment of these patients with glucocorticoids appears to worsen the diabetes insipidus. Thus, in the patient with diabetes, insipidus‐free water excretion will be extremely large. When simultaneous glucocorticoid deficiency develops, free‐water excretion decreases.

Hypothyroidism

Myxedema coma is the most severe form of hypothyroidism and is commonly associated with hyponatremia.12 In this setting, blood pressure can be low because of decreased intravascular volume and cardiovascular collapse. The hypotension can be refractory to vasopressor therapy in the absence of thyroid hormone therapy. Cardiac output and stroke volume are low. A defect in renal water excretion develops as a result of baroreceptor mediated increases in AVP along with decreased delivery of filtrate to the distal nephron. In hypothyroid rats, there is upregulation of aquaporin 2 water channels. In these animals, administration of a V2 receptor antagonist reverses the increased water channel expression and corrects the impaired response to an acute water load.11

While reduced cardiac output and blood pressure associated with severe hypothyroidism can provide a stimulatory effect for AVP release through a baroreceptor mediated mechanism, milder forms of hypothyroidism can be considered in the differential diagnosis of euvolemic hyponatremia. In this setting, impaired renal excretion of water is presumably due to increased release of AVP due to the absence of a tonic inhibitory effect of thyroid hormone in the central nervous system. The degree of hyponatremia in this setting is typically mild.

Heart Failure

Hyponatremia is a common complication of left‐sided heart failure and several studies have shown that it is an independent predictor of mortality.13, 14 A similar association between reduced survival and hyponatremia is present in advanced right‐sided heart failure in patients with pulmonary arterial hypertension.15 Patients with heart failure who are hyponatremic have higher circulating levels of neurohormones (catecholamines, renin, angiotensin II, aldosterone, and AVP) than normonatremic subjects and are more likely to have prerenal azotemia. In addition to being a marker for the extent of neurohumoral activation, hyponatremia may play a more direct role in adverse outcomes through maladaptive volume regulatory responses of cardiac myocytes and by direct effects of AVP on cardiac and coronary V1a receptors.

Heart failure is associated with arterial underfilling leading to arterial baroreceptor‐mediated activation of the neurohumoral axis. This underfilling is due to decreased cardiac output in low‐output heart failure and decreased systemic vascular resistance in high‐output heart failure. Activation of the sympathetic nervous system along with the renin‐angiotensin‐aldosterone system leads to renal salt retention while the increase in AVP is associated with water retention and hyponatremia.

Cirrhosis

Hyponatremia is a common electrolyte abnormality in patients with cirrhosis and occurs with a frequency that tends to parallel the severity of liver disease.16 Patients with a serum Na+ 130 mEq/L are more likely to have refractory ascites and require therapeutic paracentesis. Hepatic encephalopathy, hepatorenal syndrome, and spontaneous bacterial peritonitis are also more common in patients with a serum Na+ 130 mmol/L than in patients with a normal serum Na+ concentration. Hyponatremia increases morbidity and mortality from hepatic transplantation and is associated with osmotic demyelination in the postoperative period because of the large increase in Na+ concentration associated with the procedure.17

Hepatic cirrhosis is characterized by a decreased effective arterial blood volume and activation of neurohumoral effectors. Reduced effective circulating volume due to generalized and specifically to arterial splanchnic vasodilation leads to baroreceptor‐mediated nonosmotic stimulation of AVP release and an impaired ability to excrete electrolyte‐free water.18 Reduced Na+ delivery to the distal tubule because of a low glomerular filtration rate and increased proximal Na+ reabsorption adds to the susceptibility of cirrhotic patients to hyponatremia.

Hyponatremic‐Hypertensive Syndrome

The development of hyponatremia in patients with severe hypertension associated with renal artery stenosis has been called the hyponatremic‐hypertensive syndrome.19, 20 Patients with this syndrome present with a variety of signs and symptoms that include headache, confusion, postural dizziness, polyuria, polydipsia, and salt craving. In a retrospective review of 32 patients with this syndrome, most of the subjects were thin, elderly, women smokers who had atherosclerotic renal vascular disease.19 Biochemical abnormalities included not only hyponatremia, but hypokalemia and increased plasma renin activity. The mean serum Na+ concentration was 129.7 mmol/L (range 120‐135).

The precise mechanism of this syndrome is not known. Given the available data, angiotensin‐mediated thirst coupled with nonosmotic release of AVP provoked by angiotensin II and/or hypertensive encephalopathy are likely. Na+ depletion due to pressure natriuresis, and K+ depletion due to hyperaldosteronism are also likely to play a role in the pathogenesis of hyponatremia.

Pneumonia

An association between pneumonia and hyponatremia has been known for quite some time but has been poorly defined. Of the various etiologic agents, legionella is more commonly associated with hyponatremia as compared to other types of community acquired disease. As has been true for many disorders, hyponatremia is associated with longer hospital stays and hospital mortality, most likely reflecting the severity of the pneumonia rather than morbidity from the usually mild and asymptomatic hyponatremia.

Central Nervous System Disease

Hyponatremia is a frequent complication of central nervous system disease to include bacterial meningitis and traumatic brain injury. Potential mechanisms include the development of SIADH, cerebral salt wasting (CSW), or hypotonic fluid administration in the setting of impaired renal water excretion as in patients with low effective volume. Of these various causes, hyponatremia is frequently attributed to SIADH. As previously mentioned, this syndrome is characterized by hyponatremia in the setting of an inappropriately concentrated urine, increased urine Na+ concentration, and evidence of normal or slightly increased intravascular volume.

However there are patients with intracranial disease who develop hyponatremia with similar characteristics but differ in that there is clinical evidence of a contracted ECF volume. This form of hyponatremia is due to excessive renal Na+ excretion resulting from a centrally mediated process and is termed CSW. The onset of this disorder is typically seen within the first ten days following a neurosurgical procedure or after a definable event, such as a subarachnoid hemorrhage or stroke. CSW has also been described in other intracranial disorders, such as carcinomatous or infectious meningitis and metastatic carcinoma.21 The distinction between SIADH and CSW is of considerable clinical importance given the divergent nature of the treatments. Fluid restriction is the treatment of choice in SIADH, whereas the treatment of CSW comprises vigorous Na+ and volume replacement.

Treatment of Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, disorientation, restlessness and obtundation, and seizures. The principal danger of hyponatremia or hypernatremia relates to effects on central nervous system function due to changes in brain size (Figure 2).

Figure 2
Change in cell size with acute and chronic hyponatremia. Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. Rapid correction of chronic hyponatremia causes acute cellular shrinkage and can lead to osmotic demyelination.

Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. If the decrease in ECF osmolality is slow, there will be no measurable cell swelling. This pathophysiologic sequence correlates well with clinical observations. If hyponatremia is slow in onset, neurologic symptoms and permanent brain damage are unusual, even if the decreases in Na+ concentration and ECF osmolality are large. Conversely, if hyponatremia is rapid in onset, cerebral edema and significant CNS symptoms and signs can occur with lesser changes in serum Na+ concentration.

When treating a patient with hyponatremia, the Na+ concentration should be raised at the rate at which it fell. In a patient whose serum Na+ concentration has decreased rapidly (<48 hours), neurologic symptoms are frequently present and there is cerebral edema. In this setting there has not been sufficient time to remove osmoles from the brain and rapid return to normal ECF osmolality merely returns brain size to normal. In general, the development of hyponatremia in the outpatient setting is more commonly chronic in duration and should be corrected slowly. By contrast, hyponatremia of short duration is more likely to be encountered in hospitalized patients receiving intravenous free water. Use of ecstasy, exercise‐induced hyponatremia, or patients with primary polydipsia can also develop acute hyponatremia and if symptomatic may similarly require rapid correction. Raising the serum Na+ concentration by 4 mEq/L to 6 mEq/L over a several hour period is both safe and effective in preventing untoward effects of acute hyponatremia.22 A reasonable strategy to accomplish this goal is the regimen recommended for the treatment of athletes with hyponatremia and encephalopathy.23 These guidelines suggest an immediate bolus of 100 mL 3% NaCl (513 mEq/L). If there is no neurologic improvement two additional 100 mL 3% NaCl bolus infusions separated by 10 minute intervals can be given.

In patients with chronic hyponatremia (>48 hours duration) the serum Na+ concentration has fallen slowly. Neurologic symptoms are generally minimal, brain size is normal, and the number of intracellular osmoles is decreased. Sudden return of ECF osmolality to normal values will lead to cell shrinkage and possibly precipitate osmotic demyelination. Experts in the field suggest this complication can be prevented by adhering to the following limits of correction: 10 mmol/L in 24 hours, 18 mmol/L in 48 hours, and 20 mmol/L in 72 hours.22 In order to maximize patient safety, the goals of therapy should be more modest: 6 to 8 mmol/L in 24 hours, 12 to 14 mmol/L in 48 hours, and 14 to 16 mmol/L in 72 hours.

A formula designed to predict the increase in serum Na+ concentration to be expected from the infusion of 1 L of a given infusion is given below: 1.0 is indicative of no electrolyte‐free water in the urine and predicts a poor response unless water intake is severely limited.

Conservative Management of Chronic Hyponatremia
Treament Mechanism Advantages Limitations
  • Abbreviation: AVP,arginine vasopressin.

Fluid restriction (0.5‐1.0 L/day) water intake Effective, inexpensive Poor compliance
Demeclocyline (600‐1200 mg/day) Inhibits action of AVP Widely available Slow onset, nephrotoxic, expensive
Urea (30 gm/day) Osmotic diuresis Possiblerisk of osmotic demyelination Poor palatability, avoid in chronic kidney and liver disease
Lithium (up to 900 mg/daily) Inhibits action of AVP Widely available Slow onset, toxicity

When treating patients with SIADH with intravenous salt solutions, one can worsen the degree of hyponatremia if the osmolality of the administered fluid is less that the urine osmolality. One liter of isotonic saline contains 150 mEq each of Na+ and Cl and has an osmolality of 300 mOsm. In a patient with SIADH who has a urine osmolality of 600 mOsm, the administered NaCl will all be excreted in 500 mL of water (300 mOsm in 500 mL = 600 mOsm/L). The remaining 500 mL of administered fluid will be retained in the body and cause further dilution of the serum Na+ concentration. By contrast, 1 L of 3% saline contains 513 mEq each of Na+ and Cl and has an osmolality of 1026 mOsm. All of the NaCl in 1 L of this solution will be excreted in 1.7 L of water and therefore will cause the serum Na+ concentration to increase as a result of the loss of 700 mL of water.

Loop diuretics can also be employed as part of the strategy to treat chronic hyponatremia. Loop diuretics directly interfere in the countercurrent concentrating mechanism by inhibiting NaCl reabsorption in the loop of Henle. The coadministration of these drugs will enhance the effect of intravenous salt solutions by lowering urine osmolality thereby increasing the likelihood that the osmolality of the administered fluid is greater than the urine osmolality. Other measures used to treat chronic hyponatremia are given in Table 1.

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of water channels (aquaporin 2) into the apical membrane. The net effect is increased water excretion with little to no change in urinary Na+ excretion. AVP antagonists have been shown to be more effective than placebo in increasing the serum Na+ concentration in patients with modest asymptomatic hyponatremia.26, 27 These agents can be considered in patients with hyponatremia associated with heart failure or cirrhosis and in patients with irreversible SIADH but should not be used in patients with hypovolemic hyponatremia. The role of these drugs is discussed further in an accompanying article in this supplement, titled New horizons in the pharmacologic approach to hyponatremia: The V2 Receptor Antagonists.

References
  1. Hawkins R.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chem Acta.2003;337:169172.
  2. Anderson RJ,Chung HM,Kluge R,Schrier RW.Hyponatremia: a prospective analysis of its epidemiology and the pathogenetic role of vasopressin.Ann Intern Med.1985;102(2):164168.
  3. Gill G,Huda B,Boyd A, et al.Characteristics and mortality of severe hyponatraemia‐a hospital‐based study.Clin Endocrinol (Oxf).2006;65(2):246249.
  4. Waikar S,Mount D,Curhan G.Mortality after hospitalization with mild moderate and severe hyponatremia.Am J Med.2009;122:88578865.
  5. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  6. Palmer BF.Renal complications associated with use of nonsteroidal anti‐inflammatory agents.J Investig Med.1995;43(6):516533.
  7. Paut O,Lacroix F.Recent developments in the perioperative fluid management for the paediatric patient.Curr Opin Anaesthesiol.2006;19:268277.
  8. Moritz M,Ayus J.Prevention of hospital‐acquired hyponatremia: a case for using isotonic saline.Pediatrics.2003;111:227230.
  9. Hoorn EJ,Geary D,Robb M,Halperin Ml,Bohn D.Acute hyponatremia related to intravenous fluid administration in hospitalized children: an observational study.Pediatrics.2004;113(5):12791284.
  10. Choong K,Kho ME,Menon K,Bohn D.Hypotonic versus isotonic saline in hospitalized children: a systematic review.Arch Dis Child.2006;91(10):828835.
  11. Schrier R.Vasopressin and Aquaporin 2 in clinical disorders of water homeostasis.Semin Nephrol.2008;28:289296.
  12. Wartofsky L.Myxedema coma.Edocrinol Metab Clin N Am.2006;35:687698.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28(8):920921.
  14. Schrier R,Bansal S.Pulmonary hypertension, right ventricular failure, and kidney: different from left ventricular failure.Clin J Am Soc Nephrol.2008;3:12321237.
  15. Forfia PR,Mathai SC,Fisher MR, et al.Hyponatremia predicts right heart failure and poor survival in pulmonary arterial hypertension.Am J Respir Crit Care Med.2008;177(12):13641369.
  16. Angeli P,Wong F,Watson H,Ginès P;CAPPS investigators. Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44(6):15351542.
  17. Yu J,Zheng SS,Liang TB,Shen Y,Wang WL,Ke QH.Possible causes of central pontine myelinolysis after liver transplantation.World J Gastroenterol.2004;10(17):25402543.
  18. Palmer BF.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  19. Browne WL,Nair B.The hyponatremic hypertensive syndrome in renal artery stenosis: An infrequent cause of hyponatremia.J Postgrad Med.2007;53:4143.
  20. Agarwal M,Lynn K,Richards M,Nicholls M.Hyponatremic‐hypertensive syndrome with renal ischemia: an underrecognized disorder.Hypertension.1993;33:10201024.
  21. Palmer BF.Hyponatremia in patients with central nervous system disease: SIADH or CSW.Trends Endocrinol Metab.2003;14:182187.
  22. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  23. Hew‐Butler T,Ayus J,Kipps C, et al.Statement of the second international exercise‐associated hyponatremia consensus development conference, New Zealand.Clin J Sport Med.2008;18:111121.
  24. Mohmand H,Issa D,Ahmad Z,Cappuccio J,Kouides R,Sterns R.Hypertonic saline for hyponatremia: risk of inadvertent overcorrection.Clin J Am Soc Nephrol.2007;2:11101117.
  25. Perianayagam A,Sterns R,Silver S, et al.DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia.Clin J Am Soc Nephrol.2008;3:331336.
  26. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol2007;27:447457.
  27. Schrier R,Gross P,Gheorghiade M, et al.,Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
Article PDF
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S1-S7
Legacy Keywords
arginine vasopressin, hyponatremia, osmolality, osmotic demyelination
Sections
Article PDF
Article PDF

Hyponatremia is one of the most common electrolyte abnormalities encountered in clinical practice. The frequency of the disorder varies according to definition and clinical setting but has been reported to be present in 28% of patients upon hospital admission and in 7% of patients attending an ambulatory community clinic.1 Increasing age, medications, various disease states, and administration of hypotonic fluids are among the known risk factors for the disorder.

The mortality rate in hyponatremic patients is approximately 3 times that of normonatremic hospitalized patients.25 Outcomes are particularly poor in those patients whose serum sodium (Na+) falls during a hospitalization. In 1 prospective study the mortality rate in patients with a normal serum Na+ concentration was 0.2% in comparison to a mortality rate of 11.2% and 25% in patients with a serum Na+ concentration <130 mEq/L and <120 mEq/L, respectively.2 In a recent retrospective cohort study of 10,899 hospitalized patients, the incidence of hyponatremia (<135 mmol/L) at admission was 5.5%.5 As compared to those with normonatremia, these patients were more likely to require intensive care and mechanical ventilation within 48 hours of hospitalization. In addition, hospital mortality, mean length of stay, and costs were significantly greater among patients with hyponatremia than those without.

The association with hyponatremia and adverse outcomes could be the direct result of hyponatremia, the comormidities that lead to the electrolyte derangement, or both. Whatever the mechanism, hyponatremia should not be viewed as an innocuous condition. Rather, clinicians should view this disorder with urgency and institute measures to prevent any further decline in the serum Na+ concentration and initiate appropriate therapy for its correction. This review will first briefly summarize the pathogenesis of hyponatremia and then discuss various disease states encountered in the hospital setting in which hyponatremia is frequently present.

Pathogenesis of Hyponatremia

Hyponatremia is generally associated with a hypoosmolar state and is a marker for a disturbance in water balance. Stated differently, all hyponatremia is dilutional. The approach to the patient with hyponatremia is outlined in Figure 1.

Figure 1
Approach to the hyponatremic patient (EABV, effective arterial blood volume, SIADH, syndrome of inappropriate antidiuretic hormone secretion).

Is the Hyponatremia Representative of a Hypoosmolar State?

There are 3 causes of hyponatremia in which it is not associated with a hypoosmolar state. The first of these is pseudohyponatremia which involves an abnormal measurement of the serum Na+. This occurs in patients with hyperglobulinemia or hypertriglyceridemia in whom plasma water relative to plasma solids is decreased in blood, leading to less Na+ in a given volume of blood. In general, this problem is becoming less prevalent as many laboratories are using Na+ electrodes without diluting the blood such that the Na+ measurement becomes independent of plasma water and nonaqueous contents.

A second cause of hyponatremia in the absence of a hypoosmolar state involves true hyponatremia but with elevations in the concentration of another osmole. Clinical examples include hyperglycemia as seen in uncontrolled diabetes or rarely hypertonic infusion of mannitol used in the treatment of cerebral edema. The accumulation of these effective osmoles creates an osmotic force causing water to move from the intracellular to the extracellular space thus diluting the serum Na+. For every 100 mg/dL rise in glucose or mannitol, the serum Na+ will quickly fall by 1.6 mEq/L. This increase in tonicity also stimulates thirst and arginine vasopressin (AVP) secretion, both of which contribute to further water retention. As a result the plasma osmolality and serum Na+ concentration will continue to fall. Once the plasma osmolality normalizes the serum Na+ will have decreased by 2.8 mEq/L for every 100 mg/dL rise in glucose.

The third cause of hyponatremia in the absence of a hypoosmolar state is the addition of an isosmotic (or near isosmotic) non‐Na+ containing fluid to the extracellular space. This situation typically occurs during a transurethral resection of the prostate or during laprascopic surgery when large amounts of a nonconducting flushing solution containing glycine or sorbitol are absorbed systemically.

Is the Kidney's Ability to Dilute the Urine Intact?

The presence of hypotonic hyponatremia implies that water intake exceeds the ability of the kidney to excrete water. In unusual circumstances, this can occur when the kidneys ability to excrete free water is intact. However, because a normal kidney can excrete 18 L of water per day, the presence of hyponatremia with normal renal water excretion implies the patient is drinking >20 L water/day. This condition is referred to as primary polydipsia. These patients should have a urine osmolality <100 mOsm/L. While primary polydipsia is a common condition which leads to polyuria and polydipsia, it is uncommon as a sole cause of hyponatremia.

Hyponatremia in association with a maximally dilute urine can also result from more moderate fluid intake combined with extremely limited solute intake, a condition often referred to as beer potomania syndrome. In normal subjects, daily solute excretion is usually in the range of 800 mOsm to 1000 mOsm per day. Since the limit of urinary dilution is approximately 50 mOsm/L, then maximum urine output can be calculated to be approximately 16 L to 20 L per day (1000 mOsm/L/50 mOsm/L = 20). Individuals who drink beer or who are ingesting a fad diet have very limited protein intake so that daily solute excretion may be as low as 200 mOsm per day. In this setting, water intake >4 L/day (200 mOsm/50 mOsm/L = 4 L) will exceed renal water excretion and cause hyponatremia. Urine osmolality will be maximally dilute in such a patient.

In the absence of primary polydipsia, hyponatremia is associated with decreased renal water excretion and a urine that is inappropriately concentrated. It is important to note that in the presence of hyponatremia urine should be maximally dilute and a urine osmolality higher than this (>100 mOsm/L) is inappropriate. An inappropriately concentrated urine implies a defect in renal water excretion.

Excretion of water by the kidney is dependent on three factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be normal so that the tubular fluid will be diluted. Lastly, AVP levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is probably most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. In many conditions, decreased delivery of filtrate to the tip of the loop of Henle also contributes.

What is the Volume Status of the Patient?

In patients with hypotonic hyponatremia with an inappropriately concentrated urine, one needs to define whether effective arterial volume is decreased. Most causes of hyponatremia result from a decrease in effective arterial volume which leads to both baroreceptor‐mediated stimulation of AVP secretion and decreased distal delivery of filtrate to the tip of the loop of Henle. If effective arterial volume is low, extracellular fluid (ECF) volume can be low in the volume‐depleted patient (hypovolemic hyponatremia) or can be high in the edematous patient (hypervolemic hyponatremia). If effective arterial volume is normal, one is dealing with the euvolemic causes of hyponatremia (isovolemic hyponatremia).

The clinical determination of effective arterial volume is usually straightforward. On physical examination the best index of effective arterial volume is the presence or absence of an orthostatic change in pulse and blood pressure. Urinary electrolytes are also extremely useful in the assessment of effective arterial volume. Patients with a low effective arterial volume will tend to have a low urinary Na+ and Cl concentration and low fractional excretions of Na+ and Cl in the urine. Patients with euvolemic hyponatremia will be in balance and will excrete Na+ and Cl at rates that reflect dietary intake of Na+ and Cl. Generally urinary Na+ and Cl is >20 mEq/L and fractional excretions of these electrolytes are >1%.

Plasma composition can also be used to assess effective arterial volume. The blood urea nitrogen (BUN) is particularly sensitive to effective arterial volume. In patients with a normal serum creatinine concentration, a high BUN suggests a low effective arterial volume and a low BUN suggests a high effective arterial volume. The plasma uric acid can also be used as a sensitive index of effective arterial volume. In comparing patients with the syndrome of inappropriate secretion of antidiuretic hormone (SIADH) and other causes of hyponatremia, patients with low effective arterial volume tend to have an elevated serum uric acid. The serum urate is low in patients with SIADH. This is due to the fact that these patients are volume expanded although it is clinically difficult to detect the degree of volume expansion.

Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)

In patients who are determined to be clinically euvolemic, a concentrated urine and high AVP levels are inappropriate. The most common cause of this condition is SIADH. This syndrome is generally associated with diseases of the central nervous system, pulmonary diseases, and neoplasms. These conditions lead to secretion of AVP which is inappropriate both from the standpoint of plasma osmolality and effective arterial volume. A number of other etiologies cause a condition of hypoosmolality associated with euvolemia and can mimic the syndrome of inappropriate AVP secretion. These include isolated glucocorticoid deficiency (normal mineralocorticoid activity), hypothyroidism, pain, nausea, acute psychosis, and a variety of drugs.

Clinical Conditions Associated With Hyponatremia in the Hospital Setting

Post‐Operative Hyponatremia

The postoperative patient is particularly prone to developing hyponatremia. AVP levels are increased for several days following surgical procedures due to baroreceptor and nonbaroreceptor‐mediated mechanisms. These patients typically have subtle or overt decreases in effective arterial blood volume due to prolonged preoperative fasting combined with intraoperative and postoperative blood loss and third spacing of fluid. In addition to these factors which unload baroreceptors, postoperative pain, stress, anxiety, nausea, and administration of morphine can further stimulate the release of AVP. In some instances nonsteroidal antiinflmmatory drugs are given which have the effect of augmenting the hydroosmotic actions of AVP.6 In this setting of compromised ability to excrete a water load, administration of hypotonic fluid can precipitate acute iatrogenic hyponatremia.

Postoperative hyponatremia has been a major problem in pediatric populations due to the widespread practice of using hypotonic fluids for maintenance therapy. This approach is based on guidelines developed 50 years ago which were based on calculations linking energy expenditures to water and electrolyte losses.7 More recently, several groups have argued that isotonic rather than hypotonic fluids should be the routine maintenance fluid in such patients.8, 9 The pediatric community has been slow to embrace this approach out of the concern that excessive administration of Na+ would increase the risk of hypernatremia. A systematic meta‐analysis of studies comparing isotonic and hypotonic fluids in hospitalized children found the odds of developing hyponatremia following hypotonic solutions was 17.2 times greater than with isotonic fluids.10 The concern that isotonic maintenance fluids carry a risk of hypernatremia was not supported in the review. Some studies actually reported a decrease in serum Na+ concentration, presumably due to the desalination phenomenon in which hypertonic urine is excreted in volume expanded subjects with persistent vasopressin secretion.

Endocrine Disorders

Glucocorticoid Deficiency

Patients with glucocorticoid deficiency develop hyponatremia. It is important to separate this condition from that of mineralocorticoid deficiency and combined mineralocorticoid‐glucocorticoid deficiency. In patients with mineralocorticoid deficiency, ECF volume and effective arterial volume are low. This leads to baroreceptor stimulation of AVP secretion and to decreased distal delivery of filtrate to the diluting segments of the nephron. In isolated glucocorticoid deficiency, the patients are euvolemic. In these patients, for any given level of low plasma osmolality, vasopressin levels are inappropriately elevated. The administration of hydrocortisone restores the relationship between vasopressin and plasma osmolality to normal.

While it is possible to develop isolated glucocorticoid deficiency with adrenal disease, most adrenal diseases cause loss of mineralocorticoid and glucocorticoid function. Glucocorticoid deficiency in the absence of mineralocorticoid deficiency is usually due to pituitary disease. In fact, severe hyponatremia may be the initial clue to the presence of previously unrecognized hypopituitarism. Insufficient adrenal secretion of glucocorticoids may also be a complication following the long term use of exogenous glucocorticoids.

In addition to high AVP levels, AVP‐independent mechanisms lead to increases in urinary osmolality in glucocorticoid deficiency. The nature of this AVP‐independent effect is likely multifactorial. First, glucocorticoid deficiency is associated with a diminished cardiac output and an impaired systemic vascular response to hypotension. These changes will lead to a slight decline in glomerular filtration rate and increased volume absorption in the proximal tubule and thin descending limb. As a result, distal delivery of filtrate to the diluting segment will be abnormally low in glucocorticoid deficiency. Second, mineralocorticoid and glucocorticoid deficiency have both been shown to result in increased expression of aquaporin 2 water channels in the collecting duct.11 This later effect will further limit maximal urinary dilution and contribute to net water retention.

These AVP‐independent mechanisms are consistent with the clinical observation that patients with diabetes insipidus appear to improve clinically when they develop coexistent anterior pituitary insufficiency, and treatment of these patients with glucocorticoids appears to worsen the diabetes insipidus. Thus, in the patient with diabetes, insipidus‐free water excretion will be extremely large. When simultaneous glucocorticoid deficiency develops, free‐water excretion decreases.

Hypothyroidism

Myxedema coma is the most severe form of hypothyroidism and is commonly associated with hyponatremia.12 In this setting, blood pressure can be low because of decreased intravascular volume and cardiovascular collapse. The hypotension can be refractory to vasopressor therapy in the absence of thyroid hormone therapy. Cardiac output and stroke volume are low. A defect in renal water excretion develops as a result of baroreceptor mediated increases in AVP along with decreased delivery of filtrate to the distal nephron. In hypothyroid rats, there is upregulation of aquaporin 2 water channels. In these animals, administration of a V2 receptor antagonist reverses the increased water channel expression and corrects the impaired response to an acute water load.11

While reduced cardiac output and blood pressure associated with severe hypothyroidism can provide a stimulatory effect for AVP release through a baroreceptor mediated mechanism, milder forms of hypothyroidism can be considered in the differential diagnosis of euvolemic hyponatremia. In this setting, impaired renal excretion of water is presumably due to increased release of AVP due to the absence of a tonic inhibitory effect of thyroid hormone in the central nervous system. The degree of hyponatremia in this setting is typically mild.

Heart Failure

Hyponatremia is a common complication of left‐sided heart failure and several studies have shown that it is an independent predictor of mortality.13, 14 A similar association between reduced survival and hyponatremia is present in advanced right‐sided heart failure in patients with pulmonary arterial hypertension.15 Patients with heart failure who are hyponatremic have higher circulating levels of neurohormones (catecholamines, renin, angiotensin II, aldosterone, and AVP) than normonatremic subjects and are more likely to have prerenal azotemia. In addition to being a marker for the extent of neurohumoral activation, hyponatremia may play a more direct role in adverse outcomes through maladaptive volume regulatory responses of cardiac myocytes and by direct effects of AVP on cardiac and coronary V1a receptors.

Heart failure is associated with arterial underfilling leading to arterial baroreceptor‐mediated activation of the neurohumoral axis. This underfilling is due to decreased cardiac output in low‐output heart failure and decreased systemic vascular resistance in high‐output heart failure. Activation of the sympathetic nervous system along with the renin‐angiotensin‐aldosterone system leads to renal salt retention while the increase in AVP is associated with water retention and hyponatremia.

Cirrhosis

Hyponatremia is a common electrolyte abnormality in patients with cirrhosis and occurs with a frequency that tends to parallel the severity of liver disease.16 Patients with a serum Na+ 130 mEq/L are more likely to have refractory ascites and require therapeutic paracentesis. Hepatic encephalopathy, hepatorenal syndrome, and spontaneous bacterial peritonitis are also more common in patients with a serum Na+ 130 mmol/L than in patients with a normal serum Na+ concentration. Hyponatremia increases morbidity and mortality from hepatic transplantation and is associated with osmotic demyelination in the postoperative period because of the large increase in Na+ concentration associated with the procedure.17

Hepatic cirrhosis is characterized by a decreased effective arterial blood volume and activation of neurohumoral effectors. Reduced effective circulating volume due to generalized and specifically to arterial splanchnic vasodilation leads to baroreceptor‐mediated nonosmotic stimulation of AVP release and an impaired ability to excrete electrolyte‐free water.18 Reduced Na+ delivery to the distal tubule because of a low glomerular filtration rate and increased proximal Na+ reabsorption adds to the susceptibility of cirrhotic patients to hyponatremia.

Hyponatremic‐Hypertensive Syndrome

The development of hyponatremia in patients with severe hypertension associated with renal artery stenosis has been called the hyponatremic‐hypertensive syndrome.19, 20 Patients with this syndrome present with a variety of signs and symptoms that include headache, confusion, postural dizziness, polyuria, polydipsia, and salt craving. In a retrospective review of 32 patients with this syndrome, most of the subjects were thin, elderly, women smokers who had atherosclerotic renal vascular disease.19 Biochemical abnormalities included not only hyponatremia, but hypokalemia and increased plasma renin activity. The mean serum Na+ concentration was 129.7 mmol/L (range 120‐135).

The precise mechanism of this syndrome is not known. Given the available data, angiotensin‐mediated thirst coupled with nonosmotic release of AVP provoked by angiotensin II and/or hypertensive encephalopathy are likely. Na+ depletion due to pressure natriuresis, and K+ depletion due to hyperaldosteronism are also likely to play a role in the pathogenesis of hyponatremia.

Pneumonia

An association between pneumonia and hyponatremia has been known for quite some time but has been poorly defined. Of the various etiologic agents, legionella is more commonly associated with hyponatremia as compared to other types of community acquired disease. As has been true for many disorders, hyponatremia is associated with longer hospital stays and hospital mortality, most likely reflecting the severity of the pneumonia rather than morbidity from the usually mild and asymptomatic hyponatremia.

Central Nervous System Disease

Hyponatremia is a frequent complication of central nervous system disease to include bacterial meningitis and traumatic brain injury. Potential mechanisms include the development of SIADH, cerebral salt wasting (CSW), or hypotonic fluid administration in the setting of impaired renal water excretion as in patients with low effective volume. Of these various causes, hyponatremia is frequently attributed to SIADH. As previously mentioned, this syndrome is characterized by hyponatremia in the setting of an inappropriately concentrated urine, increased urine Na+ concentration, and evidence of normal or slightly increased intravascular volume.

However there are patients with intracranial disease who develop hyponatremia with similar characteristics but differ in that there is clinical evidence of a contracted ECF volume. This form of hyponatremia is due to excessive renal Na+ excretion resulting from a centrally mediated process and is termed CSW. The onset of this disorder is typically seen within the first ten days following a neurosurgical procedure or after a definable event, such as a subarachnoid hemorrhage or stroke. CSW has also been described in other intracranial disorders, such as carcinomatous or infectious meningitis and metastatic carcinoma.21 The distinction between SIADH and CSW is of considerable clinical importance given the divergent nature of the treatments. Fluid restriction is the treatment of choice in SIADH, whereas the treatment of CSW comprises vigorous Na+ and volume replacement.

Treatment of Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, disorientation, restlessness and obtundation, and seizures. The principal danger of hyponatremia or hypernatremia relates to effects on central nervous system function due to changes in brain size (Figure 2).

Figure 2
Change in cell size with acute and chronic hyponatremia. Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. Rapid correction of chronic hyponatremia causes acute cellular shrinkage and can lead to osmotic demyelination.

Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. If the decrease in ECF osmolality is slow, there will be no measurable cell swelling. This pathophysiologic sequence correlates well with clinical observations. If hyponatremia is slow in onset, neurologic symptoms and permanent brain damage are unusual, even if the decreases in Na+ concentration and ECF osmolality are large. Conversely, if hyponatremia is rapid in onset, cerebral edema and significant CNS symptoms and signs can occur with lesser changes in serum Na+ concentration.

When treating a patient with hyponatremia, the Na+ concentration should be raised at the rate at which it fell. In a patient whose serum Na+ concentration has decreased rapidly (<48 hours), neurologic symptoms are frequently present and there is cerebral edema. In this setting there has not been sufficient time to remove osmoles from the brain and rapid return to normal ECF osmolality merely returns brain size to normal. In general, the development of hyponatremia in the outpatient setting is more commonly chronic in duration and should be corrected slowly. By contrast, hyponatremia of short duration is more likely to be encountered in hospitalized patients receiving intravenous free water. Use of ecstasy, exercise‐induced hyponatremia, or patients with primary polydipsia can also develop acute hyponatremia and if symptomatic may similarly require rapid correction. Raising the serum Na+ concentration by 4 mEq/L to 6 mEq/L over a several hour period is both safe and effective in preventing untoward effects of acute hyponatremia.22 A reasonable strategy to accomplish this goal is the regimen recommended for the treatment of athletes with hyponatremia and encephalopathy.23 These guidelines suggest an immediate bolus of 100 mL 3% NaCl (513 mEq/L). If there is no neurologic improvement two additional 100 mL 3% NaCl bolus infusions separated by 10 minute intervals can be given.

In patients with chronic hyponatremia (>48 hours duration) the serum Na+ concentration has fallen slowly. Neurologic symptoms are generally minimal, brain size is normal, and the number of intracellular osmoles is decreased. Sudden return of ECF osmolality to normal values will lead to cell shrinkage and possibly precipitate osmotic demyelination. Experts in the field suggest this complication can be prevented by adhering to the following limits of correction: 10 mmol/L in 24 hours, 18 mmol/L in 48 hours, and 20 mmol/L in 72 hours.22 In order to maximize patient safety, the goals of therapy should be more modest: 6 to 8 mmol/L in 24 hours, 12 to 14 mmol/L in 48 hours, and 14 to 16 mmol/L in 72 hours.

A formula designed to predict the increase in serum Na+ concentration to be expected from the infusion of 1 L of a given infusion is given below: 1.0 is indicative of no electrolyte‐free water in the urine and predicts a poor response unless water intake is severely limited.

Conservative Management of Chronic Hyponatremia
Treament Mechanism Advantages Limitations
  • Abbreviation: AVP,arginine vasopressin.

Fluid restriction (0.5‐1.0 L/day) water intake Effective, inexpensive Poor compliance
Demeclocyline (600‐1200 mg/day) Inhibits action of AVP Widely available Slow onset, nephrotoxic, expensive
Urea (30 gm/day) Osmotic diuresis Possiblerisk of osmotic demyelination Poor palatability, avoid in chronic kidney and liver disease
Lithium (up to 900 mg/daily) Inhibits action of AVP Widely available Slow onset, toxicity

When treating patients with SIADH with intravenous salt solutions, one can worsen the degree of hyponatremia if the osmolality of the administered fluid is less that the urine osmolality. One liter of isotonic saline contains 150 mEq each of Na+ and Cl and has an osmolality of 300 mOsm. In a patient with SIADH who has a urine osmolality of 600 mOsm, the administered NaCl will all be excreted in 500 mL of water (300 mOsm in 500 mL = 600 mOsm/L). The remaining 500 mL of administered fluid will be retained in the body and cause further dilution of the serum Na+ concentration. By contrast, 1 L of 3% saline contains 513 mEq each of Na+ and Cl and has an osmolality of 1026 mOsm. All of the NaCl in 1 L of this solution will be excreted in 1.7 L of water and therefore will cause the serum Na+ concentration to increase as a result of the loss of 700 mL of water.

Loop diuretics can also be employed as part of the strategy to treat chronic hyponatremia. Loop diuretics directly interfere in the countercurrent concentrating mechanism by inhibiting NaCl reabsorption in the loop of Henle. The coadministration of these drugs will enhance the effect of intravenous salt solutions by lowering urine osmolality thereby increasing the likelihood that the osmolality of the administered fluid is greater than the urine osmolality. Other measures used to treat chronic hyponatremia are given in Table 1.

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of water channels (aquaporin 2) into the apical membrane. The net effect is increased water excretion with little to no change in urinary Na+ excretion. AVP antagonists have been shown to be more effective than placebo in increasing the serum Na+ concentration in patients with modest asymptomatic hyponatremia.26, 27 These agents can be considered in patients with hyponatremia associated with heart failure or cirrhosis and in patients with irreversible SIADH but should not be used in patients with hypovolemic hyponatremia. The role of these drugs is discussed further in an accompanying article in this supplement, titled New horizons in the pharmacologic approach to hyponatremia: The V2 Receptor Antagonists.

Hyponatremia is one of the most common electrolyte abnormalities encountered in clinical practice. The frequency of the disorder varies according to definition and clinical setting but has been reported to be present in 28% of patients upon hospital admission and in 7% of patients attending an ambulatory community clinic.1 Increasing age, medications, various disease states, and administration of hypotonic fluids are among the known risk factors for the disorder.

The mortality rate in hyponatremic patients is approximately 3 times that of normonatremic hospitalized patients.25 Outcomes are particularly poor in those patients whose serum sodium (Na+) falls during a hospitalization. In 1 prospective study the mortality rate in patients with a normal serum Na+ concentration was 0.2% in comparison to a mortality rate of 11.2% and 25% in patients with a serum Na+ concentration <130 mEq/L and <120 mEq/L, respectively.2 In a recent retrospective cohort study of 10,899 hospitalized patients, the incidence of hyponatremia (<135 mmol/L) at admission was 5.5%.5 As compared to those with normonatremia, these patients were more likely to require intensive care and mechanical ventilation within 48 hours of hospitalization. In addition, hospital mortality, mean length of stay, and costs were significantly greater among patients with hyponatremia than those without.

The association with hyponatremia and adverse outcomes could be the direct result of hyponatremia, the comormidities that lead to the electrolyte derangement, or both. Whatever the mechanism, hyponatremia should not be viewed as an innocuous condition. Rather, clinicians should view this disorder with urgency and institute measures to prevent any further decline in the serum Na+ concentration and initiate appropriate therapy for its correction. This review will first briefly summarize the pathogenesis of hyponatremia and then discuss various disease states encountered in the hospital setting in which hyponatremia is frequently present.

Pathogenesis of Hyponatremia

Hyponatremia is generally associated with a hypoosmolar state and is a marker for a disturbance in water balance. Stated differently, all hyponatremia is dilutional. The approach to the patient with hyponatremia is outlined in Figure 1.

Figure 1
Approach to the hyponatremic patient (EABV, effective arterial blood volume, SIADH, syndrome of inappropriate antidiuretic hormone secretion).

Is the Hyponatremia Representative of a Hypoosmolar State?

There are 3 causes of hyponatremia in which it is not associated with a hypoosmolar state. The first of these is pseudohyponatremia which involves an abnormal measurement of the serum Na+. This occurs in patients with hyperglobulinemia or hypertriglyceridemia in whom plasma water relative to plasma solids is decreased in blood, leading to less Na+ in a given volume of blood. In general, this problem is becoming less prevalent as many laboratories are using Na+ electrodes without diluting the blood such that the Na+ measurement becomes independent of plasma water and nonaqueous contents.

A second cause of hyponatremia in the absence of a hypoosmolar state involves true hyponatremia but with elevations in the concentration of another osmole. Clinical examples include hyperglycemia as seen in uncontrolled diabetes or rarely hypertonic infusion of mannitol used in the treatment of cerebral edema. The accumulation of these effective osmoles creates an osmotic force causing water to move from the intracellular to the extracellular space thus diluting the serum Na+. For every 100 mg/dL rise in glucose or mannitol, the serum Na+ will quickly fall by 1.6 mEq/L. This increase in tonicity also stimulates thirst and arginine vasopressin (AVP) secretion, both of which contribute to further water retention. As a result the plasma osmolality and serum Na+ concentration will continue to fall. Once the plasma osmolality normalizes the serum Na+ will have decreased by 2.8 mEq/L for every 100 mg/dL rise in glucose.

The third cause of hyponatremia in the absence of a hypoosmolar state is the addition of an isosmotic (or near isosmotic) non‐Na+ containing fluid to the extracellular space. This situation typically occurs during a transurethral resection of the prostate or during laprascopic surgery when large amounts of a nonconducting flushing solution containing glycine or sorbitol are absorbed systemically.

Is the Kidney's Ability to Dilute the Urine Intact?

The presence of hypotonic hyponatremia implies that water intake exceeds the ability of the kidney to excrete water. In unusual circumstances, this can occur when the kidneys ability to excrete free water is intact. However, because a normal kidney can excrete 18 L of water per day, the presence of hyponatremia with normal renal water excretion implies the patient is drinking >20 L water/day. This condition is referred to as primary polydipsia. These patients should have a urine osmolality <100 mOsm/L. While primary polydipsia is a common condition which leads to polyuria and polydipsia, it is uncommon as a sole cause of hyponatremia.

Hyponatremia in association with a maximally dilute urine can also result from more moderate fluid intake combined with extremely limited solute intake, a condition often referred to as beer potomania syndrome. In normal subjects, daily solute excretion is usually in the range of 800 mOsm to 1000 mOsm per day. Since the limit of urinary dilution is approximately 50 mOsm/L, then maximum urine output can be calculated to be approximately 16 L to 20 L per day (1000 mOsm/L/50 mOsm/L = 20). Individuals who drink beer or who are ingesting a fad diet have very limited protein intake so that daily solute excretion may be as low as 200 mOsm per day. In this setting, water intake >4 L/day (200 mOsm/50 mOsm/L = 4 L) will exceed renal water excretion and cause hyponatremia. Urine osmolality will be maximally dilute in such a patient.

In the absence of primary polydipsia, hyponatremia is associated with decreased renal water excretion and a urine that is inappropriately concentrated. It is important to note that in the presence of hyponatremia urine should be maximally dilute and a urine osmolality higher than this (>100 mOsm/L) is inappropriate. An inappropriately concentrated urine implies a defect in renal water excretion.

Excretion of water by the kidney is dependent on three factors. First, there must be adequate delivery of filtrate to the tip of the loop of Henle. Second, solute absorption in the ascending limb and the distal nephron must be normal so that the tubular fluid will be diluted. Lastly, AVP levels must be low in the plasma. Of these 3 requirements for water excretion, the one which is probably most important in the genesis of hyponatremia is the failure to maximally suppress AVP levels. In many conditions, decreased delivery of filtrate to the tip of the loop of Henle also contributes.

What is the Volume Status of the Patient?

In patients with hypotonic hyponatremia with an inappropriately concentrated urine, one needs to define whether effective arterial volume is decreased. Most causes of hyponatremia result from a decrease in effective arterial volume which leads to both baroreceptor‐mediated stimulation of AVP secretion and decreased distal delivery of filtrate to the tip of the loop of Henle. If effective arterial volume is low, extracellular fluid (ECF) volume can be low in the volume‐depleted patient (hypovolemic hyponatremia) or can be high in the edematous patient (hypervolemic hyponatremia). If effective arterial volume is normal, one is dealing with the euvolemic causes of hyponatremia (isovolemic hyponatremia).

The clinical determination of effective arterial volume is usually straightforward. On physical examination the best index of effective arterial volume is the presence or absence of an orthostatic change in pulse and blood pressure. Urinary electrolytes are also extremely useful in the assessment of effective arterial volume. Patients with a low effective arterial volume will tend to have a low urinary Na+ and Cl concentration and low fractional excretions of Na+ and Cl in the urine. Patients with euvolemic hyponatremia will be in balance and will excrete Na+ and Cl at rates that reflect dietary intake of Na+ and Cl. Generally urinary Na+ and Cl is >20 mEq/L and fractional excretions of these electrolytes are >1%.

Plasma composition can also be used to assess effective arterial volume. The blood urea nitrogen (BUN) is particularly sensitive to effective arterial volume. In patients with a normal serum creatinine concentration, a high BUN suggests a low effective arterial volume and a low BUN suggests a high effective arterial volume. The plasma uric acid can also be used as a sensitive index of effective arterial volume. In comparing patients with the syndrome of inappropriate secretion of antidiuretic hormone (SIADH) and other causes of hyponatremia, patients with low effective arterial volume tend to have an elevated serum uric acid. The serum urate is low in patients with SIADH. This is due to the fact that these patients are volume expanded although it is clinically difficult to detect the degree of volume expansion.

Syndrome of Inappropriate Antidiuretic Hormone Secretion (SIADH)

In patients who are determined to be clinically euvolemic, a concentrated urine and high AVP levels are inappropriate. The most common cause of this condition is SIADH. This syndrome is generally associated with diseases of the central nervous system, pulmonary diseases, and neoplasms. These conditions lead to secretion of AVP which is inappropriate both from the standpoint of plasma osmolality and effective arterial volume. A number of other etiologies cause a condition of hypoosmolality associated with euvolemia and can mimic the syndrome of inappropriate AVP secretion. These include isolated glucocorticoid deficiency (normal mineralocorticoid activity), hypothyroidism, pain, nausea, acute psychosis, and a variety of drugs.

Clinical Conditions Associated With Hyponatremia in the Hospital Setting

Post‐Operative Hyponatremia

The postoperative patient is particularly prone to developing hyponatremia. AVP levels are increased for several days following surgical procedures due to baroreceptor and nonbaroreceptor‐mediated mechanisms. These patients typically have subtle or overt decreases in effective arterial blood volume due to prolonged preoperative fasting combined with intraoperative and postoperative blood loss and third spacing of fluid. In addition to these factors which unload baroreceptors, postoperative pain, stress, anxiety, nausea, and administration of morphine can further stimulate the release of AVP. In some instances nonsteroidal antiinflmmatory drugs are given which have the effect of augmenting the hydroosmotic actions of AVP.6 In this setting of compromised ability to excrete a water load, administration of hypotonic fluid can precipitate acute iatrogenic hyponatremia.

Postoperative hyponatremia has been a major problem in pediatric populations due to the widespread practice of using hypotonic fluids for maintenance therapy. This approach is based on guidelines developed 50 years ago which were based on calculations linking energy expenditures to water and electrolyte losses.7 More recently, several groups have argued that isotonic rather than hypotonic fluids should be the routine maintenance fluid in such patients.8, 9 The pediatric community has been slow to embrace this approach out of the concern that excessive administration of Na+ would increase the risk of hypernatremia. A systematic meta‐analysis of studies comparing isotonic and hypotonic fluids in hospitalized children found the odds of developing hyponatremia following hypotonic solutions was 17.2 times greater than with isotonic fluids.10 The concern that isotonic maintenance fluids carry a risk of hypernatremia was not supported in the review. Some studies actually reported a decrease in serum Na+ concentration, presumably due to the desalination phenomenon in which hypertonic urine is excreted in volume expanded subjects with persistent vasopressin secretion.

Endocrine Disorders

Glucocorticoid Deficiency

Patients with glucocorticoid deficiency develop hyponatremia. It is important to separate this condition from that of mineralocorticoid deficiency and combined mineralocorticoid‐glucocorticoid deficiency. In patients with mineralocorticoid deficiency, ECF volume and effective arterial volume are low. This leads to baroreceptor stimulation of AVP secretion and to decreased distal delivery of filtrate to the diluting segments of the nephron. In isolated glucocorticoid deficiency, the patients are euvolemic. In these patients, for any given level of low plasma osmolality, vasopressin levels are inappropriately elevated. The administration of hydrocortisone restores the relationship between vasopressin and plasma osmolality to normal.

While it is possible to develop isolated glucocorticoid deficiency with adrenal disease, most adrenal diseases cause loss of mineralocorticoid and glucocorticoid function. Glucocorticoid deficiency in the absence of mineralocorticoid deficiency is usually due to pituitary disease. In fact, severe hyponatremia may be the initial clue to the presence of previously unrecognized hypopituitarism. Insufficient adrenal secretion of glucocorticoids may also be a complication following the long term use of exogenous glucocorticoids.

In addition to high AVP levels, AVP‐independent mechanisms lead to increases in urinary osmolality in glucocorticoid deficiency. The nature of this AVP‐independent effect is likely multifactorial. First, glucocorticoid deficiency is associated with a diminished cardiac output and an impaired systemic vascular response to hypotension. These changes will lead to a slight decline in glomerular filtration rate and increased volume absorption in the proximal tubule and thin descending limb. As a result, distal delivery of filtrate to the diluting segment will be abnormally low in glucocorticoid deficiency. Second, mineralocorticoid and glucocorticoid deficiency have both been shown to result in increased expression of aquaporin 2 water channels in the collecting duct.11 This later effect will further limit maximal urinary dilution and contribute to net water retention.

These AVP‐independent mechanisms are consistent with the clinical observation that patients with diabetes insipidus appear to improve clinically when they develop coexistent anterior pituitary insufficiency, and treatment of these patients with glucocorticoids appears to worsen the diabetes insipidus. Thus, in the patient with diabetes, insipidus‐free water excretion will be extremely large. When simultaneous glucocorticoid deficiency develops, free‐water excretion decreases.

Hypothyroidism

Myxedema coma is the most severe form of hypothyroidism and is commonly associated with hyponatremia.12 In this setting, blood pressure can be low because of decreased intravascular volume and cardiovascular collapse. The hypotension can be refractory to vasopressor therapy in the absence of thyroid hormone therapy. Cardiac output and stroke volume are low. A defect in renal water excretion develops as a result of baroreceptor mediated increases in AVP along with decreased delivery of filtrate to the distal nephron. In hypothyroid rats, there is upregulation of aquaporin 2 water channels. In these animals, administration of a V2 receptor antagonist reverses the increased water channel expression and corrects the impaired response to an acute water load.11

While reduced cardiac output and blood pressure associated with severe hypothyroidism can provide a stimulatory effect for AVP release through a baroreceptor mediated mechanism, milder forms of hypothyroidism can be considered in the differential diagnosis of euvolemic hyponatremia. In this setting, impaired renal excretion of water is presumably due to increased release of AVP due to the absence of a tonic inhibitory effect of thyroid hormone in the central nervous system. The degree of hyponatremia in this setting is typically mild.

Heart Failure

Hyponatremia is a common complication of left‐sided heart failure and several studies have shown that it is an independent predictor of mortality.13, 14 A similar association between reduced survival and hyponatremia is present in advanced right‐sided heart failure in patients with pulmonary arterial hypertension.15 Patients with heart failure who are hyponatremic have higher circulating levels of neurohormones (catecholamines, renin, angiotensin II, aldosterone, and AVP) than normonatremic subjects and are more likely to have prerenal azotemia. In addition to being a marker for the extent of neurohumoral activation, hyponatremia may play a more direct role in adverse outcomes through maladaptive volume regulatory responses of cardiac myocytes and by direct effects of AVP on cardiac and coronary V1a receptors.

Heart failure is associated with arterial underfilling leading to arterial baroreceptor‐mediated activation of the neurohumoral axis. This underfilling is due to decreased cardiac output in low‐output heart failure and decreased systemic vascular resistance in high‐output heart failure. Activation of the sympathetic nervous system along with the renin‐angiotensin‐aldosterone system leads to renal salt retention while the increase in AVP is associated with water retention and hyponatremia.

Cirrhosis

Hyponatremia is a common electrolyte abnormality in patients with cirrhosis and occurs with a frequency that tends to parallel the severity of liver disease.16 Patients with a serum Na+ 130 mEq/L are more likely to have refractory ascites and require therapeutic paracentesis. Hepatic encephalopathy, hepatorenal syndrome, and spontaneous bacterial peritonitis are also more common in patients with a serum Na+ 130 mmol/L than in patients with a normal serum Na+ concentration. Hyponatremia increases morbidity and mortality from hepatic transplantation and is associated with osmotic demyelination in the postoperative period because of the large increase in Na+ concentration associated with the procedure.17

Hepatic cirrhosis is characterized by a decreased effective arterial blood volume and activation of neurohumoral effectors. Reduced effective circulating volume due to generalized and specifically to arterial splanchnic vasodilation leads to baroreceptor‐mediated nonosmotic stimulation of AVP release and an impaired ability to excrete electrolyte‐free water.18 Reduced Na+ delivery to the distal tubule because of a low glomerular filtration rate and increased proximal Na+ reabsorption adds to the susceptibility of cirrhotic patients to hyponatremia.

Hyponatremic‐Hypertensive Syndrome

The development of hyponatremia in patients with severe hypertension associated with renal artery stenosis has been called the hyponatremic‐hypertensive syndrome.19, 20 Patients with this syndrome present with a variety of signs and symptoms that include headache, confusion, postural dizziness, polyuria, polydipsia, and salt craving. In a retrospective review of 32 patients with this syndrome, most of the subjects were thin, elderly, women smokers who had atherosclerotic renal vascular disease.19 Biochemical abnormalities included not only hyponatremia, but hypokalemia and increased plasma renin activity. The mean serum Na+ concentration was 129.7 mmol/L (range 120‐135).

The precise mechanism of this syndrome is not known. Given the available data, angiotensin‐mediated thirst coupled with nonosmotic release of AVP provoked by angiotensin II and/or hypertensive encephalopathy are likely. Na+ depletion due to pressure natriuresis, and K+ depletion due to hyperaldosteronism are also likely to play a role in the pathogenesis of hyponatremia.

Pneumonia

An association between pneumonia and hyponatremia has been known for quite some time but has been poorly defined. Of the various etiologic agents, legionella is more commonly associated with hyponatremia as compared to other types of community acquired disease. As has been true for many disorders, hyponatremia is associated with longer hospital stays and hospital mortality, most likely reflecting the severity of the pneumonia rather than morbidity from the usually mild and asymptomatic hyponatremia.

Central Nervous System Disease

Hyponatremia is a frequent complication of central nervous system disease to include bacterial meningitis and traumatic brain injury. Potential mechanisms include the development of SIADH, cerebral salt wasting (CSW), or hypotonic fluid administration in the setting of impaired renal water excretion as in patients with low effective volume. Of these various causes, hyponatremia is frequently attributed to SIADH. As previously mentioned, this syndrome is characterized by hyponatremia in the setting of an inappropriately concentrated urine, increased urine Na+ concentration, and evidence of normal or slightly increased intravascular volume.

However there are patients with intracranial disease who develop hyponatremia with similar characteristics but differ in that there is clinical evidence of a contracted ECF volume. This form of hyponatremia is due to excessive renal Na+ excretion resulting from a centrally mediated process and is termed CSW. The onset of this disorder is typically seen within the first ten days following a neurosurgical procedure or after a definable event, such as a subarachnoid hemorrhage or stroke. CSW has also been described in other intracranial disorders, such as carcinomatous or infectious meningitis and metastatic carcinoma.21 The distinction between SIADH and CSW is of considerable clinical importance given the divergent nature of the treatments. Fluid restriction is the treatment of choice in SIADH, whereas the treatment of CSW comprises vigorous Na+ and volume replacement.

Treatment of Hyponatremia

Symptoms of hyponatremia include nausea and malaise, which can be followed by headache, lethargy, muscle cramps, disorientation, restlessness and obtundation, and seizures. The principal danger of hyponatremia or hypernatremia relates to effects on central nervous system function due to changes in brain size (Figure 2).

Figure 2
Change in cell size with acute and chronic hyponatremia. Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. Rapid correction of chronic hyponatremia causes acute cellular shrinkage and can lead to osmotic demyelination.

Hyponatremia initially leads to cell swelling driven by the higher intracellular osmolality. The net result is equilibration of intracellular and extracellular osmolality at the expense of increased brain volume. Cells in general, and brain cells in particular, then respond by decreasing the number of intracellular osmoles and as intracellular osmolality decreases, cell size returns toward normal despite the presence of hyponatremia. If the decrease in ECF osmolality is slow, there will be no measurable cell swelling. This pathophysiologic sequence correlates well with clinical observations. If hyponatremia is slow in onset, neurologic symptoms and permanent brain damage are unusual, even if the decreases in Na+ concentration and ECF osmolality are large. Conversely, if hyponatremia is rapid in onset, cerebral edema and significant CNS symptoms and signs can occur with lesser changes in serum Na+ concentration.

When treating a patient with hyponatremia, the Na+ concentration should be raised at the rate at which it fell. In a patient whose serum Na+ concentration has decreased rapidly (<48 hours), neurologic symptoms are frequently present and there is cerebral edema. In this setting there has not been sufficient time to remove osmoles from the brain and rapid return to normal ECF osmolality merely returns brain size to normal. In general, the development of hyponatremia in the outpatient setting is more commonly chronic in duration and should be corrected slowly. By contrast, hyponatremia of short duration is more likely to be encountered in hospitalized patients receiving intravenous free water. Use of ecstasy, exercise‐induced hyponatremia, or patients with primary polydipsia can also develop acute hyponatremia and if symptomatic may similarly require rapid correction. Raising the serum Na+ concentration by 4 mEq/L to 6 mEq/L over a several hour period is both safe and effective in preventing untoward effects of acute hyponatremia.22 A reasonable strategy to accomplish this goal is the regimen recommended for the treatment of athletes with hyponatremia and encephalopathy.23 These guidelines suggest an immediate bolus of 100 mL 3% NaCl (513 mEq/L). If there is no neurologic improvement two additional 100 mL 3% NaCl bolus infusions separated by 10 minute intervals can be given.

In patients with chronic hyponatremia (>48 hours duration) the serum Na+ concentration has fallen slowly. Neurologic symptoms are generally minimal, brain size is normal, and the number of intracellular osmoles is decreased. Sudden return of ECF osmolality to normal values will lead to cell shrinkage and possibly precipitate osmotic demyelination. Experts in the field suggest this complication can be prevented by adhering to the following limits of correction: 10 mmol/L in 24 hours, 18 mmol/L in 48 hours, and 20 mmol/L in 72 hours.22 In order to maximize patient safety, the goals of therapy should be more modest: 6 to 8 mmol/L in 24 hours, 12 to 14 mmol/L in 48 hours, and 14 to 16 mmol/L in 72 hours.

A formula designed to predict the increase in serum Na+ concentration to be expected from the infusion of 1 L of a given infusion is given below: 1.0 is indicative of no electrolyte‐free water in the urine and predicts a poor response unless water intake is severely limited.

Conservative Management of Chronic Hyponatremia
Treament Mechanism Advantages Limitations
  • Abbreviation: AVP,arginine vasopressin.

Fluid restriction (0.5‐1.0 L/day) water intake Effective, inexpensive Poor compliance
Demeclocyline (600‐1200 mg/day) Inhibits action of AVP Widely available Slow onset, nephrotoxic, expensive
Urea (30 gm/day) Osmotic diuresis Possiblerisk of osmotic demyelination Poor palatability, avoid in chronic kidney and liver disease
Lithium (up to 900 mg/daily) Inhibits action of AVP Widely available Slow onset, toxicity

When treating patients with SIADH with intravenous salt solutions, one can worsen the degree of hyponatremia if the osmolality of the administered fluid is less that the urine osmolality. One liter of isotonic saline contains 150 mEq each of Na+ and Cl and has an osmolality of 300 mOsm. In a patient with SIADH who has a urine osmolality of 600 mOsm, the administered NaCl will all be excreted in 500 mL of water (300 mOsm in 500 mL = 600 mOsm/L). The remaining 500 mL of administered fluid will be retained in the body and cause further dilution of the serum Na+ concentration. By contrast, 1 L of 3% saline contains 513 mEq each of Na+ and Cl and has an osmolality of 1026 mOsm. All of the NaCl in 1 L of this solution will be excreted in 1.7 L of water and therefore will cause the serum Na+ concentration to increase as a result of the loss of 700 mL of water.

Loop diuretics can also be employed as part of the strategy to treat chronic hyponatremia. Loop diuretics directly interfere in the countercurrent concentrating mechanism by inhibiting NaCl reabsorption in the loop of Henle. The coadministration of these drugs will enhance the effect of intravenous salt solutions by lowering urine osmolality thereby increasing the likelihood that the osmolality of the administered fluid is greater than the urine osmolality. Other measures used to treat chronic hyponatremia are given in Table 1.

AVP antagonists block the V2 receptor located on the basolateral surface of the collecting duct thereby antagonizing the ability of AVP to cause insertion of water channels (aquaporin 2) into the apical membrane. The net effect is increased water excretion with little to no change in urinary Na+ excretion. AVP antagonists have been shown to be more effective than placebo in increasing the serum Na+ concentration in patients with modest asymptomatic hyponatremia.26, 27 These agents can be considered in patients with hyponatremia associated with heart failure or cirrhosis and in patients with irreversible SIADH but should not be used in patients with hypovolemic hyponatremia. The role of these drugs is discussed further in an accompanying article in this supplement, titled New horizons in the pharmacologic approach to hyponatremia: The V2 Receptor Antagonists.

References
  1. Hawkins R.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chem Acta.2003;337:169172.
  2. Anderson RJ,Chung HM,Kluge R,Schrier RW.Hyponatremia: a prospective analysis of its epidemiology and the pathogenetic role of vasopressin.Ann Intern Med.1985;102(2):164168.
  3. Gill G,Huda B,Boyd A, et al.Characteristics and mortality of severe hyponatraemia‐a hospital‐based study.Clin Endocrinol (Oxf).2006;65(2):246249.
  4. Waikar S,Mount D,Curhan G.Mortality after hospitalization with mild moderate and severe hyponatremia.Am J Med.2009;122:88578865.
  5. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  6. Palmer BF.Renal complications associated with use of nonsteroidal anti‐inflammatory agents.J Investig Med.1995;43(6):516533.
  7. Paut O,Lacroix F.Recent developments in the perioperative fluid management for the paediatric patient.Curr Opin Anaesthesiol.2006;19:268277.
  8. Moritz M,Ayus J.Prevention of hospital‐acquired hyponatremia: a case for using isotonic saline.Pediatrics.2003;111:227230.
  9. Hoorn EJ,Geary D,Robb M,Halperin Ml,Bohn D.Acute hyponatremia related to intravenous fluid administration in hospitalized children: an observational study.Pediatrics.2004;113(5):12791284.
  10. Choong K,Kho ME,Menon K,Bohn D.Hypotonic versus isotonic saline in hospitalized children: a systematic review.Arch Dis Child.2006;91(10):828835.
  11. Schrier R.Vasopressin and Aquaporin 2 in clinical disorders of water homeostasis.Semin Nephrol.2008;28:289296.
  12. Wartofsky L.Myxedema coma.Edocrinol Metab Clin N Am.2006;35:687698.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28(8):920921.
  14. Schrier R,Bansal S.Pulmonary hypertension, right ventricular failure, and kidney: different from left ventricular failure.Clin J Am Soc Nephrol.2008;3:12321237.
  15. Forfia PR,Mathai SC,Fisher MR, et al.Hyponatremia predicts right heart failure and poor survival in pulmonary arterial hypertension.Am J Respir Crit Care Med.2008;177(12):13641369.
  16. Angeli P,Wong F,Watson H,Ginès P;CAPPS investigators. Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44(6):15351542.
  17. Yu J,Zheng SS,Liang TB,Shen Y,Wang WL,Ke QH.Possible causes of central pontine myelinolysis after liver transplantation.World J Gastroenterol.2004;10(17):25402543.
  18. Palmer BF.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  19. Browne WL,Nair B.The hyponatremic hypertensive syndrome in renal artery stenosis: An infrequent cause of hyponatremia.J Postgrad Med.2007;53:4143.
  20. Agarwal M,Lynn K,Richards M,Nicholls M.Hyponatremic‐hypertensive syndrome with renal ischemia: an underrecognized disorder.Hypertension.1993;33:10201024.
  21. Palmer BF.Hyponatremia in patients with central nervous system disease: SIADH or CSW.Trends Endocrinol Metab.2003;14:182187.
  22. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  23. Hew‐Butler T,Ayus J,Kipps C, et al.Statement of the second international exercise‐associated hyponatremia consensus development conference, New Zealand.Clin J Sport Med.2008;18:111121.
  24. Mohmand H,Issa D,Ahmad Z,Cappuccio J,Kouides R,Sterns R.Hypertonic saline for hyponatremia: risk of inadvertent overcorrection.Clin J Am Soc Nephrol.2007;2:11101117.
  25. Perianayagam A,Sterns R,Silver S, et al.DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia.Clin J Am Soc Nephrol.2008;3:331336.
  26. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol2007;27:447457.
  27. Schrier R,Gross P,Gheorghiade M, et al.,Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
References
  1. Hawkins R.Age and gender as risk factors for hyponatremia and hypernatremia.Clin Chem Acta.2003;337:169172.
  2. Anderson RJ,Chung HM,Kluge R,Schrier RW.Hyponatremia: a prospective analysis of its epidemiology and the pathogenetic role of vasopressin.Ann Intern Med.1985;102(2):164168.
  3. Gill G,Huda B,Boyd A, et al.Characteristics and mortality of severe hyponatraemia‐a hospital‐based study.Clin Endocrinol (Oxf).2006;65(2):246249.
  4. Waikar S,Mount D,Curhan G.Mortality after hospitalization with mild moderate and severe hyponatremia.Am J Med.2009;122:88578865.
  5. Zilberberg MD,Exuzides A,Spalding J, et al.Epidemiology, clinical and economic outcomes of admission hyponatremia among hospitalized patients.Curr Med Res Opin.2008;24:16011608.
  6. Palmer BF.Renal complications associated with use of nonsteroidal anti‐inflammatory agents.J Investig Med.1995;43(6):516533.
  7. Paut O,Lacroix F.Recent developments in the perioperative fluid management for the paediatric patient.Curr Opin Anaesthesiol.2006;19:268277.
  8. Moritz M,Ayus J.Prevention of hospital‐acquired hyponatremia: a case for using isotonic saline.Pediatrics.2003;111:227230.
  9. Hoorn EJ,Geary D,Robb M,Halperin Ml,Bohn D.Acute hyponatremia related to intravenous fluid administration in hospitalized children: an observational study.Pediatrics.2004;113(5):12791284.
  10. Choong K,Kho ME,Menon K,Bohn D.Hypotonic versus isotonic saline in hospitalized children: a systematic review.Arch Dis Child.2006;91(10):828835.
  11. Schrier R.Vasopressin and Aquaporin 2 in clinical disorders of water homeostasis.Semin Nephrol.2008;28:289296.
  12. Wartofsky L.Myxedema coma.Edocrinol Metab Clin N Am.2006;35:687698.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28(8):920921.
  14. Schrier R,Bansal S.Pulmonary hypertension, right ventricular failure, and kidney: different from left ventricular failure.Clin J Am Soc Nephrol.2008;3:12321237.
  15. Forfia PR,Mathai SC,Fisher MR, et al.Hyponatremia predicts right heart failure and poor survival in pulmonary arterial hypertension.Am J Respir Crit Care Med.2008;177(12):13641369.
  16. Angeli P,Wong F,Watson H,Ginès P;CAPPS investigators. Hyponatremia in cirrhosis: results of a patient population survey.Hepatology.2006;44(6):15351542.
  17. Yu J,Zheng SS,Liang TB,Shen Y,Wang WL,Ke QH.Possible causes of central pontine myelinolysis after liver transplantation.World J Gastroenterol.2004;10(17):25402543.
  18. Palmer BF.Pathogenesis of ascites and renal salt retention in cirrhosis.J Invest Med.1999;47:183202.
  19. Browne WL,Nair B.The hyponatremic hypertensive syndrome in renal artery stenosis: An infrequent cause of hyponatremia.J Postgrad Med.2007;53:4143.
  20. Agarwal M,Lynn K,Richards M,Nicholls M.Hyponatremic‐hypertensive syndrome with renal ischemia: an underrecognized disorder.Hypertension.1993;33:10201024.
  21. Palmer BF.Hyponatremia in patients with central nervous system disease: SIADH or CSW.Trends Endocrinol Metab.2003;14:182187.
  22. Sterns R,Nigwekar S,Hix J.The treatment of hyponatremia.Semin Nephrol.2009;29:282299.
  23. Hew‐Butler T,Ayus J,Kipps C, et al.Statement of the second international exercise‐associated hyponatremia consensus development conference, New Zealand.Clin J Sport Med.2008;18:111121.
  24. Mohmand H,Issa D,Ahmad Z,Cappuccio J,Kouides R,Sterns R.Hypertonic saline for hyponatremia: risk of inadvertent overcorrection.Clin J Am Soc Nephrol.2007;2:11101117.
  25. Perianayagam A,Sterns R,Silver S, et al.DDAVP is effective in preventing and reversing inadvertent overcorrection of hyponatremia.Clin J Am Soc Nephrol.2008;3:331336.
  26. Zeltser D,Rosansky S,va Rensburg H,Verbalis J,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol2007;27:447457.
  27. Schrier R,Gross P,Gheorghiade M, et al.,Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
Issue
Journal of Hospital Medicine - 5(3)
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S1-S7
Page Number
S1-S7
Article Type
Display Headline
Diagnostic approach and management of inpatient hyponatremia
Display Headline
Diagnostic approach and management of inpatient hyponatremia
Legacy Keywords
arginine vasopressin, hyponatremia, osmolality, osmotic demyelination
Legacy Keywords
arginine vasopressin, hyponatremia, osmolality, osmotic demyelination
Sections
Article Source
Copyright © 2010 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Professor of Internal Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

Treating Hyponatremia in Heart Failure

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Managing hyponatremia in patients with heart failure

Heart failure is a common and growing problem in both industrialized and developing nations. In the U.S. alone there are estimated to be well over 5 million heart failure patients and that number is expected to double over the next few decades. There are several reasons for this pandemic, most notably the aging of the world's population, a rising incidence of heart failure risk factors including hypertension, diabetes and obesity, and improved survival post‐myocardial infarction (MI).1 Greater longevity of patients with existing heart failure as a result of treatment with drugs and devices that lower mortality and, in developing nations, a reduction in premature mortality from infectious diseases have also contributed to the increase in heart failure prevalence. Although there have been important advances in treating heart failure that have improved outcomes over the past several decades, morbidity and mortality remain unacceptably high and quality of life is substantially reduced. Thus, there is considerable need for finding new approaches for managing patients with this condition.

The Role of Neurohormonal Blockade in the Treatment of Heart Failure Patients

The pathophysiology of heart failure is complex. In patients who develop systolic dysfunction, the pathway initially involves injury to the heart and/or increases in wall stress which activates a variety of compensatory responses in an effort to reestablish homeostasis within the cardiovascular (CV) system. Many of these responses are mediated by neurohormonal systems that are stimulated both systemically and locally within the heart itself.2, 3 While this widespread neurohormonal activation has some short‐term benefits in maintaining cardiac performance, there is clear evidence that it has adverse effects when maintained over time. The deleterious effects of neurohormonal activation in heart failure include excess salt and water retention, constriction of arterial resistance and venous capacitance vessels, increased load on the heart, electrolyte abnormalities and maladaptive cardiac remodeling. The critical role of neurohormonal activation in the pathogenesis and progression of heart failure has been confirmed by the results of large scale clinical trials which show that neurohormonal blocking agents such as angiotensin converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs), beta blockers (BBs), and aldosterone blockers greatly reduce morbidity and mortality and result in a variety of other favorable effects in heart failure patients.49 Based on their profound effects on outcomes, strategies that target neurohormonal activation have emerged over the past 2 decades as the cornerstone of medical management of heart failure.

Establishing Risk in Heart Failure

Although there have been impressive gains in reducing morbidity and mortality in heart failure patients over the past 3 decades, the overall clinical course remains unfavorable in a substantial portion of this population. A wide variety of risk factors which identify patients who are more likely to do poorly in the future have been identified. These include demographic variables (eg, age), functional and structural abnormalities, hemodynamic measurements, symptomatic status, exercise capacity, quality of life, presence of comorbidities and a myriad of blood tests and biomarkers. Amongst the plethora of risk factors for poor outcome, decompensation of heart failure which results in hospitalization has been recognized as 1 of the most important prognostic indicators. The Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients With Heart Failure (OPTIMIZE‐HF) Registry which included a large fairly representative population of heart failure patients from throughout the U.S. followed a subset of patients for 60 days to 90 days immediately post‐discharge from a hospitalization that was associated with decompensated heart failure.10 Over this relatively short time period hospital readmission rate was over 30% and mortality over 9%. Thus, within 2 months to 3 months of discharge following an episode of decompensation 40% of heart failure patients had either died or were back in the hospital. Among the many risk factors that have been used to predict morbidity and mortality outcomes either during or following hospitalization, the ones that appear to be the most powerful in detecting patients who are likely to do poorly are impaired renal function,11, 12 low systolic blood pressure,13 persistence of congestion at the time of hospital discharge,14 elevation of various biomarkers such as B‐type natriuretic peptide (BNP)15 and the presence of hyponatremia.16

Hyponatremia in Heart Failure

Incidence of Hyponatremia in Heart Failure Patients

Determining the exact incidence of hyponatremia in heart failure patients has been challenging due to differences in the populations that have been studied and in the criteria used to define hyponatremia. The Acute and Chronic Therapeutic Impact of a Vasopressin Antagonist in Congestive Heart Failure (ACTIV in CHF) study reported that 21.3% of the cohort of hospitalized patients had a serum sodium below 136 mmol/L.17 Higher incidences were reported in the Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) which found that 27% of patients had serum sodium concentrations between 132 mmol/L to 135 mmol/L.18 The Evaluation Study of CHF and Pulmonary Artery Catheter Effectiveness (ESCAPE) trial reported that 18% of patients had hyponatremia defined as serum sodium concentration below 134 mmol/L.19 Data from OPTIMIZE‐HF, a registry that captured data from a large cohort of representative patients who were hospitalized with decompensated heart failure indicate that such patients have a wide distribution of admission sodium values (Figure 1).16 Overall, 19.7% of the OPTIIMIZE‐HF patients had values below 135 mmol/L.

Figure 1
Distribution of admission serum sodium in patients hospitalized with a primary discharge diagnosis of heart failure.

Risk Associated With Hyponatremia

There is considerable evidence that hyponatremia is associated with increased risk for poor outcomes in heart failure patients. One of the first reports that related hyponatremia to a poor prognosis came from Lee and Packer who analyzed 30 clinical, hemodynamic, and biochemical variables in outpatients with severe heart failure.20 Their results showed that serum sodium was the most powerful predictor of CV mortality. Similar findings have been reported by other investigators.21 In patients hospitalized for decompensated heart failure, the presence of hyponatremia has been shown to be an independent predictor of longer duration of stay.22 Results from the OPTIMIZE‐HF Registry confirmed the adverse impact of hyponatremia on length of stay during a heart failure hospitalization and also suggested that low serum sodium was associated with significantly higher in‐hospital and post‐discharge mortality rates.16 In this large cohort of hospitalized patients with decompensated heart failure, each 3 mmol/L decrease in serum sodium below 140 mmol/L increased the risk of in‐hospital and follow‐up mortality by 19.5% and 10%, respectively. This association is depicted in Figure 2. A similar adverse impact of hyponatremia on post‐discharge outcomes was seen in the results of the ACTIV in CHF study.17 Overall, 69 patients out of a total of 319 who were included in the study (21.6%) had a serum sodium that was <136 mmol/L. Mortality over a 60 day period of follow‐up was 14.5% in the hyponatremic patients compared to 4% in the 250 patients whose serum sodium was 136 mmol/L.

Figure 2
Relationship between admission serum sodium level and in‐hospital mortality. Restrictive cubic spline transformation plot with 95% confidence intervals is shown.

Although these data present a powerful argument that hyponatremia is a potent risk factor for poor outcomes in heart failure patients both during and following hospitalization, they do not determine whether or not the relationship is causal. It is possible that the poor outcomes that have been observed in hyponatremic patients are related to an association between low serum sodium levels and the more profound alterations in hemodynamics, neurohormonal activation and renal function that are seen in patients with advanced heart failure. Alternatively, increased edema in vital organs including the heart in hyponatremic patients could further impair already tenuous function and contribute to a downward spiral in the clinical course. The impact of hyponatremia in limiting the use of loop diuretics and spironolactone may also be associated with a less favorable long‐term outcome. The presence of hyponatremia may also contribute to a poor outcome as a result of effects on cognitive and neuromuscular function.23, 24 Impaired cognition could adversely affect compliance with the medical regimen while neuromuscular problems related to hyponatremia could contribute to an increased incidence of falls and other traumatic injuries that older patients with chronic diseases are already at high risk of experiencing.

Pathophysiology of Hyponatremia

The pathophysiology of hyponatremia in heart failure patients involves several different processes.25 Overall, heart failure is characterized by retention of excessive amounts of salt and water in the body. Sodium retention is related to decreased renal perfusion that is caused by the effects of reduced cardiac output, decreased renal perfusion pressure and increased afferent glomerular arteriolar resistance. A reduction in glomerular filtration and increased reabsorption of sodium and water in the proximal renal tubules leads to a reduction in the delivery of water and solute to the diluting segment of the nephron. In patients with heart failure the renin‐angiotensin system (RAS) is activated, an event that is further stimulated by the administration of loop diuretics.26 Angiotensin II (Ang II), a key effector molecule of the RAS, increases tone in renal efferent arterioles. This tends to enhance both sodium and water reabsorption both through an increase in the glomerular filtration fraction and by direct effects on the distal tubule.27 Ang II also stimulates the thirst center of the brain both directly and through stimulation of antidiuretic hormone to promote the ingestion of excessive amounts of hypotonic fluids.25 Water reabsorption in the distal portion of the nephron is governed by arginine vasopressin (AVP). High levels of AVP are seen in patients with heart failure26, 28 and there is a significant association between serum levels of this peptide and the symptomatic state of the patient. There is evidence that in heart failure patients AVP levels are elevated disproportionally to plasma osmolality and serum sodium concentrations.28, 29 Even when serum osmolality is increased in this setting by infusion of sodium, AVP levels fail to demonstrate an appropriate reduction suggesting that mechanisms other than activation of osmoreceptors are involved. The effects of AVP in the pathogenesis of hyponatremia are significant. This peptide binds to the vasopressin‐2 (V2) receptor in the collecting duct of the kidney stimulating an increase in the second messenger cyclic AMP.30 Downstream signaling initiated by cyclic AMP leads to an increase in the number and activation of aquaporin‐2 water channels on the luminal surface of epithelial cells in the collecting tubule.31 The presence of these activated pores is necessary for water permeability in the collecting duct and leads to an increase in the reabsorption of free water. Finally, the use of diuretics in the treatment of heart failure has been implicated in the development and worsening of the hyponatremic state.32

Treatment of Hyponatremia

Treatment options for dealing with hyponatremia in heart failure patients have been limited until recently. Since low cardiac output and/or diminished renal perfusion are involved, interventions which improve cardiac and renal function can reverse hyponatremia. While this can be accomplished by the use of inotropic agents, the use of drugs such as dobutamine, milrinone, and other inotropes in either stable or decompensated heart failure patients with adequate tissue perfusion is not routinely recommended due to a well‐documented increase in adverse effects, particularly in patients with coronary artery disease.33 The use of hypertonic saline is also not recommended since it may worsen the extent of volume overload in decompensated patients. Fluid restriction can be used to treat hyponatremia, particularly in hospitalized patients where stricter control on input is possible. Hyponatremic patients, however, often experience excessive thirst and restriction to less than 1000 cc to 1500 cc is rarely, if ever, successful for more than a brief period of time. The use of angiotensin converting enzyme (ACE) inhibitors has been associated with an improvement in serum sodium levels. Packer et al. treated a cohort of heart failure patients who were receiving a stable dose of diuretic with an ACE inhibitor captopril and found that over a 2‐week period the serum sodium increased from 131.2 0.5 to 135.9 0.5 mmol/L; P < 0.001).34 These investigators concluded that the RAS was involved in the pathogenesis of hyponatremia and that ACEIs increased sodium levels in hyponatremic patients, though the mechanism through which this occurs has not been delineated.

The Use of Vaptans in Treating Hyponatremia

AVP actions are mediated by an interaction of the peptide with a series of receptors located on cells throughout the body. Vaptans are nonpeptidergic agents which block the interaction of AVP with these receptors; they are classified according to which receptor subtype they affect. As mentioned earlier, activation of the V2 receptor on renal tubular cells increases collecting duct permeability to water and leads to reabsorption of free water.35 The V1A receptor is located on vascular smooth muscle cells where it mediates an increase in vasomotor tone. V1A receptors are also found on platelets and in the myometrium where they mediate aggregation and uterine contraction, respectively. Some of the AVP antagonists (eg, conivaptan) block both the V1A and V2 receptors while others (eg, tolvaptan and lixivaptan) are selective for the V2 receptor.

Tolvaptan, a V2 selective agent, has been studied extensively in heart failure as well as in patients with hyponatremia due to a variety of causes. One of the initial studies was performed in a group of 254 heart failure patients who were randomized to receive tolvaptan in doses ranging from 30 mg to 60 mg daily or placebo.36 Tolvaptan at all doses studied was associated with significant reductions in body weight and improvement in the signs and symptoms of heart failure. All doses were also associated with an increase in serum sodium levels in this study. Patients who were hyponatremic made up 28% of the study population and these patients experienced the greatest increase in serum sodium. Of note was the fact that as early as day 1 in the study 80% of tolvaptan (as opposed to 40% of placebo) patients had normalization of their serum sodium levels. These effects occurred without significant changes in blood pressure or renal function and the major side effects that were seen were polyuria, dry mouth, and thirst. This study was followed by the ACTIV in CHF trial which included a slightly larger population of 319 patients (of whom 21.3% were hyponatremic at baseline) who were hospitalized due to decompensated heart failure.17 Mean body weight decreased significantly more in patients treated with tolvaptan compared to those who received placebo. Tolvaptan‐treated patients also experienced increases in serum sodium that were greatest in the patients who were hyponatremic at baseline. These changes persisted throughout the duration of the study. On post hoc analysis, event‐free survival tended to be longer for the combined group of patients treated with tolvaptan compared to placebo but there were no differences in the rate of rehospitalization or unscheduled visits for heart failure. As in the initial study, tolvaptan was well tolerated, with dry mouth being the main side effect. There were no significant hemodynamic or renal effects.

The efficacy of vasopressin antagonism in heart failure outcome study with tolvaptan (EVEREST) study randomized 4133 patients hospitalized for decompensated heart failure to receive either tolvaptan 30 mg daily or placebo in addition to their standard therapy. The short‐term goal of the study was to assess the effects of therapy on a composite end‐point of patient assessed global clinical features and weight loss on day 7 (or at the time of hospital discharge) after starting treatment.37 The results of EVEREST demonstrated that patients treated with tolvaptan had greater improvement in the composite primary end‐point. This effect was driven by a greater reduction in body weight with active drug. Although, changes in global clinical status did not differ between the study groups, tolvaptan‐treated patients reported significantly greater improvement in dyspnea at day 1. In 1 (but not the other of the 2 component trials of EVEREST) there was also an improvement in edema. At day 1 and at discharge, the tolvaptan group with hyponatremia (defined as a serum sodium below 134 mEq/L) demonstrated significantly greater increases in serum sodium than in the hyponatremic placebo treated patients. Tolvaptan was well tolerated and serious adverse event frequencies were similar between groups, without excess renal failure or hypotension.

Patients who were enrolled in EVEREST were then followed for an average of 9.9 months on tolvaptan or placebo in order to assess the effects of treatment on the dual primary endpoints of all‐cause mortality (both superiority and noninferiority) and CV death or heart failure hospitalization.38 The results demonstrated no significant differences in either primary or secondary morbidity and mortality outcomes between tolvaptan and placebo treated patients. In the EVEREST patients with baseline serum sodium levels less than 134 mEq/L, there was a significant increase of 5.49 mEq/L 5.77 mEq/L (mean SD) at day 7 or discharge, if earlier, with tolvaptan, compared with 1.85 mEq/L 5.10 mEq/L in the placebo group. This effect was observed as early as day 1 and was maintained throughout the 40 weeks of treatment. Side effects were minimal. Overall, tolvaptan increased thirst and dry mouth, but the frequencies of major adverse events were similar in the 2 groups.

Two parallel multicenter, randomized, double‐blind, placebo‐controlled trials, termed collectively the Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2), examined the effect of tolvaptan on hypervolemic and euvolemic hyponatremia of diverse causes.39 The 448 patients included in the 2 studies were randomly assigned to either placebo or tolvaptan starting at a dose of 15 mg daily (increasing to 30 mg and then 60 mg if needed, depending on serum sodium concentrations) and followed over a 30 day period. The population included 138 patients (31%) with chronic heart failure as the cause of hyponatremia with the remainder of the population divided between patients with cirrhosis or syndrome of inappropriate antidiuretic hormone hypersecretion (SIADH) and other causes of hyponatremia. The 2 primary end points for all patients were the change in the average daily area under the curve for the serum sodium concentration from baseline to day 4 and the change from baseline to day 30. As shown in Figure 3, serum sodium concentrations increased significantly more in the tolvaptan group than in the placebo group during the first 4 days and after the full 30 days of therapy. A planned analysis of the SALT trials demonstrated that correction of hyponatremia with tolvaptan was associated with significant improvement in self‐reported mental status, particularly in patients with marked hyponatremia or SIADH. Improvements in mental health scores were positively correlated with changes in serum [Na+] in both the tolvaptan and placebo groups and reversed after cessation of therapy, suggeting that hyponatremia‐associated impairments in mental function can be significantly improved by raising the serum [Na+]. The major side effects with tolvaptan included increased thirst, dry mouth, and increased urination. Tolvaptan has been approved by the US Food and Drug Administration (FDA) for the treatment of euvolemic and hypervolemic hyponatremia.

Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Lixivaptan is another selective V2 receptor antagonist that has been studied in heart failure patients.40 In a randomized, double‐blind, placebo‐controlled, ascending single‐dose study 42 diuretic‐requiring patients with mild‐to‐moderate heart failure patients received either placebo or doses of lixivaptan ranging from 10 mg to 400 mg. Except for patients who received the 10‐mg dose, lixivaptan produced a significant and dose‐related increase in urine volume over a 4‐hour period compared with placebo. Over 24 hours increases in urine volume were greater with lixivaptan than with placebo and these increases were accompanied by significant increases in solute‐free water excretion. At higher doses of lixivaptan, serum sodium levels increased significantly. The drug was tolerated in these patients and side effects tended to be mild. The Treatment of Hyponatremia Based on Lixivaptan in NYHA Class III/IV Cardiac Patient Evaluation (BALANCE Study) is an on‐going trial that is designed to evaluate whether lixivaptan is an effective and safe agent for increasing serum sodium in heart failure patients who are volume overloaded and have hyponatremia. The secondary end‐points of the BALANCE study include all‐cause mortality, CV effects, HF hospitalization and acute change in body weight.

The combined V1A and V2 receptor antagonist conivaptan has been approved by the FDA for the treatment of euvolemic and hypervolemic hyponatremia. The acute hemodynamic effects were studied in 142 NYHA class III and IV heart failure patients. Administration of 20 mg or 40 mg of conivaptan significantly reduced pulmonary artery wedge and right atrial pressures during the 3‐hour to 6‐hour interval after intravenous administration and significantly increased urine output in a dose‐dependent manner during the first 4 hours after the dose.41 In another study 170 patients hospitalized for worsening heart failure were randomly assigned to treatment with conivaptan (20‐mg loading dose followed by 2 successive 24‐hour continuous infusions of 40, 80, or 120 mg/day) or placebo in addition to their standard therapy.42 At 24 hours each dose of conivaptan had increased urine output significantly more than placebo with the difference averaging 1.0 to 1.5 L. The mean increase in serum sodium at 24, 48, and 72 hours was significantly higher in each of the conivaptan groups compared with the placebo group. At 48 hours, conivaptan increased serum sodium by 2.25 mmol/L to 3.27 mmol/L more than placebo. Conivaptan was well tolerated in these hospitalized heart failure patients. Infusion‐site reactions for this drug which is given intravenously were the most common adverse event and administration of the drug was not associated with clinically important changes in vital signs, electrolyte disturbances, or cardiac rhythm.

The effects of conivaptan on serum sodium levels were evaluated in 84 hospitalized patients with euvolemic or hypervolemic hyponatremia defined as a serum sodium between 115 mEq/ to 129 mEq/L.43 These patients received either intravenous placebo or conivaptan administered as a 30‐minute, 20‐mg loading dose followed by a 96‐hour infusion of either 40 mg/day or 80 mg/day. The results which are depicted in Figure 4 show that both conivaptan doses were associated with highly significant increases under the sodium‐time curve during the 4‐day treatment. From baseline to the end of treatment, serum sodium increased by 0.8 0.8 mEq/L with placebo as compared to 6.3 0.7 mEq/L and 9.4 0.8 mEq/L with the 40 mg and 80 mg doses of conivaptan. Conivaptan was generally well tolerated, although infusion‐site reactions led to the withdrawal of 1 (3%) and 4 (15%) of patients given conivaptan 40 mg/day and 80 mg/day, respectively.

Figure 4
(a) Mean serum [Na ] and (b) mean change (LS) from baseline in serum [Na ] at baseline (hour 0) and each measurement time. T bars indicate SE. *P = 0.025; †P = 0.034; ‡P = 0.002; §P = 0.008; ∥P < 0.001.

The overall safety profile of the vaptans has been good. Most of the adverse effects including thirst, dry mouth and others have been minor and these agents, in general, have only minimal effects on blood pressure and renal function. In addition, the long‐term safety and tolerability of tolvaptan was demonstrated in the EVEREST trial. One theoretical concern about the use of vaptans to treat hyponatremia is that rapid correction of hyponatremia at a rate of >12 mEq/L over 24 hours can cause osmotic demyelination of brain structures with severe neurologic consequences. It has been advised that in susceptible patients (including those with severe malnutrition, alcoholism, or advanced liver disease) that sodium levels be corrected at a lower rate. It is also recommended that the drugs be initiated in hospital and that serum sodium is monitored during treatment.

Conclusions

Hyponatremia is common in heart failure patients, particularly during periods of decompensation. The presence of hyponatremia has been associated with a substantial increase in risk for longer hospitalization stay and mortality both in the hospital and following discharge. Hyponatremia has also been associated with alterations in cognitive and neuromuscular function which could further impair heart failure patients, particularly those who are elderly. The use of AVP receptor antagonists to treat hyponatremia is based on evidence that this peptide which regulates the flow of free water in the distal portion of the nephron is inappropriately elevated in heart failure patients. Administration of AVP receptor antagonists has been shown to increase and improve free water excretion and increase serum sodium levels in both euvolemic and volume overloaded hyponatremic patients. In addition to their favorable effects on serum sodium levels, the AVP receptor blockers have been shown to improve hemodynamics acutely and to increase weight loss in heart failure patients. There is some evidence that they also improve symptoms in hospitalized patients and that correction of hyponatremia is associated with improved cognitive and neuromuscular function. Currently available evidence, however, does not support a beneficial effect on long‐term outcomes such as mortality or CV hospitalizations. Additional on‐going clinical trials will provide further insights into this critical question. The overall side effect profile of the vaptans is favorable and published studies document the long‐term safety of administration of tolvaptan in heart failure patients. Thus, these agents represent an important new approach for treating hyponatremia in heart failure patients. They deserve consideration for use when hyponatremia is present during an episode of decompensated heart failure.

References
  1. Schocken DD,Benjamin EJ,Fonarow GC, et al.Prevention of heart failure: a scientific statement from the American Heart Association Councils on Epidemiology and Prevention, Clinical Cardiology, Cardiovascular Nursing, and High Blood Pressure Research; Quality of Care and Outcomes Research Interdisciplinary Working Group; and Functional Genomics and Translational Biology Interdisciplinary Working Group.Circulation.2008;117:25442565.
  2. Rouleau JL,Moye LA,de Champlain J, et al.Activation of neurohumoral systems following acute myocardial infarction.Am J Cardiol.1991;68:80D86D.
  3. Dostal DE,Baker KM.The cardiac renin‐angiotensin system: conceptual, or a regulator of cardiac function?Circ Res.1999;85:643650.
  4. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators.N Engl J Med.1991;325:293302.
  5. Granger CB,McMurray JJ,Yusuf S, et al.Effects of candesartan in patients with chronic heart failure and reduced left‐ventricular systolic function intolerant to angiotensin‐converting‐enzyme inhibitors: the CHARM‐Alternative trial.Lancet.2003;362:772776.
  6. Pfeffer MA,Braunwald E,Moye LA, et al.Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction. Results of the survival and ventricular enlargement trial. The SAVE Investigators [see comments].N Engl J Med.1992;327:669677.
  7. Packer M,Bristow MR,Cohn JN, et al.The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group.N Engl J Med.1996;334:13491355.
  8. Pitt B,Zannad F,Remme WJ, et al.The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators.N Engl J Med.1999;341:709717.
  9. Pitt B,Remme W,Zannad F, et al.Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction.N Engl J Med.2003;348:13091321.
  10. Fonarow GC,Abraham WT,Albert NM, et al.Factors identified as precipitating hospital admissions for heart failure and clinical outcomes: findings from OPTIMIZE‐HF.Arch Intern Med.2008;168:847854.
  11. Fonarow GC,Adams KF,Abraham WT,Yancy CW,Boscardin WJ.Risk stratification for in‐hospital mortality in acutely decompensated heart failure: classification and regression tree analysis.JAMA.2005;293:572580.
  12. Forman DE,Butler J,Wang Y, et al.Incidence, predictors at admission, and impact of worsening renal function among patients hospitalized with heart failure.J Am Coll Cardiol.2004;43:6167.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Systolic blood pressure at admission, clinical characteristics, and outcomes in patients hospitalized with acute heart failure.JAMA.2006;296:22172226.
  14. Lucas C,Johnson W,Hamilton MA, et al.Freedom from congestion predicts good survival despite previous class IV symptoms of heart failure.Am Heart J.2000;140:840847.
  15. Maisel A,Mueller C,Adams K, et al.State of the art: using natriuretic peptide levels in clinical practice.Eur J Heart Fail.2008;10:824839.
  16. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28:980988.
  17. Gheorghiade M,Gattis WA,O'Connor CM, et al.Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial.JAMA.2004;291:19631971.
  18. Klein L,O'Connor CM,Leimberger JD, et al.Lower serum sodium is associated with increased short‐term mortality in hospitalized patients with worsening heart failure: results from the Outcomes of a Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) study.Circulation.2005;111:24542460.
  19. Gheorghiade M,Rossi JS,Cotts W, et al.Characterization and prognostic value of persistent hyponatremia in patients with severe heart failure in the ESCAPE Trial.Arch Intern Med.2007;167:19982005.
  20. Lee WH,Packer M.Prognostic importance of serum sodium concentration and its modification by converting‐enzyme inhibition in patients with severe chronic heart failure.Circulation.1986;73:257267.
  21. Kearney MT,Fox KA,Lee AJ, et al.Predicting death due to progressive heart failure in patients with mild‐to‐moderate chronic heart failure.J Am Coll Cardiol.2002;40:18011808.
  22. Krumholz HM,Chen YT,Bradford WD,Cerese J.Variations in and correlates of length of stay in academic hospitals among patients with heart failure resulting from systolic dysfunction.Am J Manag Care.1999;5:715723.
  23. Pereira AF,Simoes do CF,de MA.The use of laboratory tests in patients with mild cognitive impairment.J Alzheimers Dis.2006;10:5358.
  24. Renneboog B,Musch W,Vandemergel X,Manto MU,Decaux G.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119:7178.
  25. Sica DA.Hyponatremia and heart failure‐‐pathophysiology and implications.Congest Heart Fail.2005;11:274277.
  26. Francis GS,Benedict C,Johnstone DE et al.Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD).Circulation.1990;82:17241729.
  27. Schuster VL,Kokko JP,Jacobson HR.Angiotensin II directly stimulates sodium transport in rabbit proximal convoluted tubules.J Clin Invest.1984;73:507515.
  28. Goldsmith SR,Francis GS,Cowley AW.Arginine vasopressin and the renal response to water loading in congestive heart failure.Am J Cardiol.1986;58:295299.
  29. Schrier RW,Berl T,Anderson RJ.Osmotic and nonosmotic control of vasopressin release.Am J Physiol.1979;236:F321F332.
  30. Verbalis JG.Vasopressin V2 receptor antagonists.J Mol Endocrinol.2002;29:19.
  31. Nielsen S,Kwon TH,Christensen BM,Promeneur D,Frokiaer J,Marples D.Physiology and pathophysiology of renal aquaporins.J Am Soc Nephrol.1999;10:647663.
  32. Sonnenblick M,Friedlander Y,Rosin AJ.Diuretic‐induced severe hyponatremia. Review and analysis of 129 reported patients.Chest.1993;103:601606.
  33. Felker GM,Benza RL,Chandler AB, et al.Heart failure etiology and response to milrinone in decompensated heart failure: results from the OPTIME‐CHF study.J Am Coll Cardiol.2003;41:9971003.
  34. Packer M,Medina N,Yushak M.Correction of dilutional hyponatremia in severe chronic heart failure by converting‐enzyme inhibition.Ann Intern Med.1984;100:782789.
  35. Birnbaumer M.Vasopressin receptors.Trends Endocrinol Metab.2000;11:406410.
  36. Gheorghiade M,Niazi I,Ouyang J, et al.Vasopressin V2‐receptor blockade with tolvaptan in patients with chronic heart failure: results from a double‐blind, randomized trial.Circulation.2003;107:26902696.
  37. Gheorghiade M,Konstam MA,Burnett JC, et al.Short‐term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials.JAMA.2007;297:13321343.
  38. Konstam MA,Gheorghiade M,Burnett JC, et al.Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial.JAMA.2007;297:13191331.
  39. Schrier RW,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  40. Abraham WT,Shamshirsaz AA,McFann K,Oren RM,Schrier RW.Aquaretic effect of lixivaptan, an oral, non‐peptide, selective V2 receptor vasopressin antagonist, in New York Heart Association functional class II and III chronic heart failure patients.J Am Coll Cardiol.2006;47:16151621.
  41. Udelson JE,Smith WB,Hendrix GH, et al.Acute hemodynamic effects of conivaptan, a dual V(1A) and V(2) vasopressin receptor antagonist, in patients with advanced heart failure.Circulation.2001;104:24172423.
  42. Goldsmith SR,Elkayam U,Haught WH,Barve A,He W.Efficacy and safety of the vasopressin V1A/V2‐receptor antagonist conivaptan in acute decompensated heart failure: a dose‐ranging pilot study.J Card Fail.2008;14:641647.
  43. Zeltser D,Rosansky S,van RH,Verbalis JG,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
Article PDF
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S33-S39
Legacy Keywords
arginine vasopressin receptor antagonists, common electrolyte disorders, heart failure, hyponatremia, pharmaceuticals
Sections
Article PDF
Article PDF

Heart failure is a common and growing problem in both industrialized and developing nations. In the U.S. alone there are estimated to be well over 5 million heart failure patients and that number is expected to double over the next few decades. There are several reasons for this pandemic, most notably the aging of the world's population, a rising incidence of heart failure risk factors including hypertension, diabetes and obesity, and improved survival post‐myocardial infarction (MI).1 Greater longevity of patients with existing heart failure as a result of treatment with drugs and devices that lower mortality and, in developing nations, a reduction in premature mortality from infectious diseases have also contributed to the increase in heart failure prevalence. Although there have been important advances in treating heart failure that have improved outcomes over the past several decades, morbidity and mortality remain unacceptably high and quality of life is substantially reduced. Thus, there is considerable need for finding new approaches for managing patients with this condition.

The Role of Neurohormonal Blockade in the Treatment of Heart Failure Patients

The pathophysiology of heart failure is complex. In patients who develop systolic dysfunction, the pathway initially involves injury to the heart and/or increases in wall stress which activates a variety of compensatory responses in an effort to reestablish homeostasis within the cardiovascular (CV) system. Many of these responses are mediated by neurohormonal systems that are stimulated both systemically and locally within the heart itself.2, 3 While this widespread neurohormonal activation has some short‐term benefits in maintaining cardiac performance, there is clear evidence that it has adverse effects when maintained over time. The deleterious effects of neurohormonal activation in heart failure include excess salt and water retention, constriction of arterial resistance and venous capacitance vessels, increased load on the heart, electrolyte abnormalities and maladaptive cardiac remodeling. The critical role of neurohormonal activation in the pathogenesis and progression of heart failure has been confirmed by the results of large scale clinical trials which show that neurohormonal blocking agents such as angiotensin converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs), beta blockers (BBs), and aldosterone blockers greatly reduce morbidity and mortality and result in a variety of other favorable effects in heart failure patients.49 Based on their profound effects on outcomes, strategies that target neurohormonal activation have emerged over the past 2 decades as the cornerstone of medical management of heart failure.

Establishing Risk in Heart Failure

Although there have been impressive gains in reducing morbidity and mortality in heart failure patients over the past 3 decades, the overall clinical course remains unfavorable in a substantial portion of this population. A wide variety of risk factors which identify patients who are more likely to do poorly in the future have been identified. These include demographic variables (eg, age), functional and structural abnormalities, hemodynamic measurements, symptomatic status, exercise capacity, quality of life, presence of comorbidities and a myriad of blood tests and biomarkers. Amongst the plethora of risk factors for poor outcome, decompensation of heart failure which results in hospitalization has been recognized as 1 of the most important prognostic indicators. The Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients With Heart Failure (OPTIMIZE‐HF) Registry which included a large fairly representative population of heart failure patients from throughout the U.S. followed a subset of patients for 60 days to 90 days immediately post‐discharge from a hospitalization that was associated with decompensated heart failure.10 Over this relatively short time period hospital readmission rate was over 30% and mortality over 9%. Thus, within 2 months to 3 months of discharge following an episode of decompensation 40% of heart failure patients had either died or were back in the hospital. Among the many risk factors that have been used to predict morbidity and mortality outcomes either during or following hospitalization, the ones that appear to be the most powerful in detecting patients who are likely to do poorly are impaired renal function,11, 12 low systolic blood pressure,13 persistence of congestion at the time of hospital discharge,14 elevation of various biomarkers such as B‐type natriuretic peptide (BNP)15 and the presence of hyponatremia.16

Hyponatremia in Heart Failure

Incidence of Hyponatremia in Heart Failure Patients

Determining the exact incidence of hyponatremia in heart failure patients has been challenging due to differences in the populations that have been studied and in the criteria used to define hyponatremia. The Acute and Chronic Therapeutic Impact of a Vasopressin Antagonist in Congestive Heart Failure (ACTIV in CHF) study reported that 21.3% of the cohort of hospitalized patients had a serum sodium below 136 mmol/L.17 Higher incidences were reported in the Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) which found that 27% of patients had serum sodium concentrations between 132 mmol/L to 135 mmol/L.18 The Evaluation Study of CHF and Pulmonary Artery Catheter Effectiveness (ESCAPE) trial reported that 18% of patients had hyponatremia defined as serum sodium concentration below 134 mmol/L.19 Data from OPTIMIZE‐HF, a registry that captured data from a large cohort of representative patients who were hospitalized with decompensated heart failure indicate that such patients have a wide distribution of admission sodium values (Figure 1).16 Overall, 19.7% of the OPTIIMIZE‐HF patients had values below 135 mmol/L.

Figure 1
Distribution of admission serum sodium in patients hospitalized with a primary discharge diagnosis of heart failure.

Risk Associated With Hyponatremia

There is considerable evidence that hyponatremia is associated with increased risk for poor outcomes in heart failure patients. One of the first reports that related hyponatremia to a poor prognosis came from Lee and Packer who analyzed 30 clinical, hemodynamic, and biochemical variables in outpatients with severe heart failure.20 Their results showed that serum sodium was the most powerful predictor of CV mortality. Similar findings have been reported by other investigators.21 In patients hospitalized for decompensated heart failure, the presence of hyponatremia has been shown to be an independent predictor of longer duration of stay.22 Results from the OPTIMIZE‐HF Registry confirmed the adverse impact of hyponatremia on length of stay during a heart failure hospitalization and also suggested that low serum sodium was associated with significantly higher in‐hospital and post‐discharge mortality rates.16 In this large cohort of hospitalized patients with decompensated heart failure, each 3 mmol/L decrease in serum sodium below 140 mmol/L increased the risk of in‐hospital and follow‐up mortality by 19.5% and 10%, respectively. This association is depicted in Figure 2. A similar adverse impact of hyponatremia on post‐discharge outcomes was seen in the results of the ACTIV in CHF study.17 Overall, 69 patients out of a total of 319 who were included in the study (21.6%) had a serum sodium that was <136 mmol/L. Mortality over a 60 day period of follow‐up was 14.5% in the hyponatremic patients compared to 4% in the 250 patients whose serum sodium was 136 mmol/L.

Figure 2
Relationship between admission serum sodium level and in‐hospital mortality. Restrictive cubic spline transformation plot with 95% confidence intervals is shown.

Although these data present a powerful argument that hyponatremia is a potent risk factor for poor outcomes in heart failure patients both during and following hospitalization, they do not determine whether or not the relationship is causal. It is possible that the poor outcomes that have been observed in hyponatremic patients are related to an association between low serum sodium levels and the more profound alterations in hemodynamics, neurohormonal activation and renal function that are seen in patients with advanced heart failure. Alternatively, increased edema in vital organs including the heart in hyponatremic patients could further impair already tenuous function and contribute to a downward spiral in the clinical course. The impact of hyponatremia in limiting the use of loop diuretics and spironolactone may also be associated with a less favorable long‐term outcome. The presence of hyponatremia may also contribute to a poor outcome as a result of effects on cognitive and neuromuscular function.23, 24 Impaired cognition could adversely affect compliance with the medical regimen while neuromuscular problems related to hyponatremia could contribute to an increased incidence of falls and other traumatic injuries that older patients with chronic diseases are already at high risk of experiencing.

Pathophysiology of Hyponatremia

The pathophysiology of hyponatremia in heart failure patients involves several different processes.25 Overall, heart failure is characterized by retention of excessive amounts of salt and water in the body. Sodium retention is related to decreased renal perfusion that is caused by the effects of reduced cardiac output, decreased renal perfusion pressure and increased afferent glomerular arteriolar resistance. A reduction in glomerular filtration and increased reabsorption of sodium and water in the proximal renal tubules leads to a reduction in the delivery of water and solute to the diluting segment of the nephron. In patients with heart failure the renin‐angiotensin system (RAS) is activated, an event that is further stimulated by the administration of loop diuretics.26 Angiotensin II (Ang II), a key effector molecule of the RAS, increases tone in renal efferent arterioles. This tends to enhance both sodium and water reabsorption both through an increase in the glomerular filtration fraction and by direct effects on the distal tubule.27 Ang II also stimulates the thirst center of the brain both directly and through stimulation of antidiuretic hormone to promote the ingestion of excessive amounts of hypotonic fluids.25 Water reabsorption in the distal portion of the nephron is governed by arginine vasopressin (AVP). High levels of AVP are seen in patients with heart failure26, 28 and there is a significant association between serum levels of this peptide and the symptomatic state of the patient. There is evidence that in heart failure patients AVP levels are elevated disproportionally to plasma osmolality and serum sodium concentrations.28, 29 Even when serum osmolality is increased in this setting by infusion of sodium, AVP levels fail to demonstrate an appropriate reduction suggesting that mechanisms other than activation of osmoreceptors are involved. The effects of AVP in the pathogenesis of hyponatremia are significant. This peptide binds to the vasopressin‐2 (V2) receptor in the collecting duct of the kidney stimulating an increase in the second messenger cyclic AMP.30 Downstream signaling initiated by cyclic AMP leads to an increase in the number and activation of aquaporin‐2 water channels on the luminal surface of epithelial cells in the collecting tubule.31 The presence of these activated pores is necessary for water permeability in the collecting duct and leads to an increase in the reabsorption of free water. Finally, the use of diuretics in the treatment of heart failure has been implicated in the development and worsening of the hyponatremic state.32

Treatment of Hyponatremia

Treatment options for dealing with hyponatremia in heart failure patients have been limited until recently. Since low cardiac output and/or diminished renal perfusion are involved, interventions which improve cardiac and renal function can reverse hyponatremia. While this can be accomplished by the use of inotropic agents, the use of drugs such as dobutamine, milrinone, and other inotropes in either stable or decompensated heart failure patients with adequate tissue perfusion is not routinely recommended due to a well‐documented increase in adverse effects, particularly in patients with coronary artery disease.33 The use of hypertonic saline is also not recommended since it may worsen the extent of volume overload in decompensated patients. Fluid restriction can be used to treat hyponatremia, particularly in hospitalized patients where stricter control on input is possible. Hyponatremic patients, however, often experience excessive thirst and restriction to less than 1000 cc to 1500 cc is rarely, if ever, successful for more than a brief period of time. The use of angiotensin converting enzyme (ACE) inhibitors has been associated with an improvement in serum sodium levels. Packer et al. treated a cohort of heart failure patients who were receiving a stable dose of diuretic with an ACE inhibitor captopril and found that over a 2‐week period the serum sodium increased from 131.2 0.5 to 135.9 0.5 mmol/L; P < 0.001).34 These investigators concluded that the RAS was involved in the pathogenesis of hyponatremia and that ACEIs increased sodium levels in hyponatremic patients, though the mechanism through which this occurs has not been delineated.

The Use of Vaptans in Treating Hyponatremia

AVP actions are mediated by an interaction of the peptide with a series of receptors located on cells throughout the body. Vaptans are nonpeptidergic agents which block the interaction of AVP with these receptors; they are classified according to which receptor subtype they affect. As mentioned earlier, activation of the V2 receptor on renal tubular cells increases collecting duct permeability to water and leads to reabsorption of free water.35 The V1A receptor is located on vascular smooth muscle cells where it mediates an increase in vasomotor tone. V1A receptors are also found on platelets and in the myometrium where they mediate aggregation and uterine contraction, respectively. Some of the AVP antagonists (eg, conivaptan) block both the V1A and V2 receptors while others (eg, tolvaptan and lixivaptan) are selective for the V2 receptor.

Tolvaptan, a V2 selective agent, has been studied extensively in heart failure as well as in patients with hyponatremia due to a variety of causes. One of the initial studies was performed in a group of 254 heart failure patients who were randomized to receive tolvaptan in doses ranging from 30 mg to 60 mg daily or placebo.36 Tolvaptan at all doses studied was associated with significant reductions in body weight and improvement in the signs and symptoms of heart failure. All doses were also associated with an increase in serum sodium levels in this study. Patients who were hyponatremic made up 28% of the study population and these patients experienced the greatest increase in serum sodium. Of note was the fact that as early as day 1 in the study 80% of tolvaptan (as opposed to 40% of placebo) patients had normalization of their serum sodium levels. These effects occurred without significant changes in blood pressure or renal function and the major side effects that were seen were polyuria, dry mouth, and thirst. This study was followed by the ACTIV in CHF trial which included a slightly larger population of 319 patients (of whom 21.3% were hyponatremic at baseline) who were hospitalized due to decompensated heart failure.17 Mean body weight decreased significantly more in patients treated with tolvaptan compared to those who received placebo. Tolvaptan‐treated patients also experienced increases in serum sodium that were greatest in the patients who were hyponatremic at baseline. These changes persisted throughout the duration of the study. On post hoc analysis, event‐free survival tended to be longer for the combined group of patients treated with tolvaptan compared to placebo but there were no differences in the rate of rehospitalization or unscheduled visits for heart failure. As in the initial study, tolvaptan was well tolerated, with dry mouth being the main side effect. There were no significant hemodynamic or renal effects.

The efficacy of vasopressin antagonism in heart failure outcome study with tolvaptan (EVEREST) study randomized 4133 patients hospitalized for decompensated heart failure to receive either tolvaptan 30 mg daily or placebo in addition to their standard therapy. The short‐term goal of the study was to assess the effects of therapy on a composite end‐point of patient assessed global clinical features and weight loss on day 7 (or at the time of hospital discharge) after starting treatment.37 The results of EVEREST demonstrated that patients treated with tolvaptan had greater improvement in the composite primary end‐point. This effect was driven by a greater reduction in body weight with active drug. Although, changes in global clinical status did not differ between the study groups, tolvaptan‐treated patients reported significantly greater improvement in dyspnea at day 1. In 1 (but not the other of the 2 component trials of EVEREST) there was also an improvement in edema. At day 1 and at discharge, the tolvaptan group with hyponatremia (defined as a serum sodium below 134 mEq/L) demonstrated significantly greater increases in serum sodium than in the hyponatremic placebo treated patients. Tolvaptan was well tolerated and serious adverse event frequencies were similar between groups, without excess renal failure or hypotension.

Patients who were enrolled in EVEREST were then followed for an average of 9.9 months on tolvaptan or placebo in order to assess the effects of treatment on the dual primary endpoints of all‐cause mortality (both superiority and noninferiority) and CV death or heart failure hospitalization.38 The results demonstrated no significant differences in either primary or secondary morbidity and mortality outcomes between tolvaptan and placebo treated patients. In the EVEREST patients with baseline serum sodium levels less than 134 mEq/L, there was a significant increase of 5.49 mEq/L 5.77 mEq/L (mean SD) at day 7 or discharge, if earlier, with tolvaptan, compared with 1.85 mEq/L 5.10 mEq/L in the placebo group. This effect was observed as early as day 1 and was maintained throughout the 40 weeks of treatment. Side effects were minimal. Overall, tolvaptan increased thirst and dry mouth, but the frequencies of major adverse events were similar in the 2 groups.

Two parallel multicenter, randomized, double‐blind, placebo‐controlled trials, termed collectively the Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2), examined the effect of tolvaptan on hypervolemic and euvolemic hyponatremia of diverse causes.39 The 448 patients included in the 2 studies were randomly assigned to either placebo or tolvaptan starting at a dose of 15 mg daily (increasing to 30 mg and then 60 mg if needed, depending on serum sodium concentrations) and followed over a 30 day period. The population included 138 patients (31%) with chronic heart failure as the cause of hyponatremia with the remainder of the population divided between patients with cirrhosis or syndrome of inappropriate antidiuretic hormone hypersecretion (SIADH) and other causes of hyponatremia. The 2 primary end points for all patients were the change in the average daily area under the curve for the serum sodium concentration from baseline to day 4 and the change from baseline to day 30. As shown in Figure 3, serum sodium concentrations increased significantly more in the tolvaptan group than in the placebo group during the first 4 days and after the full 30 days of therapy. A planned analysis of the SALT trials demonstrated that correction of hyponatremia with tolvaptan was associated with significant improvement in self‐reported mental status, particularly in patients with marked hyponatremia or SIADH. Improvements in mental health scores were positively correlated with changes in serum [Na+] in both the tolvaptan and placebo groups and reversed after cessation of therapy, suggeting that hyponatremia‐associated impairments in mental function can be significantly improved by raising the serum [Na+]. The major side effects with tolvaptan included increased thirst, dry mouth, and increased urination. Tolvaptan has been approved by the US Food and Drug Administration (FDA) for the treatment of euvolemic and hypervolemic hyponatremia.

Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Lixivaptan is another selective V2 receptor antagonist that has been studied in heart failure patients.40 In a randomized, double‐blind, placebo‐controlled, ascending single‐dose study 42 diuretic‐requiring patients with mild‐to‐moderate heart failure patients received either placebo or doses of lixivaptan ranging from 10 mg to 400 mg. Except for patients who received the 10‐mg dose, lixivaptan produced a significant and dose‐related increase in urine volume over a 4‐hour period compared with placebo. Over 24 hours increases in urine volume were greater with lixivaptan than with placebo and these increases were accompanied by significant increases in solute‐free water excretion. At higher doses of lixivaptan, serum sodium levels increased significantly. The drug was tolerated in these patients and side effects tended to be mild. The Treatment of Hyponatremia Based on Lixivaptan in NYHA Class III/IV Cardiac Patient Evaluation (BALANCE Study) is an on‐going trial that is designed to evaluate whether lixivaptan is an effective and safe agent for increasing serum sodium in heart failure patients who are volume overloaded and have hyponatremia. The secondary end‐points of the BALANCE study include all‐cause mortality, CV effects, HF hospitalization and acute change in body weight.

The combined V1A and V2 receptor antagonist conivaptan has been approved by the FDA for the treatment of euvolemic and hypervolemic hyponatremia. The acute hemodynamic effects were studied in 142 NYHA class III and IV heart failure patients. Administration of 20 mg or 40 mg of conivaptan significantly reduced pulmonary artery wedge and right atrial pressures during the 3‐hour to 6‐hour interval after intravenous administration and significantly increased urine output in a dose‐dependent manner during the first 4 hours after the dose.41 In another study 170 patients hospitalized for worsening heart failure were randomly assigned to treatment with conivaptan (20‐mg loading dose followed by 2 successive 24‐hour continuous infusions of 40, 80, or 120 mg/day) or placebo in addition to their standard therapy.42 At 24 hours each dose of conivaptan had increased urine output significantly more than placebo with the difference averaging 1.0 to 1.5 L. The mean increase in serum sodium at 24, 48, and 72 hours was significantly higher in each of the conivaptan groups compared with the placebo group. At 48 hours, conivaptan increased serum sodium by 2.25 mmol/L to 3.27 mmol/L more than placebo. Conivaptan was well tolerated in these hospitalized heart failure patients. Infusion‐site reactions for this drug which is given intravenously were the most common adverse event and administration of the drug was not associated with clinically important changes in vital signs, electrolyte disturbances, or cardiac rhythm.

The effects of conivaptan on serum sodium levels were evaluated in 84 hospitalized patients with euvolemic or hypervolemic hyponatremia defined as a serum sodium between 115 mEq/ to 129 mEq/L.43 These patients received either intravenous placebo or conivaptan administered as a 30‐minute, 20‐mg loading dose followed by a 96‐hour infusion of either 40 mg/day or 80 mg/day. The results which are depicted in Figure 4 show that both conivaptan doses were associated with highly significant increases under the sodium‐time curve during the 4‐day treatment. From baseline to the end of treatment, serum sodium increased by 0.8 0.8 mEq/L with placebo as compared to 6.3 0.7 mEq/L and 9.4 0.8 mEq/L with the 40 mg and 80 mg doses of conivaptan. Conivaptan was generally well tolerated, although infusion‐site reactions led to the withdrawal of 1 (3%) and 4 (15%) of patients given conivaptan 40 mg/day and 80 mg/day, respectively.

Figure 4
(a) Mean serum [Na ] and (b) mean change (LS) from baseline in serum [Na ] at baseline (hour 0) and each measurement time. T bars indicate SE. *P = 0.025; †P = 0.034; ‡P = 0.002; §P = 0.008; ∥P < 0.001.

The overall safety profile of the vaptans has been good. Most of the adverse effects including thirst, dry mouth and others have been minor and these agents, in general, have only minimal effects on blood pressure and renal function. In addition, the long‐term safety and tolerability of tolvaptan was demonstrated in the EVEREST trial. One theoretical concern about the use of vaptans to treat hyponatremia is that rapid correction of hyponatremia at a rate of >12 mEq/L over 24 hours can cause osmotic demyelination of brain structures with severe neurologic consequences. It has been advised that in susceptible patients (including those with severe malnutrition, alcoholism, or advanced liver disease) that sodium levels be corrected at a lower rate. It is also recommended that the drugs be initiated in hospital and that serum sodium is monitored during treatment.

Conclusions

Hyponatremia is common in heart failure patients, particularly during periods of decompensation. The presence of hyponatremia has been associated with a substantial increase in risk for longer hospitalization stay and mortality both in the hospital and following discharge. Hyponatremia has also been associated with alterations in cognitive and neuromuscular function which could further impair heart failure patients, particularly those who are elderly. The use of AVP receptor antagonists to treat hyponatremia is based on evidence that this peptide which regulates the flow of free water in the distal portion of the nephron is inappropriately elevated in heart failure patients. Administration of AVP receptor antagonists has been shown to increase and improve free water excretion and increase serum sodium levels in both euvolemic and volume overloaded hyponatremic patients. In addition to their favorable effects on serum sodium levels, the AVP receptor blockers have been shown to improve hemodynamics acutely and to increase weight loss in heart failure patients. There is some evidence that they also improve symptoms in hospitalized patients and that correction of hyponatremia is associated with improved cognitive and neuromuscular function. Currently available evidence, however, does not support a beneficial effect on long‐term outcomes such as mortality or CV hospitalizations. Additional on‐going clinical trials will provide further insights into this critical question. The overall side effect profile of the vaptans is favorable and published studies document the long‐term safety of administration of tolvaptan in heart failure patients. Thus, these agents represent an important new approach for treating hyponatremia in heart failure patients. They deserve consideration for use when hyponatremia is present during an episode of decompensated heart failure.

Heart failure is a common and growing problem in both industrialized and developing nations. In the U.S. alone there are estimated to be well over 5 million heart failure patients and that number is expected to double over the next few decades. There are several reasons for this pandemic, most notably the aging of the world's population, a rising incidence of heart failure risk factors including hypertension, diabetes and obesity, and improved survival post‐myocardial infarction (MI).1 Greater longevity of patients with existing heart failure as a result of treatment with drugs and devices that lower mortality and, in developing nations, a reduction in premature mortality from infectious diseases have also contributed to the increase in heart failure prevalence. Although there have been important advances in treating heart failure that have improved outcomes over the past several decades, morbidity and mortality remain unacceptably high and quality of life is substantially reduced. Thus, there is considerable need for finding new approaches for managing patients with this condition.

The Role of Neurohormonal Blockade in the Treatment of Heart Failure Patients

The pathophysiology of heart failure is complex. In patients who develop systolic dysfunction, the pathway initially involves injury to the heart and/or increases in wall stress which activates a variety of compensatory responses in an effort to reestablish homeostasis within the cardiovascular (CV) system. Many of these responses are mediated by neurohormonal systems that are stimulated both systemically and locally within the heart itself.2, 3 While this widespread neurohormonal activation has some short‐term benefits in maintaining cardiac performance, there is clear evidence that it has adverse effects when maintained over time. The deleterious effects of neurohormonal activation in heart failure include excess salt and water retention, constriction of arterial resistance and venous capacitance vessels, increased load on the heart, electrolyte abnormalities and maladaptive cardiac remodeling. The critical role of neurohormonal activation in the pathogenesis and progression of heart failure has been confirmed by the results of large scale clinical trials which show that neurohormonal blocking agents such as angiotensin converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs), beta blockers (BBs), and aldosterone blockers greatly reduce morbidity and mortality and result in a variety of other favorable effects in heart failure patients.49 Based on their profound effects on outcomes, strategies that target neurohormonal activation have emerged over the past 2 decades as the cornerstone of medical management of heart failure.

Establishing Risk in Heart Failure

Although there have been impressive gains in reducing morbidity and mortality in heart failure patients over the past 3 decades, the overall clinical course remains unfavorable in a substantial portion of this population. A wide variety of risk factors which identify patients who are more likely to do poorly in the future have been identified. These include demographic variables (eg, age), functional and structural abnormalities, hemodynamic measurements, symptomatic status, exercise capacity, quality of life, presence of comorbidities and a myriad of blood tests and biomarkers. Amongst the plethora of risk factors for poor outcome, decompensation of heart failure which results in hospitalization has been recognized as 1 of the most important prognostic indicators. The Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients With Heart Failure (OPTIMIZE‐HF) Registry which included a large fairly representative population of heart failure patients from throughout the U.S. followed a subset of patients for 60 days to 90 days immediately post‐discharge from a hospitalization that was associated with decompensated heart failure.10 Over this relatively short time period hospital readmission rate was over 30% and mortality over 9%. Thus, within 2 months to 3 months of discharge following an episode of decompensation 40% of heart failure patients had either died or were back in the hospital. Among the many risk factors that have been used to predict morbidity and mortality outcomes either during or following hospitalization, the ones that appear to be the most powerful in detecting patients who are likely to do poorly are impaired renal function,11, 12 low systolic blood pressure,13 persistence of congestion at the time of hospital discharge,14 elevation of various biomarkers such as B‐type natriuretic peptide (BNP)15 and the presence of hyponatremia.16

Hyponatremia in Heart Failure

Incidence of Hyponatremia in Heart Failure Patients

Determining the exact incidence of hyponatremia in heart failure patients has been challenging due to differences in the populations that have been studied and in the criteria used to define hyponatremia. The Acute and Chronic Therapeutic Impact of a Vasopressin Antagonist in Congestive Heart Failure (ACTIV in CHF) study reported that 21.3% of the cohort of hospitalized patients had a serum sodium below 136 mmol/L.17 Higher incidences were reported in the Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) which found that 27% of patients had serum sodium concentrations between 132 mmol/L to 135 mmol/L.18 The Evaluation Study of CHF and Pulmonary Artery Catheter Effectiveness (ESCAPE) trial reported that 18% of patients had hyponatremia defined as serum sodium concentration below 134 mmol/L.19 Data from OPTIMIZE‐HF, a registry that captured data from a large cohort of representative patients who were hospitalized with decompensated heart failure indicate that such patients have a wide distribution of admission sodium values (Figure 1).16 Overall, 19.7% of the OPTIIMIZE‐HF patients had values below 135 mmol/L.

Figure 1
Distribution of admission serum sodium in patients hospitalized with a primary discharge diagnosis of heart failure.

Risk Associated With Hyponatremia

There is considerable evidence that hyponatremia is associated with increased risk for poor outcomes in heart failure patients. One of the first reports that related hyponatremia to a poor prognosis came from Lee and Packer who analyzed 30 clinical, hemodynamic, and biochemical variables in outpatients with severe heart failure.20 Their results showed that serum sodium was the most powerful predictor of CV mortality. Similar findings have been reported by other investigators.21 In patients hospitalized for decompensated heart failure, the presence of hyponatremia has been shown to be an independent predictor of longer duration of stay.22 Results from the OPTIMIZE‐HF Registry confirmed the adverse impact of hyponatremia on length of stay during a heart failure hospitalization and also suggested that low serum sodium was associated with significantly higher in‐hospital and post‐discharge mortality rates.16 In this large cohort of hospitalized patients with decompensated heart failure, each 3 mmol/L decrease in serum sodium below 140 mmol/L increased the risk of in‐hospital and follow‐up mortality by 19.5% and 10%, respectively. This association is depicted in Figure 2. A similar adverse impact of hyponatremia on post‐discharge outcomes was seen in the results of the ACTIV in CHF study.17 Overall, 69 patients out of a total of 319 who were included in the study (21.6%) had a serum sodium that was <136 mmol/L. Mortality over a 60 day period of follow‐up was 14.5% in the hyponatremic patients compared to 4% in the 250 patients whose serum sodium was 136 mmol/L.

Figure 2
Relationship between admission serum sodium level and in‐hospital mortality. Restrictive cubic spline transformation plot with 95% confidence intervals is shown.

Although these data present a powerful argument that hyponatremia is a potent risk factor for poor outcomes in heart failure patients both during and following hospitalization, they do not determine whether or not the relationship is causal. It is possible that the poor outcomes that have been observed in hyponatremic patients are related to an association between low serum sodium levels and the more profound alterations in hemodynamics, neurohormonal activation and renal function that are seen in patients with advanced heart failure. Alternatively, increased edema in vital organs including the heart in hyponatremic patients could further impair already tenuous function and contribute to a downward spiral in the clinical course. The impact of hyponatremia in limiting the use of loop diuretics and spironolactone may also be associated with a less favorable long‐term outcome. The presence of hyponatremia may also contribute to a poor outcome as a result of effects on cognitive and neuromuscular function.23, 24 Impaired cognition could adversely affect compliance with the medical regimen while neuromuscular problems related to hyponatremia could contribute to an increased incidence of falls and other traumatic injuries that older patients with chronic diseases are already at high risk of experiencing.

Pathophysiology of Hyponatremia

The pathophysiology of hyponatremia in heart failure patients involves several different processes.25 Overall, heart failure is characterized by retention of excessive amounts of salt and water in the body. Sodium retention is related to decreased renal perfusion that is caused by the effects of reduced cardiac output, decreased renal perfusion pressure and increased afferent glomerular arteriolar resistance. A reduction in glomerular filtration and increased reabsorption of sodium and water in the proximal renal tubules leads to a reduction in the delivery of water and solute to the diluting segment of the nephron. In patients with heart failure the renin‐angiotensin system (RAS) is activated, an event that is further stimulated by the administration of loop diuretics.26 Angiotensin II (Ang II), a key effector molecule of the RAS, increases tone in renal efferent arterioles. This tends to enhance both sodium and water reabsorption both through an increase in the glomerular filtration fraction and by direct effects on the distal tubule.27 Ang II also stimulates the thirst center of the brain both directly and through stimulation of antidiuretic hormone to promote the ingestion of excessive amounts of hypotonic fluids.25 Water reabsorption in the distal portion of the nephron is governed by arginine vasopressin (AVP). High levels of AVP are seen in patients with heart failure26, 28 and there is a significant association between serum levels of this peptide and the symptomatic state of the patient. There is evidence that in heart failure patients AVP levels are elevated disproportionally to plasma osmolality and serum sodium concentrations.28, 29 Even when serum osmolality is increased in this setting by infusion of sodium, AVP levels fail to demonstrate an appropriate reduction suggesting that mechanisms other than activation of osmoreceptors are involved. The effects of AVP in the pathogenesis of hyponatremia are significant. This peptide binds to the vasopressin‐2 (V2) receptor in the collecting duct of the kidney stimulating an increase in the second messenger cyclic AMP.30 Downstream signaling initiated by cyclic AMP leads to an increase in the number and activation of aquaporin‐2 water channels on the luminal surface of epithelial cells in the collecting tubule.31 The presence of these activated pores is necessary for water permeability in the collecting duct and leads to an increase in the reabsorption of free water. Finally, the use of diuretics in the treatment of heart failure has been implicated in the development and worsening of the hyponatremic state.32

Treatment of Hyponatremia

Treatment options for dealing with hyponatremia in heart failure patients have been limited until recently. Since low cardiac output and/or diminished renal perfusion are involved, interventions which improve cardiac and renal function can reverse hyponatremia. While this can be accomplished by the use of inotropic agents, the use of drugs such as dobutamine, milrinone, and other inotropes in either stable or decompensated heart failure patients with adequate tissue perfusion is not routinely recommended due to a well‐documented increase in adverse effects, particularly in patients with coronary artery disease.33 The use of hypertonic saline is also not recommended since it may worsen the extent of volume overload in decompensated patients. Fluid restriction can be used to treat hyponatremia, particularly in hospitalized patients where stricter control on input is possible. Hyponatremic patients, however, often experience excessive thirst and restriction to less than 1000 cc to 1500 cc is rarely, if ever, successful for more than a brief period of time. The use of angiotensin converting enzyme (ACE) inhibitors has been associated with an improvement in serum sodium levels. Packer et al. treated a cohort of heart failure patients who were receiving a stable dose of diuretic with an ACE inhibitor captopril and found that over a 2‐week period the serum sodium increased from 131.2 0.5 to 135.9 0.5 mmol/L; P < 0.001).34 These investigators concluded that the RAS was involved in the pathogenesis of hyponatremia and that ACEIs increased sodium levels in hyponatremic patients, though the mechanism through which this occurs has not been delineated.

The Use of Vaptans in Treating Hyponatremia

AVP actions are mediated by an interaction of the peptide with a series of receptors located on cells throughout the body. Vaptans are nonpeptidergic agents which block the interaction of AVP with these receptors; they are classified according to which receptor subtype they affect. As mentioned earlier, activation of the V2 receptor on renal tubular cells increases collecting duct permeability to water and leads to reabsorption of free water.35 The V1A receptor is located on vascular smooth muscle cells where it mediates an increase in vasomotor tone. V1A receptors are also found on platelets and in the myometrium where they mediate aggregation and uterine contraction, respectively. Some of the AVP antagonists (eg, conivaptan) block both the V1A and V2 receptors while others (eg, tolvaptan and lixivaptan) are selective for the V2 receptor.

Tolvaptan, a V2 selective agent, has been studied extensively in heart failure as well as in patients with hyponatremia due to a variety of causes. One of the initial studies was performed in a group of 254 heart failure patients who were randomized to receive tolvaptan in doses ranging from 30 mg to 60 mg daily or placebo.36 Tolvaptan at all doses studied was associated with significant reductions in body weight and improvement in the signs and symptoms of heart failure. All doses were also associated with an increase in serum sodium levels in this study. Patients who were hyponatremic made up 28% of the study population and these patients experienced the greatest increase in serum sodium. Of note was the fact that as early as day 1 in the study 80% of tolvaptan (as opposed to 40% of placebo) patients had normalization of their serum sodium levels. These effects occurred without significant changes in blood pressure or renal function and the major side effects that were seen were polyuria, dry mouth, and thirst. This study was followed by the ACTIV in CHF trial which included a slightly larger population of 319 patients (of whom 21.3% were hyponatremic at baseline) who were hospitalized due to decompensated heart failure.17 Mean body weight decreased significantly more in patients treated with tolvaptan compared to those who received placebo. Tolvaptan‐treated patients also experienced increases in serum sodium that were greatest in the patients who were hyponatremic at baseline. These changes persisted throughout the duration of the study. On post hoc analysis, event‐free survival tended to be longer for the combined group of patients treated with tolvaptan compared to placebo but there were no differences in the rate of rehospitalization or unscheduled visits for heart failure. As in the initial study, tolvaptan was well tolerated, with dry mouth being the main side effect. There were no significant hemodynamic or renal effects.

The efficacy of vasopressin antagonism in heart failure outcome study with tolvaptan (EVEREST) study randomized 4133 patients hospitalized for decompensated heart failure to receive either tolvaptan 30 mg daily or placebo in addition to their standard therapy. The short‐term goal of the study was to assess the effects of therapy on a composite end‐point of patient assessed global clinical features and weight loss on day 7 (or at the time of hospital discharge) after starting treatment.37 The results of EVEREST demonstrated that patients treated with tolvaptan had greater improvement in the composite primary end‐point. This effect was driven by a greater reduction in body weight with active drug. Although, changes in global clinical status did not differ between the study groups, tolvaptan‐treated patients reported significantly greater improvement in dyspnea at day 1. In 1 (but not the other of the 2 component trials of EVEREST) there was also an improvement in edema. At day 1 and at discharge, the tolvaptan group with hyponatremia (defined as a serum sodium below 134 mEq/L) demonstrated significantly greater increases in serum sodium than in the hyponatremic placebo treated patients. Tolvaptan was well tolerated and serious adverse event frequencies were similar between groups, without excess renal failure or hypotension.

Patients who were enrolled in EVEREST were then followed for an average of 9.9 months on tolvaptan or placebo in order to assess the effects of treatment on the dual primary endpoints of all‐cause mortality (both superiority and noninferiority) and CV death or heart failure hospitalization.38 The results demonstrated no significant differences in either primary or secondary morbidity and mortality outcomes between tolvaptan and placebo treated patients. In the EVEREST patients with baseline serum sodium levels less than 134 mEq/L, there was a significant increase of 5.49 mEq/L 5.77 mEq/L (mean SD) at day 7 or discharge, if earlier, with tolvaptan, compared with 1.85 mEq/L 5.10 mEq/L in the placebo group. This effect was observed as early as day 1 and was maintained throughout the 40 weeks of treatment. Side effects were minimal. Overall, tolvaptan increased thirst and dry mouth, but the frequencies of major adverse events were similar in the 2 groups.

Two parallel multicenter, randomized, double‐blind, placebo‐controlled trials, termed collectively the Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2 (SALT‐1 and SALT‐2), examined the effect of tolvaptan on hypervolemic and euvolemic hyponatremia of diverse causes.39 The 448 patients included in the 2 studies were randomly assigned to either placebo or tolvaptan starting at a dose of 15 mg daily (increasing to 30 mg and then 60 mg if needed, depending on serum sodium concentrations) and followed over a 30 day period. The population included 138 patients (31%) with chronic heart failure as the cause of hyponatremia with the remainder of the population divided between patients with cirrhosis or syndrome of inappropriate antidiuretic hormone hypersecretion (SIADH) and other causes of hyponatremia. The 2 primary end points for all patients were the change in the average daily area under the curve for the serum sodium concentration from baseline to day 4 and the change from baseline to day 30. As shown in Figure 3, serum sodium concentrations increased significantly more in the tolvaptan group than in the placebo group during the first 4 days and after the full 30 days of therapy. A planned analysis of the SALT trials demonstrated that correction of hyponatremia with tolvaptan was associated with significant improvement in self‐reported mental status, particularly in patients with marked hyponatremia or SIADH. Improvements in mental health scores were positively correlated with changes in serum [Na+] in both the tolvaptan and placebo groups and reversed after cessation of therapy, suggeting that hyponatremia‐associated impairments in mental function can be significantly improved by raising the serum [Na+]. The major side effects with tolvaptan included increased thirst, dry mouth, and increased urination. Tolvaptan has been approved by the US Food and Drug Administration (FDA) for the treatment of euvolemic and hypervolemic hyponatremia.

Figure 3
Mean serum sodium concentrations according to the day of patient visit in the SALT‐1 and SALT‐2 trials. Schrier RW, Gheorghiade M, Berl T, et al. Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099–2112. Copyright 2006 Massachusetts Medical Society. All rights reserved. Asterisks indicate P < 0.001 for the comparison between tolvaptan and placebo treated patients. Daggers indicate P < 0.01 for the comparison between tolvaptan and placebo. Tolvaptan was discontinued on day 30. Circles denote patients receiving tolvaptan, and squares denote patients receiving placebo. Horizontal lines indicate the lower limit of the normal range for the serum sodium concentration. Vertical lines indicate the end of the treatment period. HN denotes hyponatremia. Abbreviation: SALT‐1/SALT‐2, Study of Ascending Levels of Tolvaptan in Hyponatremia 1 and 2.

Lixivaptan is another selective V2 receptor antagonist that has been studied in heart failure patients.40 In a randomized, double‐blind, placebo‐controlled, ascending single‐dose study 42 diuretic‐requiring patients with mild‐to‐moderate heart failure patients received either placebo or doses of lixivaptan ranging from 10 mg to 400 mg. Except for patients who received the 10‐mg dose, lixivaptan produced a significant and dose‐related increase in urine volume over a 4‐hour period compared with placebo. Over 24 hours increases in urine volume were greater with lixivaptan than with placebo and these increases were accompanied by significant increases in solute‐free water excretion. At higher doses of lixivaptan, serum sodium levels increased significantly. The drug was tolerated in these patients and side effects tended to be mild. The Treatment of Hyponatremia Based on Lixivaptan in NYHA Class III/IV Cardiac Patient Evaluation (BALANCE Study) is an on‐going trial that is designed to evaluate whether lixivaptan is an effective and safe agent for increasing serum sodium in heart failure patients who are volume overloaded and have hyponatremia. The secondary end‐points of the BALANCE study include all‐cause mortality, CV effects, HF hospitalization and acute change in body weight.

The combined V1A and V2 receptor antagonist conivaptan has been approved by the FDA for the treatment of euvolemic and hypervolemic hyponatremia. The acute hemodynamic effects were studied in 142 NYHA class III and IV heart failure patients. Administration of 20 mg or 40 mg of conivaptan significantly reduced pulmonary artery wedge and right atrial pressures during the 3‐hour to 6‐hour interval after intravenous administration and significantly increased urine output in a dose‐dependent manner during the first 4 hours after the dose.41 In another study 170 patients hospitalized for worsening heart failure were randomly assigned to treatment with conivaptan (20‐mg loading dose followed by 2 successive 24‐hour continuous infusions of 40, 80, or 120 mg/day) or placebo in addition to their standard therapy.42 At 24 hours each dose of conivaptan had increased urine output significantly more than placebo with the difference averaging 1.0 to 1.5 L. The mean increase in serum sodium at 24, 48, and 72 hours was significantly higher in each of the conivaptan groups compared with the placebo group. At 48 hours, conivaptan increased serum sodium by 2.25 mmol/L to 3.27 mmol/L more than placebo. Conivaptan was well tolerated in these hospitalized heart failure patients. Infusion‐site reactions for this drug which is given intravenously were the most common adverse event and administration of the drug was not associated with clinically important changes in vital signs, electrolyte disturbances, or cardiac rhythm.

The effects of conivaptan on serum sodium levels were evaluated in 84 hospitalized patients with euvolemic or hypervolemic hyponatremia defined as a serum sodium between 115 mEq/ to 129 mEq/L.43 These patients received either intravenous placebo or conivaptan administered as a 30‐minute, 20‐mg loading dose followed by a 96‐hour infusion of either 40 mg/day or 80 mg/day. The results which are depicted in Figure 4 show that both conivaptan doses were associated with highly significant increases under the sodium‐time curve during the 4‐day treatment. From baseline to the end of treatment, serum sodium increased by 0.8 0.8 mEq/L with placebo as compared to 6.3 0.7 mEq/L and 9.4 0.8 mEq/L with the 40 mg and 80 mg doses of conivaptan. Conivaptan was generally well tolerated, although infusion‐site reactions led to the withdrawal of 1 (3%) and 4 (15%) of patients given conivaptan 40 mg/day and 80 mg/day, respectively.

Figure 4
(a) Mean serum [Na ] and (b) mean change (LS) from baseline in serum [Na ] at baseline (hour 0) and each measurement time. T bars indicate SE. *P = 0.025; †P = 0.034; ‡P = 0.002; §P = 0.008; ∥P < 0.001.

The overall safety profile of the vaptans has been good. Most of the adverse effects including thirst, dry mouth and others have been minor and these agents, in general, have only minimal effects on blood pressure and renal function. In addition, the long‐term safety and tolerability of tolvaptan was demonstrated in the EVEREST trial. One theoretical concern about the use of vaptans to treat hyponatremia is that rapid correction of hyponatremia at a rate of >12 mEq/L over 24 hours can cause osmotic demyelination of brain structures with severe neurologic consequences. It has been advised that in susceptible patients (including those with severe malnutrition, alcoholism, or advanced liver disease) that sodium levels be corrected at a lower rate. It is also recommended that the drugs be initiated in hospital and that serum sodium is monitored during treatment.

Conclusions

Hyponatremia is common in heart failure patients, particularly during periods of decompensation. The presence of hyponatremia has been associated with a substantial increase in risk for longer hospitalization stay and mortality both in the hospital and following discharge. Hyponatremia has also been associated with alterations in cognitive and neuromuscular function which could further impair heart failure patients, particularly those who are elderly. The use of AVP receptor antagonists to treat hyponatremia is based on evidence that this peptide which regulates the flow of free water in the distal portion of the nephron is inappropriately elevated in heart failure patients. Administration of AVP receptor antagonists has been shown to increase and improve free water excretion and increase serum sodium levels in both euvolemic and volume overloaded hyponatremic patients. In addition to their favorable effects on serum sodium levels, the AVP receptor blockers have been shown to improve hemodynamics acutely and to increase weight loss in heart failure patients. There is some evidence that they also improve symptoms in hospitalized patients and that correction of hyponatremia is associated with improved cognitive and neuromuscular function. Currently available evidence, however, does not support a beneficial effect on long‐term outcomes such as mortality or CV hospitalizations. Additional on‐going clinical trials will provide further insights into this critical question. The overall side effect profile of the vaptans is favorable and published studies document the long‐term safety of administration of tolvaptan in heart failure patients. Thus, these agents represent an important new approach for treating hyponatremia in heart failure patients. They deserve consideration for use when hyponatremia is present during an episode of decompensated heart failure.

References
  1. Schocken DD,Benjamin EJ,Fonarow GC, et al.Prevention of heart failure: a scientific statement from the American Heart Association Councils on Epidemiology and Prevention, Clinical Cardiology, Cardiovascular Nursing, and High Blood Pressure Research; Quality of Care and Outcomes Research Interdisciplinary Working Group; and Functional Genomics and Translational Biology Interdisciplinary Working Group.Circulation.2008;117:25442565.
  2. Rouleau JL,Moye LA,de Champlain J, et al.Activation of neurohumoral systems following acute myocardial infarction.Am J Cardiol.1991;68:80D86D.
  3. Dostal DE,Baker KM.The cardiac renin‐angiotensin system: conceptual, or a regulator of cardiac function?Circ Res.1999;85:643650.
  4. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators.N Engl J Med.1991;325:293302.
  5. Granger CB,McMurray JJ,Yusuf S, et al.Effects of candesartan in patients with chronic heart failure and reduced left‐ventricular systolic function intolerant to angiotensin‐converting‐enzyme inhibitors: the CHARM‐Alternative trial.Lancet.2003;362:772776.
  6. Pfeffer MA,Braunwald E,Moye LA, et al.Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction. Results of the survival and ventricular enlargement trial. The SAVE Investigators [see comments].N Engl J Med.1992;327:669677.
  7. Packer M,Bristow MR,Cohn JN, et al.The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group.N Engl J Med.1996;334:13491355.
  8. Pitt B,Zannad F,Remme WJ, et al.The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators.N Engl J Med.1999;341:709717.
  9. Pitt B,Remme W,Zannad F, et al.Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction.N Engl J Med.2003;348:13091321.
  10. Fonarow GC,Abraham WT,Albert NM, et al.Factors identified as precipitating hospital admissions for heart failure and clinical outcomes: findings from OPTIMIZE‐HF.Arch Intern Med.2008;168:847854.
  11. Fonarow GC,Adams KF,Abraham WT,Yancy CW,Boscardin WJ.Risk stratification for in‐hospital mortality in acutely decompensated heart failure: classification and regression tree analysis.JAMA.2005;293:572580.
  12. Forman DE,Butler J,Wang Y, et al.Incidence, predictors at admission, and impact of worsening renal function among patients hospitalized with heart failure.J Am Coll Cardiol.2004;43:6167.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Systolic blood pressure at admission, clinical characteristics, and outcomes in patients hospitalized with acute heart failure.JAMA.2006;296:22172226.
  14. Lucas C,Johnson W,Hamilton MA, et al.Freedom from congestion predicts good survival despite previous class IV symptoms of heart failure.Am Heart J.2000;140:840847.
  15. Maisel A,Mueller C,Adams K, et al.State of the art: using natriuretic peptide levels in clinical practice.Eur J Heart Fail.2008;10:824839.
  16. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28:980988.
  17. Gheorghiade M,Gattis WA,O'Connor CM, et al.Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial.JAMA.2004;291:19631971.
  18. Klein L,O'Connor CM,Leimberger JD, et al.Lower serum sodium is associated with increased short‐term mortality in hospitalized patients with worsening heart failure: results from the Outcomes of a Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) study.Circulation.2005;111:24542460.
  19. Gheorghiade M,Rossi JS,Cotts W, et al.Characterization and prognostic value of persistent hyponatremia in patients with severe heart failure in the ESCAPE Trial.Arch Intern Med.2007;167:19982005.
  20. Lee WH,Packer M.Prognostic importance of serum sodium concentration and its modification by converting‐enzyme inhibition in patients with severe chronic heart failure.Circulation.1986;73:257267.
  21. Kearney MT,Fox KA,Lee AJ, et al.Predicting death due to progressive heart failure in patients with mild‐to‐moderate chronic heart failure.J Am Coll Cardiol.2002;40:18011808.
  22. Krumholz HM,Chen YT,Bradford WD,Cerese J.Variations in and correlates of length of stay in academic hospitals among patients with heart failure resulting from systolic dysfunction.Am J Manag Care.1999;5:715723.
  23. Pereira AF,Simoes do CF,de MA.The use of laboratory tests in patients with mild cognitive impairment.J Alzheimers Dis.2006;10:5358.
  24. Renneboog B,Musch W,Vandemergel X,Manto MU,Decaux G.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119:7178.
  25. Sica DA.Hyponatremia and heart failure‐‐pathophysiology and implications.Congest Heart Fail.2005;11:274277.
  26. Francis GS,Benedict C,Johnstone DE et al.Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD).Circulation.1990;82:17241729.
  27. Schuster VL,Kokko JP,Jacobson HR.Angiotensin II directly stimulates sodium transport in rabbit proximal convoluted tubules.J Clin Invest.1984;73:507515.
  28. Goldsmith SR,Francis GS,Cowley AW.Arginine vasopressin and the renal response to water loading in congestive heart failure.Am J Cardiol.1986;58:295299.
  29. Schrier RW,Berl T,Anderson RJ.Osmotic and nonosmotic control of vasopressin release.Am J Physiol.1979;236:F321F332.
  30. Verbalis JG.Vasopressin V2 receptor antagonists.J Mol Endocrinol.2002;29:19.
  31. Nielsen S,Kwon TH,Christensen BM,Promeneur D,Frokiaer J,Marples D.Physiology and pathophysiology of renal aquaporins.J Am Soc Nephrol.1999;10:647663.
  32. Sonnenblick M,Friedlander Y,Rosin AJ.Diuretic‐induced severe hyponatremia. Review and analysis of 129 reported patients.Chest.1993;103:601606.
  33. Felker GM,Benza RL,Chandler AB, et al.Heart failure etiology and response to milrinone in decompensated heart failure: results from the OPTIME‐CHF study.J Am Coll Cardiol.2003;41:9971003.
  34. Packer M,Medina N,Yushak M.Correction of dilutional hyponatremia in severe chronic heart failure by converting‐enzyme inhibition.Ann Intern Med.1984;100:782789.
  35. Birnbaumer M.Vasopressin receptors.Trends Endocrinol Metab.2000;11:406410.
  36. Gheorghiade M,Niazi I,Ouyang J, et al.Vasopressin V2‐receptor blockade with tolvaptan in patients with chronic heart failure: results from a double‐blind, randomized trial.Circulation.2003;107:26902696.
  37. Gheorghiade M,Konstam MA,Burnett JC, et al.Short‐term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials.JAMA.2007;297:13321343.
  38. Konstam MA,Gheorghiade M,Burnett JC, et al.Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial.JAMA.2007;297:13191331.
  39. Schrier RW,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  40. Abraham WT,Shamshirsaz AA,McFann K,Oren RM,Schrier RW.Aquaretic effect of lixivaptan, an oral, non‐peptide, selective V2 receptor vasopressin antagonist, in New York Heart Association functional class II and III chronic heart failure patients.J Am Coll Cardiol.2006;47:16151621.
  41. Udelson JE,Smith WB,Hendrix GH, et al.Acute hemodynamic effects of conivaptan, a dual V(1A) and V(2) vasopressin receptor antagonist, in patients with advanced heart failure.Circulation.2001;104:24172423.
  42. Goldsmith SR,Elkayam U,Haught WH,Barve A,He W.Efficacy and safety of the vasopressin V1A/V2‐receptor antagonist conivaptan in acute decompensated heart failure: a dose‐ranging pilot study.J Card Fail.2008;14:641647.
  43. Zeltser D,Rosansky S,van RH,Verbalis JG,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
References
  1. Schocken DD,Benjamin EJ,Fonarow GC, et al.Prevention of heart failure: a scientific statement from the American Heart Association Councils on Epidemiology and Prevention, Clinical Cardiology, Cardiovascular Nursing, and High Blood Pressure Research; Quality of Care and Outcomes Research Interdisciplinary Working Group; and Functional Genomics and Translational Biology Interdisciplinary Working Group.Circulation.2008;117:25442565.
  2. Rouleau JL,Moye LA,de Champlain J, et al.Activation of neurohumoral systems following acute myocardial infarction.Am J Cardiol.1991;68:80D86D.
  3. Dostal DE,Baker KM.The cardiac renin‐angiotensin system: conceptual, or a regulator of cardiac function?Circ Res.1999;85:643650.
  4. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators.N Engl J Med.1991;325:293302.
  5. Granger CB,McMurray JJ,Yusuf S, et al.Effects of candesartan in patients with chronic heart failure and reduced left‐ventricular systolic function intolerant to angiotensin‐converting‐enzyme inhibitors: the CHARM‐Alternative trial.Lancet.2003;362:772776.
  6. Pfeffer MA,Braunwald E,Moye LA, et al.Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction. Results of the survival and ventricular enlargement trial. The SAVE Investigators [see comments].N Engl J Med.1992;327:669677.
  7. Packer M,Bristow MR,Cohn JN, et al.The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group.N Engl J Med.1996;334:13491355.
  8. Pitt B,Zannad F,Remme WJ, et al.The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators.N Engl J Med.1999;341:709717.
  9. Pitt B,Remme W,Zannad F, et al.Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction.N Engl J Med.2003;348:13091321.
  10. Fonarow GC,Abraham WT,Albert NM, et al.Factors identified as precipitating hospital admissions for heart failure and clinical outcomes: findings from OPTIMIZE‐HF.Arch Intern Med.2008;168:847854.
  11. Fonarow GC,Adams KF,Abraham WT,Yancy CW,Boscardin WJ.Risk stratification for in‐hospital mortality in acutely decompensated heart failure: classification and regression tree analysis.JAMA.2005;293:572580.
  12. Forman DE,Butler J,Wang Y, et al.Incidence, predictors at admission, and impact of worsening renal function among patients hospitalized with heart failure.J Am Coll Cardiol.2004;43:6167.
  13. Gheorghiade M,Abraham WT,Albert NM, et al.Systolic blood pressure at admission, clinical characteristics, and outcomes in patients hospitalized with acute heart failure.JAMA.2006;296:22172226.
  14. Lucas C,Johnson W,Hamilton MA, et al.Freedom from congestion predicts good survival despite previous class IV symptoms of heart failure.Am Heart J.2000;140:840847.
  15. Maisel A,Mueller C,Adams K, et al.State of the art: using natriuretic peptide levels in clinical practice.Eur J Heart Fail.2008;10:824839.
  16. Gheorghiade M,Abraham WT,Albert NM, et al.Relationship between admission serum sodium concentration and clinical outcomes in patients hospitalized for heart failure: an analysis from the OPTIMIZE‐HF registry.Eur Heart J.2007;28:980988.
  17. Gheorghiade M,Gattis WA,O'Connor CM, et al.Effects of tolvaptan, a vasopressin antagonist, in patients hospitalized with worsening heart failure: a randomized controlled trial.JAMA.2004;291:19631971.
  18. Klein L,O'Connor CM,Leimberger JD, et al.Lower serum sodium is associated with increased short‐term mortality in hospitalized patients with worsening heart failure: results from the Outcomes of a Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure (OPTIME‐CHF) study.Circulation.2005;111:24542460.
  19. Gheorghiade M,Rossi JS,Cotts W, et al.Characterization and prognostic value of persistent hyponatremia in patients with severe heart failure in the ESCAPE Trial.Arch Intern Med.2007;167:19982005.
  20. Lee WH,Packer M.Prognostic importance of serum sodium concentration and its modification by converting‐enzyme inhibition in patients with severe chronic heart failure.Circulation.1986;73:257267.
  21. Kearney MT,Fox KA,Lee AJ, et al.Predicting death due to progressive heart failure in patients with mild‐to‐moderate chronic heart failure.J Am Coll Cardiol.2002;40:18011808.
  22. Krumholz HM,Chen YT,Bradford WD,Cerese J.Variations in and correlates of length of stay in academic hospitals among patients with heart failure resulting from systolic dysfunction.Am J Manag Care.1999;5:715723.
  23. Pereira AF,Simoes do CF,de MA.The use of laboratory tests in patients with mild cognitive impairment.J Alzheimers Dis.2006;10:5358.
  24. Renneboog B,Musch W,Vandemergel X,Manto MU,Decaux G.Mild chronic hyponatremia is associated with falls, unsteadiness, and attention deficits.Am J Med.2006;119:7178.
  25. Sica DA.Hyponatremia and heart failure‐‐pathophysiology and implications.Congest Heart Fail.2005;11:274277.
  26. Francis GS,Benedict C,Johnstone DE et al.Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD).Circulation.1990;82:17241729.
  27. Schuster VL,Kokko JP,Jacobson HR.Angiotensin II directly stimulates sodium transport in rabbit proximal convoluted tubules.J Clin Invest.1984;73:507515.
  28. Goldsmith SR,Francis GS,Cowley AW.Arginine vasopressin and the renal response to water loading in congestive heart failure.Am J Cardiol.1986;58:295299.
  29. Schrier RW,Berl T,Anderson RJ.Osmotic and nonosmotic control of vasopressin release.Am J Physiol.1979;236:F321F332.
  30. Verbalis JG.Vasopressin V2 receptor antagonists.J Mol Endocrinol.2002;29:19.
  31. Nielsen S,Kwon TH,Christensen BM,Promeneur D,Frokiaer J,Marples D.Physiology and pathophysiology of renal aquaporins.J Am Soc Nephrol.1999;10:647663.
  32. Sonnenblick M,Friedlander Y,Rosin AJ.Diuretic‐induced severe hyponatremia. Review and analysis of 129 reported patients.Chest.1993;103:601606.
  33. Felker GM,Benza RL,Chandler AB, et al.Heart failure etiology and response to milrinone in decompensated heart failure: results from the OPTIME‐CHF study.J Am Coll Cardiol.2003;41:9971003.
  34. Packer M,Medina N,Yushak M.Correction of dilutional hyponatremia in severe chronic heart failure by converting‐enzyme inhibition.Ann Intern Med.1984;100:782789.
  35. Birnbaumer M.Vasopressin receptors.Trends Endocrinol Metab.2000;11:406410.
  36. Gheorghiade M,Niazi I,Ouyang J, et al.Vasopressin V2‐receptor blockade with tolvaptan in patients with chronic heart failure: results from a double‐blind, randomized trial.Circulation.2003;107:26902696.
  37. Gheorghiade M,Konstam MA,Burnett JC, et al.Short‐term clinical effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for heart failure: the EVEREST Clinical Status Trials.JAMA.2007;297:13321343.
  38. Konstam MA,Gheorghiade M,Burnett JC, et al.Effects of oral tolvaptan in patients hospitalized for worsening heart failure: the EVEREST Outcome Trial.JAMA.2007;297:13191331.
  39. Schrier RW,Gross P,Gheorghiade M, et al.Tolvaptan, a selective oral vasopressin V2‐receptor antagonist, for hyponatremia.N Engl J Med.2006;355:20992112.
  40. Abraham WT,Shamshirsaz AA,McFann K,Oren RM,Schrier RW.Aquaretic effect of lixivaptan, an oral, non‐peptide, selective V2 receptor vasopressin antagonist, in New York Heart Association functional class II and III chronic heart failure patients.J Am Coll Cardiol.2006;47:16151621.
  41. Udelson JE,Smith WB,Hendrix GH, et al.Acute hemodynamic effects of conivaptan, a dual V(1A) and V(2) vasopressin receptor antagonist, in patients with advanced heart failure.Circulation.2001;104:24172423.
  42. Goldsmith SR,Elkayam U,Haught WH,Barve A,He W.Efficacy and safety of the vasopressin V1A/V2‐receptor antagonist conivaptan in acute decompensated heart failure: a dose‐ranging pilot study.J Card Fail.2008;14:641647.
  43. Zeltser D,Rosansky S,van RH,Verbalis JG,Smith N.Assessment of the efficacy and safety of intravenous conivaptan in euvolemic and hypervolemic hyponatremia.Am J Nephrol.2007;27:447457.
Issue
Journal of Hospital Medicine - 5(3)
Issue
Journal of Hospital Medicine - 5(3)
Page Number
S33-S39
Page Number
S33-S39
Article Type
Display Headline
Managing hyponatremia in patients with heart failure
Display Headline
Managing hyponatremia in patients with heart failure
Legacy Keywords
arginine vasopressin receptor antagonists, common electrolyte disorders, heart failure, hyponatremia, pharmaceuticals
Legacy Keywords
arginine vasopressin receptor antagonists, common electrolyte disorders, heart failure, hyponatremia, pharmaceuticals
Sections
Article Source
Copyright © 2010 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Professor of Medicine, Director, Advanced Heart Failure Treatment Program, University of California, San Diego, 200 West Arbor Street, San Diego, California 92103‐8411
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

Smartphones for Clinical Communication

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
The use of smartphones for clinical communication on internal medicine wards

The scope and importance of communication between clinicians in the delivery of health care is increasingly being recognized.1, 2 Poor communication is known to be a source of inefficiency and errors within healthcare.36 The major issues with communication include the frequent use of interruptive communication mechanisms and the difficulty of knowing who to contact.2 Traditional paging remains the primary method to contact a physician despite being disruptive, inefficient, and predisposing to errors.710 The identification of the responsible physician for a patient can also be complicated with the numerous call schedules, different coverage rules, vacations, and protected academic time for residents. In 1 observational study, 25% of calls in a hospital were attempts to identify a responsible individual for a specific role.11 A recent study revealed that 14% of pages went to the wrong physician and that 47% of these errant pages warranted an urgent response.12 In our institution, the process for a nurse to identify which physician to contact regarding a patient care issue can be complex (Figure 1).

Figure 1
Process that nurses may need to go through to find out whom to contact regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

The use of email and mobile phone technology has been recommended as a method to improve communication.2 It can improve communication between clinicians by providing a method of triaging based on importance: instantly by telephone for urgent issues and less disruptively by email for nonurgent issues. There is limited literature on the use of email for improving communication between clinicians or on the use of smartphones. In a previous pilot study, we found that with minimal training, residents were able to use smartphones on general medical wards for clinical communication and that technical challenges were minimal.13 In an intensive care setting, the use of wireless emails using smartphones was perceived by staff to improve communication.14

Methods

Aim

To evaluate the use of smartphones to improve the communication processes on an academic general internal medicine service.

Setting

We conducted the study within the General Internal Medicine service at the Toronto General Hospital, an academic teaching hospital. The service is comprised of 4 teaching teams, each with 1 staff physician, 1 senior resident, 2 to 3 junior residents, and medical students. The environment is typically characterized by a high volume of medically complex patients and constant turnover of physicians with varying degrees of experience. Ethical review of the study was performed by the University Health Network Research Ethics Board.

Program Description

Recognizing the difficulty in implementing complex interventions within clinical care, we used a staged approach applying the standard Plan‐Do‐Study‐Act (PDSA) methodology for quality improvement.15 This involved developing a component of the system, releasing it to the users to learn from their experience, then taking this feedback to make changes and continuing with the next cycle. This allowed early identification and quicker resolution of issues.

Smartphones as Clinical Communication Tools

The first change implemented was to supplement the standard numeric pager with smartphones as the primary communication device for the residents. The BlackBerry device (Research in Motion, Ontario, Canada) was selected because it is an efficient, usable device with voice and secure email functionality that also happens to be the standard smartphone for our hospital administrators. Beginning in March 2008, all residents on the internal medicine service were equipped with a smartphone that they kept for their entire rotation. Contact lists of all devices were prepopulated with the hospital phone numbers that were important in coordinating patient care. These included numbers such as hospital flow coordinators, radiology reporting rooms, interventional radiology, and microbiology.

To facilitate reaching the most responsible resident for patients, we provided an additional smartphone, the Team BlackBerry, that was carried by the senior resident of each team during regular hours and then passed to the resident that was covering the team at night and on weekends. With the new process, a nurse would only require knowledge of which team the patient belonged to in order to contact the most responsible resident (Figure 2). The Team BlackBerry also received critical laboratory and medication alerts that contained clinical decision support. The laboratory and medication alerts were generated through the hospital rule‐based clinical decision support system (Misys Insight).

Figure 2
New process for nurses to contact a physician regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Web‐paging to Improve Nurse‐Physician Communication

In May 2008, we implemented a platform for text paging that we called web‐paging that allowed the nurses to efficiently send email messages securely through a hospital intranet page to the Team BlackBerry. A nurse would access the form from any computer on the wards, select the appropriate team, provide relevant information, and then send the message. Although the form would send an email, the term web‐paging was used to create familiarity with their previous process of contacting a resident, namely paging. This process reserved voice communication for urgent matters only while routing less urgent issues through the web‐paging form. Furthermore, emails were categorized into three priority levels:

  • Call back requested messages were for issues that were not life threatening but needed immediate action or required discussion with a physician.

  • E‐mail response requested messages were for issues that needed an action but could wait until the physician was available, such as cosigning an order.

  • Information only with no response necessary messages facilitated 1‐way communication, such as updating the team of recent vital signs for a patient.

Physicians would receive prompt notification of a new email on their smartphone, and they could reply to the email or call back using a link within the email. Nurses were able to view the physician's email responses from an email account that was shared amongst the nurses on each ward.

Program Evaluation

The outcome measures of the study were residents' use of smartphones and perceptions of residents and nurses on the new communication process. Resident use of the smartphones was measured by the volume and the frequency of phone calls and emails over a 3‐month period from September 2008 to November 2008. Residents' perceptions were measured by a survey administered prior to the start of their clinical rotation and at the end of their rotation (Table 1). Data collection for residents occurred between June 2008 and February 2009. Nurses' perceptions were measured by surveys administered to the nurses working on either day shifts or night shifts over a 1‐week period prior to the intervention in March 2008 and then 6 months after in September 2008 (Table 2).

Resident Perceptions on the Change in Communication Process
Pre‐Survey (n = 59) Median, (mode, mean) Post‐Survey (n = 65) Median, (mode, mean) P value
Training level
Postgraduate Year 1 39 (66.1%) 43 (66.2%)
Postgraduate Year 2 17 (28.8%) 19 (29.2%)
Postgraduate Year 3 3 (5.1%) 3 (4.6%)
Currently own and use a personal digital assistant 30 (55.6%) 38 (62.3%)
Currently own a BlackBerry 6 (10.5%) 5 (8.3%)
The following questions used a Likert scale with 1 representing strongly disagree and 5 representing strongly agree
Q1. I never have issues accessing a phone to discuss patient care issues 3 (2, 2.8) 4 (5, 3.9) <0.001
Q2. I often waste a lot of time waiting for my pages to be answered 4 (4, 4.1) 4 (4, 3.3) 0.004
Q3. Communicating with my team often takes a lot of time 4 (4, 3.5) 2 (1, 2.5) <0.001
Q4. I have quick and easy access to contact information (eg, allied health, departments, consultants, etc) necessary for providing patient care 3 (2, 2.6) 4 (4, 3.7) <0.001
Q5. My primary communication device (pager/ BlackBerry) is non‐disruptive to my workflow 3 (3, 2.7) 4 (4, 3.4) 0.002
Q6. My primary communication device (pager/ BlackBerry) helps me prioritize my tasks 3 (3, 2.9) 4 (4, 3.7) <0.001
Q7. Email and/or text messaging is something I find useful for communication about patient care 4 (3, 3.6) 5 (5, 4.4) <0.001
Q8. Overall, I am satisfied with my primary communication device (pager/BlackBerry) 3 (3, 2.9) 4 (5, 4.2) <0.001
Q9. Overall, being phoned on the BlackBerry (instead of receiving a page) is disruptive to me 3 (2, 3.0)
Q10. I consider Internet access on the BlackBerry useful for looking up information related to patient care. 3 (3, 3.0)
Q11. I prefer not having a BlackBerry at all than having to deal with technical difficulties when I use it 2 (1, 1.9)
Q12. I feel that overall a BlackBerry saves me time 4 (5, 4.3)
Nursing Perceptions on the Change in Communication Process
Pre‐Survey (n = 27) Median, Mode Post‐Survey (n = 35) Median, Mode P Value
Day shifts 18 (66.7%) 20 (57.1%)
Total number of times you tried to page, email or telephone a resident or medical student in the shift just completed. 3.70 2.51 0.16
Did you need to repeatedly try to contact (page, email, or call) a resident at any time during the shift regarding the same issue? 80% 34.3% 0.0012
Total amount of time that you spent in the last shift trying to contact doctors or other health care providers (minutes). 27.6 11.1 <0.001
The following questions used a Likert scale with 1 representing Strongly Disagree and 5 representing Strongly Agree:
Overall, it is straightforward to contact the resident taking care of my patients. 4 (4) 4 (4) 0.27
Overall, I am satisfied with physicians' response time when I need to contact them urgently. 3 (4) 4 (4) 0.54
I spend a lot of time away from the bedside just trying to contact physicians. 3 (4) 3 (3) 0.17
I like being able to call and reach the doctor directly. 4 (5)
Overall I am satisfied with the BlackBerry/email communication system. 4 (4)

Data Analysis

We present utilization as the mean number of messages sent and received daily. We report survey responses as the mean scores on a 5‐point Likert scale. For ordinal data, we described the survey responses using median and mode, and the Mann Whitney U test was used to test for differences between before and after responses. For parametric data, unpaired t‐tests were used to look for differences in the perceptions before and after the intervention. All P values are 2‐sided.

Results

Smartphone Usage

Usage of emails and calls for the team devices over the three months is shown in Table 3. Most communications were through the Team BlackBerry devices with the individual devices showing less usage than the team device with 2.1 sent emails per day and 4.0 received emails per day. On the days with highest use, there were 7 calls received per hour and 6 emails received per hour at peak times through the day to a Team BlackBerry. Web‐paging was the largest source of emails sent to Team BlackBerrys (42%). Critical laboratory or medication alerts represented 17.1% and the remainder (40.9%) were other communications such as communication from residents, staff physicians, pharmacists, and allied health professionals. Of the web‐paging communications, 35.1% requested a call back, 22.6% requested email response, and 42.2% were informational items only that did not require a response.

Average Daily Communications Using the Team BlackBerry
Phone Calls Emails
Incoming Outgoing Received Sent
Average 9.1 6.6 14.3 2.8
Minimum 0 0 0 0
Maximum 35 45 57 13
Median 8 4 13.5 2

Resident Perceptions

Of the 91 residents, 59 residents completed the presurveys (response rate 65%) and 65 completed the postsurveys (response rate 71%) (Table 1). There was a statistically significant improvement in all of the items measured. There were also 26 postsurvey comments with 22 describing positive aspects and 9 describing negative aspects. Specific positive attributes of the new communication system that were mentioned were: easier to communicate with other physicians within the team (n = 6), easier to communicate with other health care members of the team (nurses, allied health) (n = 3), and increased mobility (n = 2). The following are some examples:

  • Blackberrys are great for contacting team member, able to mobile in hospital instead of waiting by phone ‐ since we're so busy

  • Excellent for interpersonal communication. It definitely sped up patients care ie, consults, getting investigations done and discharges. Everyone should have one.

  • Text messaging, especially in group format so that all members participate in conversations is the primary communication tool of choice. Emailing is a necessity of the modern workforce.

The 2 predominant issues described were that the direct calling by nurses was disruptive (n = 6) and voice mail was not useful (n = 3):

  • I dislike the voicemail as it is like paging back but you also need to make a phone call to your phone list. Also we are always being interrupted which is disruptive to work but prefer BlackBerrys.

  • Helpful but often disruptive such as getting calls when seeing patients.

Nursing Perceptions

From the typical staff of 94 full‐time and part‐time nurses, 27 of 48 (56%) completed the preintervention survey, and 35 of 54 (65%) completed the postintervention survey. There was a perceived significant decrease in the need to repeatedly try to contact a physician for the same issue (80% vs. 34.3%, P = 0.001) and a significant decrease in the perceived amount of time spent trying to contact clinicians (27.6 minutes vs. 11.1 minutes, P < 0.001) (Table 2). On 5 point Likert scales, there was no perceived improvement in communication areas such as finding out who to reach and resident response time. Nurses appreciated the ability to call the physicians directly for urgent issues (median response 4, mode of 5).

Discussion

Principal Results

In this pilot study of using smartphones and email, residents perceived that it significantly improved their efficiency. Nurses perceived a reduction in the time spent trying to communicate with clinicians.

There were significant lessons learned. First, we found that residents readily utilized smartphones and felt that it improved their efficiency. Staff physicians and residents have outlined multiple concerns over increased workload, increasing patient complexity, and longer work hours. Therefore, interventions that improve physician productivity are essential in maintaining and improving patient care.16 From residents' comments, it appears their improved efficiency is derived not only from improved communication within the medical team, but also within the interprofessional team. Efficiency was also perceived to improve from the greater mobility of being able to walk while returning a page or while waiting for a page to be returned.

Second, we found that changing the communication process was complicated. While the number of calls and emails per day appeared manageable, the peak times had very high volumes and may actually be unmanageable. By facilitating the process of reaching the responsible physician, we may have lowered the threshold for contacting the physician and thus actually increased communication volumes. It is unclear whether the end result is beneficial as improved management of patients is balanced by the increased interruptions. Further study is required to understand the effect of increasing communication on patient outcomes.

Finally, residents indicated quite strongly that they did not want telephone calls to be the primary mode of contact as they found the frequent telephone calls too disruptive. A direct call to a physician created significant disruption to workflow requiring interrupting a task to answer the call or deferring the call to later review of a voice mail message. Accessing voice mail would typically take at least a minute, much more time than the few seconds it takes to review a numeric page or a text page. Residents quickly provided feedback that this process was unacceptable and reduced their ability to deliver care effectively. Furthermore, nurses were frustrated by the added time required to leave voice messages and the poor response rate. As information within voice mails was not acted upon, nurses quickly learned to just keep calling.

Costs of implementation were significantly more than the numeric paging system. With a cost of $5/month for a numeric pager, the cost of numeric pagers for 4 medical teams with 4 physicians each was $80/month (all costs in Canadian dollars). From our implementation, the approximate cost of an individual smartphone was $80/month while team‐based devices were $160/month due to much higher usage. With 4 individual smartphones and one team‐based smartphone per team, costs with the new system were approximately $1,600/month. While implementing smartphones creates a significantly greater expense, it may be worthwhile if improved communication leads to more efficient and higher quality care. Our current system is funded through the hospital to allow for further evaluation, and whether other hospitals provide smartphones for clinical communications likely would depend on whether they provided a cost effective alternative to numeric pagers. This would likely require some more tangible benefit than perceived improved efficiency.

Comparison With Prior Work

Similar to the study of wireless email using smartphones in the Intensive Care Unit, physicians in our study perceived an improvement in efficiency.14 Our study adds further information to the literature since we implemented on general medical wards in an academic teaching hospital. Our study is consistent with the previous studies have documented physicians' perceptions of improved performance with mobile phones.7, 17

Limitations

There are limitations to this study. In order to improve communication, we implemented a complex intervention with several components: (1) smartphones for residents, (2) Web‐paging for nurses, and (3) a new process of identifying the most responsible physician using Team BlackBerrys. From our surveys, it is difficult to know the relative impact of each individual component. Other interventions such as team‐based alphanumeric pagers may achieve similar improvements to nurses' perceptions of improved communication.

As this was a pilot study assessing new communication methods, we informally assessed perceptions using surveys which had not been validated. While the use of formally validated surveys would be more useful, we were unable to find any that specifically addressed the innovation in communication seen with smartphones. Validity issues with the instruments and the low response rates may have contributed to the incongruent result seen with nurses' surveys in that there was a perceived improvement in time to contact physicians but not in the ease of contact of physicians or the satisfaction of the communication method. This incongruent result may also reflect that in spite of the fact that the nurses were accustomed to the new system and that things may have improved, communication issues still remain and likely would benefit from further study.

It would have also been useful to determine the effect of this intervention on numeric pages sent to traditional pagers, which was not captured. It is also important to realize that we did not replace the pagers but instead supplemented residents with smartphones. To replace pagers may involve significant costs to upgrade reception within hospital walls to reduce areas of little or no reception. Finally, this study was completed at 1 academic health science center, and the generalizability of the findings may be limited.

Future Study

There is great opportunity to improve communication between clinicians, and the use of smartphones, email and improved identification of the most responsible physician are some methods to achieving this. Further research would be useful to determine the relative importance of each component. With smartphones, it is would be useful to know whether the benefits seen were primarily due to the mobile email or having another phone available or other features. Finally, the use of web‐paging and email to communicate increases the documentation of communications, but a further improvement could be providing acknowledgement to nurses that web‐pages were received and read. This would close the loop on communications, ensuring that there are no lost communications and that escalation occurs promptly for urgent communications.

In summary, we implemented and evaluated a system to route and prioritize clinical communications combining the use of phone calls and secure email messaging to smartphones. Residents strongly perceived an improvement in communication with smartphones. Further objective clinical evaluation is necessary to determine if this intervention improves efficiency and more importantly, the quality of care.

Acknowledgements

The investigators retained all control over study design, data collection, analysis and interpretation, and preparation of reports.

References
  1. O'Leary KJ,Liebovitz DM,Baker DW.How hospitalists spend their time: insights on efficiency and safety.J Hosp Med.2006;1(2):8893.
  2. Coiera E.When conversation is better than computation.J Am Med Inform Assoc.2000;7(3):277286.
  3. Brennan TA,Leape LL,Laird NM, et al.Incidence of adverse events and negligence in hospitalized patients. Results of the Harvard Medical Practice Study I.N Engl J Med.1991;324(6):370376.
  4. Woods DM,Holl JL,Angst DB, et al.Gaps in pediatric clinician communication and opportunities for improvement.J Healthc Qual.2008;30(5):4354.
  5. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Newby L,Hamilton JD.The quality in Australian Health Care Study.Med J Aust.1995;163(9):458471.
  6. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Hamilton JD.Quality in Australian Health Care Study.Med J Aust.1996;164(12):754.
  7. Soto RG,Chu LF,Goldman JM,Rampil IJ,Ruskin KJ.Communication in critical care environments: mobile telephones improve patient care.Anesth Analg.2006;102(2):535541.
  8. Blum NJ,Lieu TA.Interrupted care. The effects of paging on pediatric resident activities.Am J Dis Child.1992;146(7):806808.
  9. Harvey R,Jarrett PG,Peltekian KM.Patterns of paging medical interns during night calls at two teaching hospitals.CMAJ.1994;151(3):307311.
  10. Katz MH,Schroeder SA.The sounds of the hospital. Paging patterns in three teaching hospitals.N Engl J Med.1988;319(24):15851589.
  11. Coiera E,Tombs V.Communication behaviours in a hospital setting: an observational study.BMJ1998;316(7132):673676.
  12. Wong BM,Quan S,Cheung CM, et al.Why isn't anyone returning my page? Frequency and clinical importance of pages sent to the wrong physician.Arch Intern Med.2009;169(11):10721073.
  13. Quan S,Wu RC,Morra D, et al.Demonstrating the BlackBerry as a clinical communication tool: a pilot study conducted through the centre for innovation in complex care.Healthc Q2008;11(4):9498.
  14. O'Connor C,Friedrich JO,Scales DC,Adhikari NK.The use of wireless email to improve healthcare team communication.J Am Med Inform Assoc.2009;16(5):705713.
  15. Berwick DM.A primer on leading the improvement of systems.BMJ.1996;312(7031):619622.
  16. Okie S.An elusive balance—residents' work hours and the continuity of care.N Engl J Med.2007;356(26):26652667.
  17. Ramesh J,Carter AO,Campbell MH, et al.Use of mobile phones by medical staff at Queen Elizabeth Hospital, Barbados: evidence for both benefit and harm.J Hosp Infect.2008;70(2):160165.
Article PDF
Issue
Journal of Hospital Medicine - 5(9)
Page Number
553-559
Legacy Keywords
cellular phones, email, handheld computers, hospital communication systems, telecommunications
Sections
Article PDF
Article PDF

The scope and importance of communication between clinicians in the delivery of health care is increasingly being recognized.1, 2 Poor communication is known to be a source of inefficiency and errors within healthcare.36 The major issues with communication include the frequent use of interruptive communication mechanisms and the difficulty of knowing who to contact.2 Traditional paging remains the primary method to contact a physician despite being disruptive, inefficient, and predisposing to errors.710 The identification of the responsible physician for a patient can also be complicated with the numerous call schedules, different coverage rules, vacations, and protected academic time for residents. In 1 observational study, 25% of calls in a hospital were attempts to identify a responsible individual for a specific role.11 A recent study revealed that 14% of pages went to the wrong physician and that 47% of these errant pages warranted an urgent response.12 In our institution, the process for a nurse to identify which physician to contact regarding a patient care issue can be complex (Figure 1).

Figure 1
Process that nurses may need to go through to find out whom to contact regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

The use of email and mobile phone technology has been recommended as a method to improve communication.2 It can improve communication between clinicians by providing a method of triaging based on importance: instantly by telephone for urgent issues and less disruptively by email for nonurgent issues. There is limited literature on the use of email for improving communication between clinicians or on the use of smartphones. In a previous pilot study, we found that with minimal training, residents were able to use smartphones on general medical wards for clinical communication and that technical challenges were minimal.13 In an intensive care setting, the use of wireless emails using smartphones was perceived by staff to improve communication.14

Methods

Aim

To evaluate the use of smartphones to improve the communication processes on an academic general internal medicine service.

Setting

We conducted the study within the General Internal Medicine service at the Toronto General Hospital, an academic teaching hospital. The service is comprised of 4 teaching teams, each with 1 staff physician, 1 senior resident, 2 to 3 junior residents, and medical students. The environment is typically characterized by a high volume of medically complex patients and constant turnover of physicians with varying degrees of experience. Ethical review of the study was performed by the University Health Network Research Ethics Board.

Program Description

Recognizing the difficulty in implementing complex interventions within clinical care, we used a staged approach applying the standard Plan‐Do‐Study‐Act (PDSA) methodology for quality improvement.15 This involved developing a component of the system, releasing it to the users to learn from their experience, then taking this feedback to make changes and continuing with the next cycle. This allowed early identification and quicker resolution of issues.

Smartphones as Clinical Communication Tools

The first change implemented was to supplement the standard numeric pager with smartphones as the primary communication device for the residents. The BlackBerry device (Research in Motion, Ontario, Canada) was selected because it is an efficient, usable device with voice and secure email functionality that also happens to be the standard smartphone for our hospital administrators. Beginning in March 2008, all residents on the internal medicine service were equipped with a smartphone that they kept for their entire rotation. Contact lists of all devices were prepopulated with the hospital phone numbers that were important in coordinating patient care. These included numbers such as hospital flow coordinators, radiology reporting rooms, interventional radiology, and microbiology.

To facilitate reaching the most responsible resident for patients, we provided an additional smartphone, the Team BlackBerry, that was carried by the senior resident of each team during regular hours and then passed to the resident that was covering the team at night and on weekends. With the new process, a nurse would only require knowledge of which team the patient belonged to in order to contact the most responsible resident (Figure 2). The Team BlackBerry also received critical laboratory and medication alerts that contained clinical decision support. The laboratory and medication alerts were generated through the hospital rule‐based clinical decision support system (Misys Insight).

Figure 2
New process for nurses to contact a physician regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Web‐paging to Improve Nurse‐Physician Communication

In May 2008, we implemented a platform for text paging that we called web‐paging that allowed the nurses to efficiently send email messages securely through a hospital intranet page to the Team BlackBerry. A nurse would access the form from any computer on the wards, select the appropriate team, provide relevant information, and then send the message. Although the form would send an email, the term web‐paging was used to create familiarity with their previous process of contacting a resident, namely paging. This process reserved voice communication for urgent matters only while routing less urgent issues through the web‐paging form. Furthermore, emails were categorized into three priority levels:

  • Call back requested messages were for issues that were not life threatening but needed immediate action or required discussion with a physician.

  • E‐mail response requested messages were for issues that needed an action but could wait until the physician was available, such as cosigning an order.

  • Information only with no response necessary messages facilitated 1‐way communication, such as updating the team of recent vital signs for a patient.

Physicians would receive prompt notification of a new email on their smartphone, and they could reply to the email or call back using a link within the email. Nurses were able to view the physician's email responses from an email account that was shared amongst the nurses on each ward.

Program Evaluation

The outcome measures of the study were residents' use of smartphones and perceptions of residents and nurses on the new communication process. Resident use of the smartphones was measured by the volume and the frequency of phone calls and emails over a 3‐month period from September 2008 to November 2008. Residents' perceptions were measured by a survey administered prior to the start of their clinical rotation and at the end of their rotation (Table 1). Data collection for residents occurred between June 2008 and February 2009. Nurses' perceptions were measured by surveys administered to the nurses working on either day shifts or night shifts over a 1‐week period prior to the intervention in March 2008 and then 6 months after in September 2008 (Table 2).

Resident Perceptions on the Change in Communication Process
Pre‐Survey (n = 59) Median, (mode, mean) Post‐Survey (n = 65) Median, (mode, mean) P value
Training level
Postgraduate Year 1 39 (66.1%) 43 (66.2%)
Postgraduate Year 2 17 (28.8%) 19 (29.2%)
Postgraduate Year 3 3 (5.1%) 3 (4.6%)
Currently own and use a personal digital assistant 30 (55.6%) 38 (62.3%)
Currently own a BlackBerry 6 (10.5%) 5 (8.3%)
The following questions used a Likert scale with 1 representing strongly disagree and 5 representing strongly agree
Q1. I never have issues accessing a phone to discuss patient care issues 3 (2, 2.8) 4 (5, 3.9) <0.001
Q2. I often waste a lot of time waiting for my pages to be answered 4 (4, 4.1) 4 (4, 3.3) 0.004
Q3. Communicating with my team often takes a lot of time 4 (4, 3.5) 2 (1, 2.5) <0.001
Q4. I have quick and easy access to contact information (eg, allied health, departments, consultants, etc) necessary for providing patient care 3 (2, 2.6) 4 (4, 3.7) <0.001
Q5. My primary communication device (pager/ BlackBerry) is non‐disruptive to my workflow 3 (3, 2.7) 4 (4, 3.4) 0.002
Q6. My primary communication device (pager/ BlackBerry) helps me prioritize my tasks 3 (3, 2.9) 4 (4, 3.7) <0.001
Q7. Email and/or text messaging is something I find useful for communication about patient care 4 (3, 3.6) 5 (5, 4.4) <0.001
Q8. Overall, I am satisfied with my primary communication device (pager/BlackBerry) 3 (3, 2.9) 4 (5, 4.2) <0.001
Q9. Overall, being phoned on the BlackBerry (instead of receiving a page) is disruptive to me 3 (2, 3.0)
Q10. I consider Internet access on the BlackBerry useful for looking up information related to patient care. 3 (3, 3.0)
Q11. I prefer not having a BlackBerry at all than having to deal with technical difficulties when I use it 2 (1, 1.9)
Q12. I feel that overall a BlackBerry saves me time 4 (5, 4.3)
Nursing Perceptions on the Change in Communication Process
Pre‐Survey (n = 27) Median, Mode Post‐Survey (n = 35) Median, Mode P Value
Day shifts 18 (66.7%) 20 (57.1%)
Total number of times you tried to page, email or telephone a resident or medical student in the shift just completed. 3.70 2.51 0.16
Did you need to repeatedly try to contact (page, email, or call) a resident at any time during the shift regarding the same issue? 80% 34.3% 0.0012
Total amount of time that you spent in the last shift trying to contact doctors or other health care providers (minutes). 27.6 11.1 <0.001
The following questions used a Likert scale with 1 representing Strongly Disagree and 5 representing Strongly Agree:
Overall, it is straightforward to contact the resident taking care of my patients. 4 (4) 4 (4) 0.27
Overall, I am satisfied with physicians' response time when I need to contact them urgently. 3 (4) 4 (4) 0.54
I spend a lot of time away from the bedside just trying to contact physicians. 3 (4) 3 (3) 0.17
I like being able to call and reach the doctor directly. 4 (5)
Overall I am satisfied with the BlackBerry/email communication system. 4 (4)

Data Analysis

We present utilization as the mean number of messages sent and received daily. We report survey responses as the mean scores on a 5‐point Likert scale. For ordinal data, we described the survey responses using median and mode, and the Mann Whitney U test was used to test for differences between before and after responses. For parametric data, unpaired t‐tests were used to look for differences in the perceptions before and after the intervention. All P values are 2‐sided.

Results

Smartphone Usage

Usage of emails and calls for the team devices over the three months is shown in Table 3. Most communications were through the Team BlackBerry devices with the individual devices showing less usage than the team device with 2.1 sent emails per day and 4.0 received emails per day. On the days with highest use, there were 7 calls received per hour and 6 emails received per hour at peak times through the day to a Team BlackBerry. Web‐paging was the largest source of emails sent to Team BlackBerrys (42%). Critical laboratory or medication alerts represented 17.1% and the remainder (40.9%) were other communications such as communication from residents, staff physicians, pharmacists, and allied health professionals. Of the web‐paging communications, 35.1% requested a call back, 22.6% requested email response, and 42.2% were informational items only that did not require a response.

Average Daily Communications Using the Team BlackBerry
Phone Calls Emails
Incoming Outgoing Received Sent
Average 9.1 6.6 14.3 2.8
Minimum 0 0 0 0
Maximum 35 45 57 13
Median 8 4 13.5 2

Resident Perceptions

Of the 91 residents, 59 residents completed the presurveys (response rate 65%) and 65 completed the postsurveys (response rate 71%) (Table 1). There was a statistically significant improvement in all of the items measured. There were also 26 postsurvey comments with 22 describing positive aspects and 9 describing negative aspects. Specific positive attributes of the new communication system that were mentioned were: easier to communicate with other physicians within the team (n = 6), easier to communicate with other health care members of the team (nurses, allied health) (n = 3), and increased mobility (n = 2). The following are some examples:

  • Blackberrys are great for contacting team member, able to mobile in hospital instead of waiting by phone ‐ since we're so busy

  • Excellent for interpersonal communication. It definitely sped up patients care ie, consults, getting investigations done and discharges. Everyone should have one.

  • Text messaging, especially in group format so that all members participate in conversations is the primary communication tool of choice. Emailing is a necessity of the modern workforce.

The 2 predominant issues described were that the direct calling by nurses was disruptive (n = 6) and voice mail was not useful (n = 3):

  • I dislike the voicemail as it is like paging back but you also need to make a phone call to your phone list. Also we are always being interrupted which is disruptive to work but prefer BlackBerrys.

  • Helpful but often disruptive such as getting calls when seeing patients.

Nursing Perceptions

From the typical staff of 94 full‐time and part‐time nurses, 27 of 48 (56%) completed the preintervention survey, and 35 of 54 (65%) completed the postintervention survey. There was a perceived significant decrease in the need to repeatedly try to contact a physician for the same issue (80% vs. 34.3%, P = 0.001) and a significant decrease in the perceived amount of time spent trying to contact clinicians (27.6 minutes vs. 11.1 minutes, P < 0.001) (Table 2). On 5 point Likert scales, there was no perceived improvement in communication areas such as finding out who to reach and resident response time. Nurses appreciated the ability to call the physicians directly for urgent issues (median response 4, mode of 5).

Discussion

Principal Results

In this pilot study of using smartphones and email, residents perceived that it significantly improved their efficiency. Nurses perceived a reduction in the time spent trying to communicate with clinicians.

There were significant lessons learned. First, we found that residents readily utilized smartphones and felt that it improved their efficiency. Staff physicians and residents have outlined multiple concerns over increased workload, increasing patient complexity, and longer work hours. Therefore, interventions that improve physician productivity are essential in maintaining and improving patient care.16 From residents' comments, it appears their improved efficiency is derived not only from improved communication within the medical team, but also within the interprofessional team. Efficiency was also perceived to improve from the greater mobility of being able to walk while returning a page or while waiting for a page to be returned.

Second, we found that changing the communication process was complicated. While the number of calls and emails per day appeared manageable, the peak times had very high volumes and may actually be unmanageable. By facilitating the process of reaching the responsible physician, we may have lowered the threshold for contacting the physician and thus actually increased communication volumes. It is unclear whether the end result is beneficial as improved management of patients is balanced by the increased interruptions. Further study is required to understand the effect of increasing communication on patient outcomes.

Finally, residents indicated quite strongly that they did not want telephone calls to be the primary mode of contact as they found the frequent telephone calls too disruptive. A direct call to a physician created significant disruption to workflow requiring interrupting a task to answer the call or deferring the call to later review of a voice mail message. Accessing voice mail would typically take at least a minute, much more time than the few seconds it takes to review a numeric page or a text page. Residents quickly provided feedback that this process was unacceptable and reduced their ability to deliver care effectively. Furthermore, nurses were frustrated by the added time required to leave voice messages and the poor response rate. As information within voice mails was not acted upon, nurses quickly learned to just keep calling.

Costs of implementation were significantly more than the numeric paging system. With a cost of $5/month for a numeric pager, the cost of numeric pagers for 4 medical teams with 4 physicians each was $80/month (all costs in Canadian dollars). From our implementation, the approximate cost of an individual smartphone was $80/month while team‐based devices were $160/month due to much higher usage. With 4 individual smartphones and one team‐based smartphone per team, costs with the new system were approximately $1,600/month. While implementing smartphones creates a significantly greater expense, it may be worthwhile if improved communication leads to more efficient and higher quality care. Our current system is funded through the hospital to allow for further evaluation, and whether other hospitals provide smartphones for clinical communications likely would depend on whether they provided a cost effective alternative to numeric pagers. This would likely require some more tangible benefit than perceived improved efficiency.

Comparison With Prior Work

Similar to the study of wireless email using smartphones in the Intensive Care Unit, physicians in our study perceived an improvement in efficiency.14 Our study adds further information to the literature since we implemented on general medical wards in an academic teaching hospital. Our study is consistent with the previous studies have documented physicians' perceptions of improved performance with mobile phones.7, 17

Limitations

There are limitations to this study. In order to improve communication, we implemented a complex intervention with several components: (1) smartphones for residents, (2) Web‐paging for nurses, and (3) a new process of identifying the most responsible physician using Team BlackBerrys. From our surveys, it is difficult to know the relative impact of each individual component. Other interventions such as team‐based alphanumeric pagers may achieve similar improvements to nurses' perceptions of improved communication.

As this was a pilot study assessing new communication methods, we informally assessed perceptions using surveys which had not been validated. While the use of formally validated surveys would be more useful, we were unable to find any that specifically addressed the innovation in communication seen with smartphones. Validity issues with the instruments and the low response rates may have contributed to the incongruent result seen with nurses' surveys in that there was a perceived improvement in time to contact physicians but not in the ease of contact of physicians or the satisfaction of the communication method. This incongruent result may also reflect that in spite of the fact that the nurses were accustomed to the new system and that things may have improved, communication issues still remain and likely would benefit from further study.

It would have also been useful to determine the effect of this intervention on numeric pages sent to traditional pagers, which was not captured. It is also important to realize that we did not replace the pagers but instead supplemented residents with smartphones. To replace pagers may involve significant costs to upgrade reception within hospital walls to reduce areas of little or no reception. Finally, this study was completed at 1 academic health science center, and the generalizability of the findings may be limited.

Future Study

There is great opportunity to improve communication between clinicians, and the use of smartphones, email and improved identification of the most responsible physician are some methods to achieving this. Further research would be useful to determine the relative importance of each component. With smartphones, it is would be useful to know whether the benefits seen were primarily due to the mobile email or having another phone available or other features. Finally, the use of web‐paging and email to communicate increases the documentation of communications, but a further improvement could be providing acknowledgement to nurses that web‐pages were received and read. This would close the loop on communications, ensuring that there are no lost communications and that escalation occurs promptly for urgent communications.

In summary, we implemented and evaluated a system to route and prioritize clinical communications combining the use of phone calls and secure email messaging to smartphones. Residents strongly perceived an improvement in communication with smartphones. Further objective clinical evaluation is necessary to determine if this intervention improves efficiency and more importantly, the quality of care.

Acknowledgements

The investigators retained all control over study design, data collection, analysis and interpretation, and preparation of reports.

The scope and importance of communication between clinicians in the delivery of health care is increasingly being recognized.1, 2 Poor communication is known to be a source of inefficiency and errors within healthcare.36 The major issues with communication include the frequent use of interruptive communication mechanisms and the difficulty of knowing who to contact.2 Traditional paging remains the primary method to contact a physician despite being disruptive, inefficient, and predisposing to errors.710 The identification of the responsible physician for a patient can also be complicated with the numerous call schedules, different coverage rules, vacations, and protected academic time for residents. In 1 observational study, 25% of calls in a hospital were attempts to identify a responsible individual for a specific role.11 A recent study revealed that 14% of pages went to the wrong physician and that 47% of these errant pages warranted an urgent response.12 In our institution, the process for a nurse to identify which physician to contact regarding a patient care issue can be complex (Figure 1).

Figure 1
Process that nurses may need to go through to find out whom to contact regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

The use of email and mobile phone technology has been recommended as a method to improve communication.2 It can improve communication between clinicians by providing a method of triaging based on importance: instantly by telephone for urgent issues and less disruptively by email for nonurgent issues. There is limited literature on the use of email for improving communication between clinicians or on the use of smartphones. In a previous pilot study, we found that with minimal training, residents were able to use smartphones on general medical wards for clinical communication and that technical challenges were minimal.13 In an intensive care setting, the use of wireless emails using smartphones was perceived by staff to improve communication.14

Methods

Aim

To evaluate the use of smartphones to improve the communication processes on an academic general internal medicine service.

Setting

We conducted the study within the General Internal Medicine service at the Toronto General Hospital, an academic teaching hospital. The service is comprised of 4 teaching teams, each with 1 staff physician, 1 senior resident, 2 to 3 junior residents, and medical students. The environment is typically characterized by a high volume of medically complex patients and constant turnover of physicians with varying degrees of experience. Ethical review of the study was performed by the University Health Network Research Ethics Board.

Program Description

Recognizing the difficulty in implementing complex interventions within clinical care, we used a staged approach applying the standard Plan‐Do‐Study‐Act (PDSA) methodology for quality improvement.15 This involved developing a component of the system, releasing it to the users to learn from their experience, then taking this feedback to make changes and continuing with the next cycle. This allowed early identification and quicker resolution of issues.

Smartphones as Clinical Communication Tools

The first change implemented was to supplement the standard numeric pager with smartphones as the primary communication device for the residents. The BlackBerry device (Research in Motion, Ontario, Canada) was selected because it is an efficient, usable device with voice and secure email functionality that also happens to be the standard smartphone for our hospital administrators. Beginning in March 2008, all residents on the internal medicine service were equipped with a smartphone that they kept for their entire rotation. Contact lists of all devices were prepopulated with the hospital phone numbers that were important in coordinating patient care. These included numbers such as hospital flow coordinators, radiology reporting rooms, interventional radiology, and microbiology.

To facilitate reaching the most responsible resident for patients, we provided an additional smartphone, the Team BlackBerry, that was carried by the senior resident of each team during regular hours and then passed to the resident that was covering the team at night and on weekends. With the new process, a nurse would only require knowledge of which team the patient belonged to in order to contact the most responsible resident (Figure 2). The Team BlackBerry also received critical laboratory and medication alerts that contained clinical decision support. The laboratory and medication alerts were generated through the hospital rule‐based clinical decision support system (Misys Insight).

Figure 2
New process for nurses to contact a physician regarding a care issue for their patient. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

Web‐paging to Improve Nurse‐Physician Communication

In May 2008, we implemented a platform for text paging that we called web‐paging that allowed the nurses to efficiently send email messages securely through a hospital intranet page to the Team BlackBerry. A nurse would access the form from any computer on the wards, select the appropriate team, provide relevant information, and then send the message. Although the form would send an email, the term web‐paging was used to create familiarity with their previous process of contacting a resident, namely paging. This process reserved voice communication for urgent matters only while routing less urgent issues through the web‐paging form. Furthermore, emails were categorized into three priority levels:

  • Call back requested messages were for issues that were not life threatening but needed immediate action or required discussion with a physician.

  • E‐mail response requested messages were for issues that needed an action but could wait until the physician was available, such as cosigning an order.

  • Information only with no response necessary messages facilitated 1‐way communication, such as updating the team of recent vital signs for a patient.

Physicians would receive prompt notification of a new email on their smartphone, and they could reply to the email or call back using a link within the email. Nurses were able to view the physician's email responses from an email account that was shared amongst the nurses on each ward.

Program Evaluation

The outcome measures of the study were residents' use of smartphones and perceptions of residents and nurses on the new communication process. Resident use of the smartphones was measured by the volume and the frequency of phone calls and emails over a 3‐month period from September 2008 to November 2008. Residents' perceptions were measured by a survey administered prior to the start of their clinical rotation and at the end of their rotation (Table 1). Data collection for residents occurred between June 2008 and February 2009. Nurses' perceptions were measured by surveys administered to the nurses working on either day shifts or night shifts over a 1‐week period prior to the intervention in March 2008 and then 6 months after in September 2008 (Table 2).

Resident Perceptions on the Change in Communication Process
Pre‐Survey (n = 59) Median, (mode, mean) Post‐Survey (n = 65) Median, (mode, mean) P value
Training level
Postgraduate Year 1 39 (66.1%) 43 (66.2%)
Postgraduate Year 2 17 (28.8%) 19 (29.2%)
Postgraduate Year 3 3 (5.1%) 3 (4.6%)
Currently own and use a personal digital assistant 30 (55.6%) 38 (62.3%)
Currently own a BlackBerry 6 (10.5%) 5 (8.3%)
The following questions used a Likert scale with 1 representing strongly disagree and 5 representing strongly agree
Q1. I never have issues accessing a phone to discuss patient care issues 3 (2, 2.8) 4 (5, 3.9) <0.001
Q2. I often waste a lot of time waiting for my pages to be answered 4 (4, 4.1) 4 (4, 3.3) 0.004
Q3. Communicating with my team often takes a lot of time 4 (4, 3.5) 2 (1, 2.5) <0.001
Q4. I have quick and easy access to contact information (eg, allied health, departments, consultants, etc) necessary for providing patient care 3 (2, 2.6) 4 (4, 3.7) <0.001
Q5. My primary communication device (pager/ BlackBerry) is non‐disruptive to my workflow 3 (3, 2.7) 4 (4, 3.4) 0.002
Q6. My primary communication device (pager/ BlackBerry) helps me prioritize my tasks 3 (3, 2.9) 4 (4, 3.7) <0.001
Q7. Email and/or text messaging is something I find useful for communication about patient care 4 (3, 3.6) 5 (5, 4.4) <0.001
Q8. Overall, I am satisfied with my primary communication device (pager/BlackBerry) 3 (3, 2.9) 4 (5, 4.2) <0.001
Q9. Overall, being phoned on the BlackBerry (instead of receiving a page) is disruptive to me 3 (2, 3.0)
Q10. I consider Internet access on the BlackBerry useful for looking up information related to patient care. 3 (3, 3.0)
Q11. I prefer not having a BlackBerry at all than having to deal with technical difficulties when I use it 2 (1, 1.9)
Q12. I feel that overall a BlackBerry saves me time 4 (5, 4.3)
Nursing Perceptions on the Change in Communication Process
Pre‐Survey (n = 27) Median, Mode Post‐Survey (n = 35) Median, Mode P Value
Day shifts 18 (66.7%) 20 (57.1%)
Total number of times you tried to page, email or telephone a resident or medical student in the shift just completed. 3.70 2.51 0.16
Did you need to repeatedly try to contact (page, email, or call) a resident at any time during the shift regarding the same issue? 80% 34.3% 0.0012
Total amount of time that you spent in the last shift trying to contact doctors or other health care providers (minutes). 27.6 11.1 <0.001
The following questions used a Likert scale with 1 representing Strongly Disagree and 5 representing Strongly Agree:
Overall, it is straightforward to contact the resident taking care of my patients. 4 (4) 4 (4) 0.27
Overall, I am satisfied with physicians' response time when I need to contact them urgently. 3 (4) 4 (4) 0.54
I spend a lot of time away from the bedside just trying to contact physicians. 3 (4) 3 (3) 0.17
I like being able to call and reach the doctor directly. 4 (5)
Overall I am satisfied with the BlackBerry/email communication system. 4 (4)

Data Analysis

We present utilization as the mean number of messages sent and received daily. We report survey responses as the mean scores on a 5‐point Likert scale. For ordinal data, we described the survey responses using median and mode, and the Mann Whitney U test was used to test for differences between before and after responses. For parametric data, unpaired t‐tests were used to look for differences in the perceptions before and after the intervention. All P values are 2‐sided.

Results

Smartphone Usage

Usage of emails and calls for the team devices over the three months is shown in Table 3. Most communications were through the Team BlackBerry devices with the individual devices showing less usage than the team device with 2.1 sent emails per day and 4.0 received emails per day. On the days with highest use, there were 7 calls received per hour and 6 emails received per hour at peak times through the day to a Team BlackBerry. Web‐paging was the largest source of emails sent to Team BlackBerrys (42%). Critical laboratory or medication alerts represented 17.1% and the remainder (40.9%) were other communications such as communication from residents, staff physicians, pharmacists, and allied health professionals. Of the web‐paging communications, 35.1% requested a call back, 22.6% requested email response, and 42.2% were informational items only that did not require a response.

Average Daily Communications Using the Team BlackBerry
Phone Calls Emails
Incoming Outgoing Received Sent
Average 9.1 6.6 14.3 2.8
Minimum 0 0 0 0
Maximum 35 45 57 13
Median 8 4 13.5 2

Resident Perceptions

Of the 91 residents, 59 residents completed the presurveys (response rate 65%) and 65 completed the postsurveys (response rate 71%) (Table 1). There was a statistically significant improvement in all of the items measured. There were also 26 postsurvey comments with 22 describing positive aspects and 9 describing negative aspects. Specific positive attributes of the new communication system that were mentioned were: easier to communicate with other physicians within the team (n = 6), easier to communicate with other health care members of the team (nurses, allied health) (n = 3), and increased mobility (n = 2). The following are some examples:

  • Blackberrys are great for contacting team member, able to mobile in hospital instead of waiting by phone ‐ since we're so busy

  • Excellent for interpersonal communication. It definitely sped up patients care ie, consults, getting investigations done and discharges. Everyone should have one.

  • Text messaging, especially in group format so that all members participate in conversations is the primary communication tool of choice. Emailing is a necessity of the modern workforce.

The 2 predominant issues described were that the direct calling by nurses was disruptive (n = 6) and voice mail was not useful (n = 3):

  • I dislike the voicemail as it is like paging back but you also need to make a phone call to your phone list. Also we are always being interrupted which is disruptive to work but prefer BlackBerrys.

  • Helpful but often disruptive such as getting calls when seeing patients.

Nursing Perceptions

From the typical staff of 94 full‐time and part‐time nurses, 27 of 48 (56%) completed the preintervention survey, and 35 of 54 (65%) completed the postintervention survey. There was a perceived significant decrease in the need to repeatedly try to contact a physician for the same issue (80% vs. 34.3%, P = 0.001) and a significant decrease in the perceived amount of time spent trying to contact clinicians (27.6 minutes vs. 11.1 minutes, P < 0.001) (Table 2). On 5 point Likert scales, there was no perceived improvement in communication areas such as finding out who to reach and resident response time. Nurses appreciated the ability to call the physicians directly for urgent issues (median response 4, mode of 5).

Discussion

Principal Results

In this pilot study of using smartphones and email, residents perceived that it significantly improved their efficiency. Nurses perceived a reduction in the time spent trying to communicate with clinicians.

There were significant lessons learned. First, we found that residents readily utilized smartphones and felt that it improved their efficiency. Staff physicians and residents have outlined multiple concerns over increased workload, increasing patient complexity, and longer work hours. Therefore, interventions that improve physician productivity are essential in maintaining and improving patient care.16 From residents' comments, it appears their improved efficiency is derived not only from improved communication within the medical team, but also within the interprofessional team. Efficiency was also perceived to improve from the greater mobility of being able to walk while returning a page or while waiting for a page to be returned.

Second, we found that changing the communication process was complicated. While the number of calls and emails per day appeared manageable, the peak times had very high volumes and may actually be unmanageable. By facilitating the process of reaching the responsible physician, we may have lowered the threshold for contacting the physician and thus actually increased communication volumes. It is unclear whether the end result is beneficial as improved management of patients is balanced by the increased interruptions. Further study is required to understand the effect of increasing communication on patient outcomes.

Finally, residents indicated quite strongly that they did not want telephone calls to be the primary mode of contact as they found the frequent telephone calls too disruptive. A direct call to a physician created significant disruption to workflow requiring interrupting a task to answer the call or deferring the call to later review of a voice mail message. Accessing voice mail would typically take at least a minute, much more time than the few seconds it takes to review a numeric page or a text page. Residents quickly provided feedback that this process was unacceptable and reduced their ability to deliver care effectively. Furthermore, nurses were frustrated by the added time required to leave voice messages and the poor response rate. As information within voice mails was not acted upon, nurses quickly learned to just keep calling.

Costs of implementation were significantly more than the numeric paging system. With a cost of $5/month for a numeric pager, the cost of numeric pagers for 4 medical teams with 4 physicians each was $80/month (all costs in Canadian dollars). From our implementation, the approximate cost of an individual smartphone was $80/month while team‐based devices were $160/month due to much higher usage. With 4 individual smartphones and one team‐based smartphone per team, costs with the new system were approximately $1,600/month. While implementing smartphones creates a significantly greater expense, it may be worthwhile if improved communication leads to more efficient and higher quality care. Our current system is funded through the hospital to allow for further evaluation, and whether other hospitals provide smartphones for clinical communications likely would depend on whether they provided a cost effective alternative to numeric pagers. This would likely require some more tangible benefit than perceived improved efficiency.

Comparison With Prior Work

Similar to the study of wireless email using smartphones in the Intensive Care Unit, physicians in our study perceived an improvement in efficiency.14 Our study adds further information to the literature since we implemented on general medical wards in an academic teaching hospital. Our study is consistent with the previous studies have documented physicians' perceptions of improved performance with mobile phones.7, 17

Limitations

There are limitations to this study. In order to improve communication, we implemented a complex intervention with several components: (1) smartphones for residents, (2) Web‐paging for nurses, and (3) a new process of identifying the most responsible physician using Team BlackBerrys. From our surveys, it is difficult to know the relative impact of each individual component. Other interventions such as team‐based alphanumeric pagers may achieve similar improvements to nurses' perceptions of improved communication.

As this was a pilot study assessing new communication methods, we informally assessed perceptions using surveys which had not been validated. While the use of formally validated surveys would be more useful, we were unable to find any that specifically addressed the innovation in communication seen with smartphones. Validity issues with the instruments and the low response rates may have contributed to the incongruent result seen with nurses' surveys in that there was a perceived improvement in time to contact physicians but not in the ease of contact of physicians or the satisfaction of the communication method. This incongruent result may also reflect that in spite of the fact that the nurses were accustomed to the new system and that things may have improved, communication issues still remain and likely would benefit from further study.

It would have also been useful to determine the effect of this intervention on numeric pages sent to traditional pagers, which was not captured. It is also important to realize that we did not replace the pagers but instead supplemented residents with smartphones. To replace pagers may involve significant costs to upgrade reception within hospital walls to reduce areas of little or no reception. Finally, this study was completed at 1 academic health science center, and the generalizability of the findings may be limited.

Future Study

There is great opportunity to improve communication between clinicians, and the use of smartphones, email and improved identification of the most responsible physician are some methods to achieving this. Further research would be useful to determine the relative importance of each component. With smartphones, it is would be useful to know whether the benefits seen were primarily due to the mobile email or having another phone available or other features. Finally, the use of web‐paging and email to communicate increases the documentation of communications, but a further improvement could be providing acknowledgement to nurses that web‐pages were received and read. This would close the loop on communications, ensuring that there are no lost communications and that escalation occurs promptly for urgent communications.

In summary, we implemented and evaluated a system to route and prioritize clinical communications combining the use of phone calls and secure email messaging to smartphones. Residents strongly perceived an improvement in communication with smartphones. Further objective clinical evaluation is necessary to determine if this intervention improves efficiency and more importantly, the quality of care.

Acknowledgements

The investigators retained all control over study design, data collection, analysis and interpretation, and preparation of reports.

References
  1. O'Leary KJ,Liebovitz DM,Baker DW.How hospitalists spend their time: insights on efficiency and safety.J Hosp Med.2006;1(2):8893.
  2. Coiera E.When conversation is better than computation.J Am Med Inform Assoc.2000;7(3):277286.
  3. Brennan TA,Leape LL,Laird NM, et al.Incidence of adverse events and negligence in hospitalized patients. Results of the Harvard Medical Practice Study I.N Engl J Med.1991;324(6):370376.
  4. Woods DM,Holl JL,Angst DB, et al.Gaps in pediatric clinician communication and opportunities for improvement.J Healthc Qual.2008;30(5):4354.
  5. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Newby L,Hamilton JD.The quality in Australian Health Care Study.Med J Aust.1995;163(9):458471.
  6. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Hamilton JD.Quality in Australian Health Care Study.Med J Aust.1996;164(12):754.
  7. Soto RG,Chu LF,Goldman JM,Rampil IJ,Ruskin KJ.Communication in critical care environments: mobile telephones improve patient care.Anesth Analg.2006;102(2):535541.
  8. Blum NJ,Lieu TA.Interrupted care. The effects of paging on pediatric resident activities.Am J Dis Child.1992;146(7):806808.
  9. Harvey R,Jarrett PG,Peltekian KM.Patterns of paging medical interns during night calls at two teaching hospitals.CMAJ.1994;151(3):307311.
  10. Katz MH,Schroeder SA.The sounds of the hospital. Paging patterns in three teaching hospitals.N Engl J Med.1988;319(24):15851589.
  11. Coiera E,Tombs V.Communication behaviours in a hospital setting: an observational study.BMJ1998;316(7132):673676.
  12. Wong BM,Quan S,Cheung CM, et al.Why isn't anyone returning my page? Frequency and clinical importance of pages sent to the wrong physician.Arch Intern Med.2009;169(11):10721073.
  13. Quan S,Wu RC,Morra D, et al.Demonstrating the BlackBerry as a clinical communication tool: a pilot study conducted through the centre for innovation in complex care.Healthc Q2008;11(4):9498.
  14. O'Connor C,Friedrich JO,Scales DC,Adhikari NK.The use of wireless email to improve healthcare team communication.J Am Med Inform Assoc.2009;16(5):705713.
  15. Berwick DM.A primer on leading the improvement of systems.BMJ.1996;312(7031):619622.
  16. Okie S.An elusive balance—residents' work hours and the continuity of care.N Engl J Med.2007;356(26):26652667.
  17. Ramesh J,Carter AO,Campbell MH, et al.Use of mobile phones by medical staff at Queen Elizabeth Hospital, Barbados: evidence for both benefit and harm.J Hosp Infect.2008;70(2):160165.
References
  1. O'Leary KJ,Liebovitz DM,Baker DW.How hospitalists spend their time: insights on efficiency and safety.J Hosp Med.2006;1(2):8893.
  2. Coiera E.When conversation is better than computation.J Am Med Inform Assoc.2000;7(3):277286.
  3. Brennan TA,Leape LL,Laird NM, et al.Incidence of adverse events and negligence in hospitalized patients. Results of the Harvard Medical Practice Study I.N Engl J Med.1991;324(6):370376.
  4. Woods DM,Holl JL,Angst DB, et al.Gaps in pediatric clinician communication and opportunities for improvement.J Healthc Qual.2008;30(5):4354.
  5. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Newby L,Hamilton JD.The quality in Australian Health Care Study.Med J Aust.1995;163(9):458471.
  6. Wilson RM,Runciman WB,Gibberd RW,Harrison BT,Hamilton JD.Quality in Australian Health Care Study.Med J Aust.1996;164(12):754.
  7. Soto RG,Chu LF,Goldman JM,Rampil IJ,Ruskin KJ.Communication in critical care environments: mobile telephones improve patient care.Anesth Analg.2006;102(2):535541.
  8. Blum NJ,Lieu TA.Interrupted care. The effects of paging on pediatric resident activities.Am J Dis Child.1992;146(7):806808.
  9. Harvey R,Jarrett PG,Peltekian KM.Patterns of paging medical interns during night calls at two teaching hospitals.CMAJ.1994;151(3):307311.
  10. Katz MH,Schroeder SA.The sounds of the hospital. Paging patterns in three teaching hospitals.N Engl J Med.1988;319(24):15851589.
  11. Coiera E,Tombs V.Communication behaviours in a hospital setting: an observational study.BMJ1998;316(7132):673676.
  12. Wong BM,Quan S,Cheung CM, et al.Why isn't anyone returning my page? Frequency and clinical importance of pages sent to the wrong physician.Arch Intern Med.2009;169(11):10721073.
  13. Quan S,Wu RC,Morra D, et al.Demonstrating the BlackBerry as a clinical communication tool: a pilot study conducted through the centre for innovation in complex care.Healthc Q2008;11(4):9498.
  14. O'Connor C,Friedrich JO,Scales DC,Adhikari NK.The use of wireless email to improve healthcare team communication.J Am Med Inform Assoc.2009;16(5):705713.
  15. Berwick DM.A primer on leading the improvement of systems.BMJ.1996;312(7031):619622.
  16. Okie S.An elusive balance—residents' work hours and the continuity of care.N Engl J Med.2007;356(26):26652667.
  17. Ramesh J,Carter AO,Campbell MH, et al.Use of mobile phones by medical staff at Queen Elizabeth Hospital, Barbados: evidence for both benefit and harm.J Hosp Infect.2008;70(2):160165.
Issue
Journal of Hospital Medicine - 5(9)
Issue
Journal of Hospital Medicine - 5(9)
Page Number
553-559
Page Number
553-559
Article Type
Display Headline
The use of smartphones for clinical communication on internal medicine wards
Display Headline
The use of smartphones for clinical communication on internal medicine wards
Legacy Keywords
cellular phones, email, handheld computers, hospital communication systems, telecommunications
Legacy Keywords
cellular phones, email, handheld computers, hospital communication systems, telecommunications
Sections
Article Source
Copyright © 2010 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Toronto General Hospital, 14EN‐222, 200Elizabeth St, Toronto, Ontario, Canada, M5G 2C4
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

Glycemic Control in the Hospital

Article Type
Changed
Sun, 05/28/2017 - 20:10
Display Headline
Effects of an educational program and a standardized insulin order form on glycemic outcomes in non‐critically ill hospitalized patients

Hyperglycemia is common in hospitalized patients, and hyperglycemia has been associated with poor hospital outcomes. The adverse physiologic effects of acute hyperglycemia are well established1 and several clinical studies have linked hyperglycemia with poor clinical outcomes in certain patient populations.28 Although the optimal target range for inpatient glycemic control has not yet been defined, these studies support the goal of metabolic control for hospital patients. However, there are many barriers to achieving adequate glycemic control in the hospital, and blood glucoses in the hospital are often far from recommended targets.9, 10 One barrier appears to be the low priority given to glycemic control in the hospital. Hyperglycemia in the hospital is often ignored,11 and insulin regimens are often chosen for simplicity as opposed to effectiveness.12 Other barriers to glycemic control in the hospital include the physiologic effects (stress) of acute illness, and the frequent nutritional changes and interruptions that occur.

Most hyperglycemic patients on a general medicine unit are treated with subcutaneous insulin, but the optimal strategy for prescribing insulin in the hospital remains uncertain. A technical review of the literature on the management of diabetes in the hospital setting from 2004 recommends prescribing insulin in a way that mimics physiologic insulin secretion (ie, physiologic or basal‐bolus insulin).1 This approach has been promulgated by experts, but there has been very little research to support these recommendations. One small, randomized trial concluded that a basal‐bolus approach achieved better glycemic control than the use of sliding‐scale insulin alone,13 and 2 quality improvement studies using a before/after design have demonstrated improvements in glycemic control after the implementation of interventions designed to encourage physiologic insulin use.14, 15

In this study we hypothesized that a few simple interventions (education for physicians and nurses, and a standardized insulin order form) would lead to a higher rate of basal‐bolus insulin use and simultaneously improve glycemic control and patient safety.

Methods

Study Design

This study was performed at the University of Michigan Hospital over a 6‐month period, and the protocol was approved by the Institutional Review Board. We performed a quasi‐experimental study comparing 3 patient groups. The intervention group (IG) was subject to all of the interventions discussed below (physician education, nurse education, and the standardized order form). The concurrent control group (CCG) was hospitalized during the same time period as the IG, but was only subject to 1 of the interventions (physician education). These patients were cared for by the same physicians as the IG, but on a different unit where the nurses had not received the education and where the standardized insulin order form was not available. Patients were admitted to the IG unit or the CCG unit via the institution's usual admission process. In addition, we examined an historic control group (HCG) which was hospitalized during the same months of the year, but 2 years prior. The HCG was not subject to any of the interventions.

Interventions

Standardized Subcutaneous Insulin Order Form

This form (Supporting Information Appendix 1) was designed to encourage physicians to prescribe insulin in a physiologic way, providing basal, nutritional, and correctional insulin. The form is based on best practice guidelines,1 and is in agreement with the principles of the inpatient management of diabetes and hyperglycemia endorsed by several professional organizations.16, 17 The form was engineered by a multidisciplinary team, including an endocrinologist, several hospitalists, several nurses, a certified diabetes educator, a pharmacist, and others. It is derived from the extensive experience of the University of Michigan Hospital Intensive Insulin Program (HIIP) in the Division of Endocrinology, and on work done by the Society of Hospital Medicine (SHM) Glycemic Control Task Force.1719 This form was only used in the care of patients in the IG. The form, which was not approved for use on other floors, did not creep to other units. The standardized order form was the only way to order insulin or to modify the insulin regimen on the IG unit. The frequency of review or revision of the insulin orders was left to the discretion of the inpatient physicians.

Physician/Midlevel Provider Education

Physicians and midlevel providers caring for patients in the IG and the CCG were given specific education about the best practice recommendations for the management of diabetes and hyperglycemia in hospitalized patients. This education was based on the principles of anticipatory, physiologic insulin use. On nonhouse staff services, the education was provided to the attending physicians and midlevel providers, and on house staff services, the education was provided to the residents. All physician education was provided by the physician authors (D.W. and R.G.). A summary of the content of the physician education is provided in Supporting Information Appendix 2.

Nurse Education

Nurses caring for patients in the IG were given education similar to that which was provided to the physicians (see above), with an emphasis on practical issues related to delivering physiologic insulin. It included topics such as blood glucose monitoring, and the real‐time manipulation of nutritional insulin doses in accordance with the clinical situation (decision‐making that was specifically delegated to the nursing staff by the order set).

Patients

Patients were eligible for inclusion in the analysis if they met the following inclusion criteria: they were admitted to the inpatient General Internal Medicine Services; subcutaneous insulin was provided to the patient during the hospitalization; they had at least 2 blood glucose values >180 mg/dL; they were discharged from the hospital on a pharmacologic glucose lowering agent (insulin or oral); and their total length‐of‐stay was 3 days to 14 days. Patients were excluded from the analysis if they were admitted with a primary diagnosis of diabetic ketoacidosis, diabetic hyperosmolar state, or hypoglycemia. Up to 10 consecutive days of glucose data were recorded for each patient, and the first day on which blood glucose information was available from the admitting floor was excluded from the analysis. Also, specific patient‐days were not analyzed if there were no bedside glucoses recorded, or if the patient was treated with an IV insulin infusion on that day.

Outcomes

The primary outcome was glycemic control. The primary unit of measure was the patient‐day (ie, all of the information for 1 patient on a single qualifying day). This was done to correct for the phenomenon of frequent repeat testing in response to abnormal values. It also allows for a more clinically relevant description of the actual glycemic control on a given day. Specifically, each patient‐day was categorized as in‐range (70‐180 mg/dL), hyperglycemic (>180 mg/dL), severely hyperglycemic (>250 mg/dL), hypoglycemic (<70 mg/dL), and/or severely hypoglycemic (<50 mg/dL). The primary endpoint was glycemic control in‐range. For a patient‐day to be in‐range, all readings for that particular day were within 70 mg/dL to180 mg/dL. For the readings that were not in the desired range, a minimum of 1 deviant reading in a particular day constituted classification into that category, and a single out‐of‐range patient‐day could be included in 1 or more of the out‐of‐range categories (eg, a patient‐day could be categorized as both severely hyperglycemic and hypoglycemic if it contained glucose readings in both of those ranges).

The day‐weighted mean blood glucose value was also calculated for each of the groups. This calculation utilized the mean blood glucose for each patient‐day, and then averaged these values for each group. These metrics have been endorsed as appropriate measures of glycemic control by the SHM Glycemic Control Task Force.20

Other Data

Several other clinical features were also examined, including the following: primary diagnoses listed in the hospital discharge summary for each patient (3 maximum); possible confounders including patient weight, length‐of‐stay, days receiving tube feeds, days receiving parenteral nutrition, and days during which patients were treated with high‐dose glucocorticoids (>10 mg/day of prednisone, or its equivalent) or oral diabetes medications; and the composition of the insulin regimen on each hospital day. Definitions of insulin regimens are provided in Table 1.

Definitions of the Insulin Regimens Prescribed for Each Patient‐Day
  • Correctional insulin (also known as sliding scale or as needed insulin) was allowed as part of any insulin regimen above. Correctional insulin was only recorded when it was unaccompanied by a scheduled insulin.

Any basal insulin dayAny day in which intermediate‐acting or long‐acting, scheduled insulin was given.
Basal insulin alone dayA day in which intermediate‐acting or long‐acting insulin was the only scheduled insulin given.
Any nutritional insulin dayAny day in which rapid‐acting or short‐acting, scheduled insulin was given.
Nutritional insulin alone dayA day in which rapid‐acting or short‐acting insulin was the only scheduled insulin given.
Basal plus nutritional dayA day in which both scheduled, intermediate‐acting or long‐acting insulin and scheduled, rapid‐acting or short‐acting insulin were given.
Pre‐mixed insulin dayAny day in which a pre‐mixed combination insulin was given.
Basal plus nutritional or pre‐mixed insulin dayA composite of the basal plus nutritional day category and the mixed insulin day category described above. This group includes any day in which either a pre‐mixed combination insulin was given OR a day in which both: (a) scheduled, intermediate‐acting or long‐acting and (b) scheduled, rapid‐acting or short‐acting insulin were given.
Sliding scale insulin alone day*Any day when only correctional (as needed) insulin was given.

Statistical Analysis

Bivariate analyses (chi‐square, and t‐tests) were carried out to compare demographic characteristics of the intervention and control populations. Since there were multiple glucose readings nested within individuals, multilevel mixed‐effects logistic regression was used to evaluate the association between the intervention and outcomes. A 2‐level hierarchical model was developed in which patient‐days were nested within patients; this accounted for the correlation between glycemic control across days for a given patient. Patient‐day was modeled as a random intercept and the log likelihood was estimated using adaptive Gaussian quadrature with 7 integration points. Alpha was set at 0.05, 2‐tailed. The final model was adjusted for gender, age, weight, length‐of‐stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was too infrequent to be considered in the adjusted analysis. All analyses were conducted in Stata/IC 10.0 (College Station, TX).

Results

A total of 245 patients provided 1315 patient‐days. Patient demographics are shown in Table 2. The patients' weight, length‐of‐stay, and primary diagnoses were similar across the 3 groups. There was a higher percentage of males in the IG as compared to the HCG.

Demographic Characteristics by Group
 IGCCGP Value IG vs. CCGHCGP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

  • Misc/other includes: malignancies, fluid/electrolyte, hematologic, transplant‐related, cardiac, venous thromboemolism, pulmonary, and dermatologic.

Number of patients8486 75 
Age, years, mean (SD)59.3 (15.3)60.4 (15.9)0.6859.2 (17.2)0.96
Range (n = 245)18‐9420‐87 24‐92 
Weight in kg, mean (SD)92.2 (29.5)89.5 (27.2)0.5794.2 (35.4)0.69
Range (n = 237)40‐19840‐188 42‐235 
Sex, n (%) (n = 245)  0.17 0.04
Male45 (53.6)37 (43.0) 28 (37.3) 
Female39(46.4)49 (57.0) 47 (62.7) 
Length of stay, mean (SD)7.6 (3.3)7.4 (3.0)0.627.0 (2.5)0.14
Range (n = 245)4‐154‐15 4‐14 
Number of diagnoses169158 160 
Primary diagnoses, n (%)  0.56 0.10
Infections40 (23.7)45 (28.5) 49 (30.6) 
Gastrointestinal33 (19.5)19 (12.0) 14 (8.8) 
Rheumatologic13 (7.7)12 (7.6) 18 (11.2) 
Renal14 (8.3)10 (6.3) 16 (10.0) 
Diabetes‐related11 (6.5)11 (7.0) 10 (6.2) 
Neurologic8 (4.7)11 (7.0) 11 (6.9) 
*Misc/other50 (29.6)50 (31.6) 42 (26.3) 

Table 3 shows the insulin regimens used in the different groups. The use of basal insulin was similar between groups. Congruent with the goals of the education session and the order set, patients in the IG were more likely to be treated with a combination of basal and nutritional insulin than patients in the other groups. Patients in the HCG were more likely to be treated with a premixed insulin than patients in the other groups. However, even when premixed insulin was categorized as a form of basal plus nutritional insulin and combined into a composite group with the combined basal and nutritional days, this type of regimen remained more common in the IG than in the HCG. The rate of sliding scale insulin use alone (ie, without any scheduled insulin) was similar in the 3 groups.

Insulin Regimen, Oral Diabetes Agent Use and Nutritional Information by Group
Patient‐days on the followingIG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions of the insulin regimens are shown in Table 1.

  • Abbreviations: IG, intervention group; CCG, concurrent control group; HCG, historic control group.

Sliding scale alone, n (%)105 (23.2)130 (27.6)0.1289 (22.8)0.89
Basal alone, n (%)132 (29.1)231 (49.0)<0.01199 (50.9)<0.01
Nutritional alone, n (%)22 (4.9)5 (1.1)<0.018 (2.0)0.03
Basal plus nutritional, n (%)166 (36.6)71 (15.1)<0.0114 (3.6)<0.01
Pre‐mixed insulin included, n (%)27 (6.0)32 (6.8)0.6078 (20.0)<0.01
No insulin, n (%)1 (<1)2 (<1)0.593 (<1)0.28
Any basal, n (%)325 (71.7)334 (70.9)0.78291 (74.4)0.38
Any nutritional, n (%)215 (47.5)108 (22.9)<0.01100 (25.6)<0.01
Basal plus nutritional or pre‐mixed, n (%)193 (42.6)103 (21.9)<0.0192 (23.5)<0.01
Oral diabetes agents, n (%)79 (17.4)83 (17.6)0.9474 (18.9)0.58
Sulfonylureas, n (%)40 (8.8)63 (13.4)0.0337 (9.5)0.75
Parenteral nutrition/tube feeds, n (%)0 (0)18 (3.8) 8 (2.0) 
High dose corticosteroids, n (%)66 (14.6)93 (19.8)0.0451 (13.0)0.52

Other relevant measures are also shown in Table 3. The use of oral diabetes agents was similar in the 3 groups. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was infrequent.

A comparison of glycemic control in the three groups is shown in Table 4. In contrast to the CCG, patients in the IG experienced more days within the target glucose range (17% vs. 10.6%, P < 0.01), fewer days with severe hyperglycemia (48.3% vs. 59.2%, P < 0.01), and had a lower day‐weighted average blood glucose (195.9 vs. 212.6, P < 0.01). Compared to the HCG, patients in the IG experienced similar rates of hyperglycemia, but fewer hypoglycemic days (5.1% vs. 9.2%, P = 0.02).

Glycemic Control by Group
 IG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions for glycemic control are provided in the text.

  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

Patient‐days     
In range, n (%)77 (17.0)50 (10.6)<0.0166 (16.9)0.98
Out of range, n (%)376 (83.0)421 (89.4)<0.01325 (83.1)0.98
Hyperglycemic, n (%)289 (63.8)310 (65.8)0.52248 (63.4)0.91
Severely hyperglycemic, n (%)219 (48.3)279 (59.2)<0.01176 (45.0)0.32
Hypoglycemic, n (%)23 (5.1)36 (7.6)0.1136 (9.2)0.02
Severely hypoglycemic, n (%)13 (2.9)10 (2.1)0.4715 (3.8)0.44
Day weighted average blood glucose (SD)195.9 (66.8)212.6 (73.4)<0.01190.5 (63.1)0.25

The percentages of patients with severe hyperglycemia in each group are shown in Figure 1 by hospital day. Severe hyperglycemia was common, but there was a trend towards a decrease in the prevalence of severe hyperglycemia with increasing hospital days for all study groups, although it was consistently higher in the CCG than in the IG. Figure 2 shows the types of insulin regimens used by hospital day (composite for all groups). The use of basal plus nutritional insulin (the recommended regimen) increased gradually with increasing hospital days. When taken together, the information in both figures support the hypothesis that the use of the recommended insulin regimen may have contributed to the modest improvements in glycemic control seen in the IG.

Figure 1
Severe Hyperglycemia by hospital day.
Figure 2
Regimen by hospital day.

In the final adjusted regression model, the intervention had a positive impact on glycemic control (Table 5). Subjects in the IG had a 72% increase in the odds of being in the target glucose range when compared to subjects in the CCG (P = 0.01). In addition, subjects in the IG had a 35% reduction in the odds of being severely hyperglycemic when compared to those in the CCG (P < 0.01). Finally, the odds ratio (OR) for being hypoglycemic among intervention subjects was 0.59 (P = 0.06) when compared to subjects in the CCG and 0.48 (P = 0.01) when compared to subjects in the HCG.

Multivariable Analysis of Glycemic Control
 Adjusted OR* IG vs. CCG95% CIP value IG vs. CCGAdjusted OR* IG vs. HCG95% CIP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; CI, confidence interval; HCG, historic control group; IG, intervention group; OR, odds ratio.

  • Adjusted for gender, age, weight, length of stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids.

  • This odds ratio reflects the unadjusted analysis as this model failed to converge after adjusting for the covariates.

In range1.721.16,2.550.011.080.74,1.580.68
Hyperglycemic0.930.70,1.220.580.950.71,1.280.74
Severely Hyperglycemic0.650.49,0.85<0.011.100.82,1.470.52
Hypoglycemic0.590.34,1.020.060.480.27,0.850.01
Severely Hypoglycemic1.360.59,3.140.470.970.29,1.440.28

Discussion

In this study, we investigated the effects of a standardized insulin order set, coupled with physician and nurse education, on glycemic control in hyperglycemic hospitalized patients. These interventions were designed to encourage a standardized approach to the treatment of hyperglycemia in hospitalized patients, based on the principles of physiologic insulin use, as described above. Our data suggest that the interventions did, indeed, alter the way insulin was prescribed, as more patients in the IG received a combination of basal plus nutritional insulin (the recommended regimen) than in the other groups. These interventions were associated with improved glycemic outcomes in the IG as compared to the CCG. The IG experienced a higher percentage of days in the target range and a trend toward fewer hypoglycemic days than the CCG. Although the IG experienced a similar percentage of days in the target range, it had significantly fewer hypoglycemic days than the HCG.

It is useful to consider the results of our study in the context of 2 other similar studies performed by Schnipper et al.14 and Maynard et al.15 Although each of these 3 studies have different study designs, they are similar in intent (to test the effects of simple quality improvement interventions on glycemic control in the hospital) and results (all showed significant improvements in some aspect of glycemic control). In our study, and the study by Maynard et al.,15 the interventions also led to decreases in the rates of hypoglycemia, whereas Schnipper et al.14 observed no difference in hypoglycemia. Of interest, in each of the three studies the interventions were associated with an increase in the use of some type of scheduled insulin. In our study and the study from Schnipper et al.14 the baseline use of basal insulin was quite high, and the interventions were associated with a significant increase in the addition of nutritional insulin. In the Maynard et al.15 study, the baseline use of sliding scale insulin alone was prevalent, and the interventions resulted in an increase in the use of basal insulin. The results of these studies, taken together, prompt us to conclude that the interventions employed in these studies are likely to lead to more frequent prescription of scheduled (anticipatory) insulin, and a modest improvement in glycemic control, without an increase (and perhaps with a decrease) in hypoglycemia.

A few of our study results are unexpected, or difficult to explain. In contrast to the other studies discussed above, our interventions did not affect the frequency of the use of sliding‐scale insulin alone (without any scheduled insulin), which was similar in the 3 groups. Although the reason for this is uncertain, we hypothesize that the high baseline use of basal insulin in our institution, and the lack of a hard stop preventing the use of sliding scale insulin alone explain this finding. Also, it is difficult to explain why measures of hyperglycemia were similar between the IG and the HCG despite the fact that the HCG was less often treated with a combination of basal and nutritional insulin and more often treated with mixed insulin.

There are several different mechanisms by which the interventions might have resulted in improved glycemic control in the IG compared to CCG. Our data clearly shows that insulin was prescribed differently in the IG, and the more frequent use of a combination of scheduled basal and nutritional insulin might have contributed to the differences between the groups. However, the effects of our interventions clearly went beyond physician education into the realm of true process improvement and standardization. The standardized order form was designed to prompt physicians to use a basal‐bolus insulin regimen. The order form also created nursing expectations of how insulin should be ordered, and clarified the roles of the different insulins that were prescribed.

On the medication administration record, each insulin was labeled as basal insulin (to be given even when fasting) or nutritional insulin (to be given along with the meal). The nurses caring for the IG also attended an education program that reinforced the role of the nurse in the bedside management of insulin administration. Specifically, nurses were taught to assess the premeal blood glucose and the patient's nutritional situation before giving the nutritional insulin (ie, Does the patient have food available? Will he tolerate eating the food?). In situations where is was not clear if the patient would be able to tolerate the ordered nutrition, the order set empowered the nurse to give the nutritional insulin after the meal, and to reduce the dose to match the patient's actual intake. These interventions resulted in some fundamental improvements in the nursing process of delivering insulin to the patient, and these changes might have resulted in improvements via mechanisms that are difficult to directly measure. Since the same physicians cared for both the IG and the CCG, interventions other than physician education clearly contributed to the observed improvements in the IG.

This study was not a randomized study, and there could be important undetected differences between the groups. However, all of the patients were admitted to the General Medicine Inpatient Services and the comparison of the general patient demographics and primary diagnoses between the groups do not suggest major differences.

Although the improvements in glycemic control seen in this study were statistically significant, they were quantitatively modest. The rates of hyperglycemia seen in this study, on the other hand, are quite remarkable. Both the American Diabetes Association and the American College of Endocrinology have recommended that blood glucoses in hospitalized patients not exceed a maximum value of 180 mg/dL, but the day‐weighted average blood glucose in this study was above that for each group. Even in the IG, over 80% of all patient‐days included at least 1 blood glucose value outside of the target range. These data suggest that better strategies for achieving metabolic control in hospitalized patients are needed.

It is worth mentioning that our interventions were not aggressively enforced. While the use of the order set was mandatory for the IG, it was flexible enough to allow for substantial practice variation, especially with respect to the dose of insulin prescribed. Although the education sessions discussed the specifics of insulin dosing in hospitalized patients, the order form did not offer dosing guidelines. It is possible that our interventions may have had a larger impact if a starting dose of insulin had been specified on the form. Although the insulin order form prompted physicians to act, there were no forced functions. Also, not all house staff attended the education sessions for physicians, and there was no feedback provided to physicians related to how they might improve their adherence to the recommendations presented in the educational module. Therefore, it is likely that more aggressive interventions could have led to greater changes in physician practice.

In conclusion, this study demonstrates that interventions including physician and nurse education and a standardized insulin order set can lead to improvement in glycemic control and patient safety in hospitalized patients treated with subcutaneous insulin. However, the observed improvements are modest, and poor metabolic control remains common, despite these interventions. These data suggest that standardization of the process of ordering and delivering subcutaneous insulin in the hospital may lead to a reduction in both hyperglycemia and hypoglycemia. However, it is clear that the interventions used in this study were not potent enough to achieve the recommended glycemic targets for the majority of patients. Additional research is needed to determine the best strategy for achieving safe and effective metabolic control in hospitalized, hyperglycemic, noncritically ill patients.

Acknowledgements

The authors thank David Conway for his work in data collection and management.

References
  1. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  2. Van den Berghe G,Wouters P,Weekers F, et al.Intensive insulin therapy in the critically ill patients.N Engl J Med.2001;345:13591367.
  3. Van den Berghe G,Wilmer A,Hermans G, et al.Intensive insulin therapy in the medical ICU.N Engl J Med.2006;354:449461.
  4. Furnary AP,Zerr KJ,Grunkemeier GL,Starr A.Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures.Ann Thorac Surg.1999;67:352360; discussion360–352.
  5. Furnary AP,Gao G,Grunkemeier GL, et al.Continuous insulin infusion reduces mortality in patients with diabetes undergoing coronary artery bypass grafting.J Thorac Cardiovasc Surg.2003;125:10071021.
  6. Umpierrez GE,Isaacs SD,Bazargan N,You X,Thaler LM,Kitabchi AE.Hyperglycemia: an independent marker of in‐hospital mortality in patients with undiagnosed diabetes.J Clin Endocrinol Metab.2002;87:978982.
  7. Pomposelli JJ,Baxter JK,Babineau TJ, et al.Early postoperative glucose control predicts nosocomial infection rate in diabetic patients.JPEN J Parenter Enteral Nutr.1998;22:7781.
  8. McAlister FA,Majumdar SR,Blitz S,Rowe BH,Romney J,Marrie TJ.The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community‐acquired pneumonia.Diabetes Care.2005;28:810815.
  9. Umpierrez G,Maynard G.Glycemic chaos (not glycemic control) still the rule for inpatient care: how do we stop the insanity?J Hosp Med.2006;1:141144.
  10. Boord JB,Greevy RA,Braithwaite SS, et al.Evaluation of hospital glycemic control at US Academic Medical Centers.J Hosp Med.2009;4:3544.
  11. Knecht LA,Gauthier SM,Castro JC, et al.Diabetes care in the hospital: is there clinical inertia?J Hosp Med.2006;1:151160.
  12. Schnipper JL,Barsky EE,Shaykevich S,Fitzmaurice G,Pendergrass ML.Inpatient management of diabetes and hyperglycemia among general medicine patients at a large teaching hospital.J Hosp Med.2006;1:145150.
  13. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  14. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  15. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  16. Garber AJ,Moghissi ES,Bransome ED, et al.American College of Endocrinology position statement on inpatient diabetes and metabolic control.Endocr Pract.2004;10:7782.
  17. Society of Hospital Medicine. Glycemic Control Resource Room. Available at: http://www.hospitalmedicine.org/AM/Template.cfm?Section= Quality_Improvement_Resource_Rooms3(5 Suppl):1728.
  18. Maynard G,Wesorick DH,O'Malley C,Inzucchi SE.Subcutaneous insulin order sets and protocols: effective design and implementation strategies.J Hosp Med.2008;3(5 Suppl):2941.
  19. Schnipper JL,Magee M,Larsen K,Inzucchi SE,Maynard G.Society of Hospital Medicine Glycemic Control Task Force summary: practical recommendations for assessing the impact of glycemic control efforts.J Hosp Med.2008;3(5 Suppl):6675.
Article PDF
Issue
Journal of Hospital Medicine - 5(8)
Page Number
438-445
Legacy Keywords
diabetes mellitus, hospitalization, hyperglycemia, inpatient, insulin
Sections
Article PDF
Article PDF

Hyperglycemia is common in hospitalized patients, and hyperglycemia has been associated with poor hospital outcomes. The adverse physiologic effects of acute hyperglycemia are well established1 and several clinical studies have linked hyperglycemia with poor clinical outcomes in certain patient populations.28 Although the optimal target range for inpatient glycemic control has not yet been defined, these studies support the goal of metabolic control for hospital patients. However, there are many barriers to achieving adequate glycemic control in the hospital, and blood glucoses in the hospital are often far from recommended targets.9, 10 One barrier appears to be the low priority given to glycemic control in the hospital. Hyperglycemia in the hospital is often ignored,11 and insulin regimens are often chosen for simplicity as opposed to effectiveness.12 Other barriers to glycemic control in the hospital include the physiologic effects (stress) of acute illness, and the frequent nutritional changes and interruptions that occur.

Most hyperglycemic patients on a general medicine unit are treated with subcutaneous insulin, but the optimal strategy for prescribing insulin in the hospital remains uncertain. A technical review of the literature on the management of diabetes in the hospital setting from 2004 recommends prescribing insulin in a way that mimics physiologic insulin secretion (ie, physiologic or basal‐bolus insulin).1 This approach has been promulgated by experts, but there has been very little research to support these recommendations. One small, randomized trial concluded that a basal‐bolus approach achieved better glycemic control than the use of sliding‐scale insulin alone,13 and 2 quality improvement studies using a before/after design have demonstrated improvements in glycemic control after the implementation of interventions designed to encourage physiologic insulin use.14, 15

In this study we hypothesized that a few simple interventions (education for physicians and nurses, and a standardized insulin order form) would lead to a higher rate of basal‐bolus insulin use and simultaneously improve glycemic control and patient safety.

Methods

Study Design

This study was performed at the University of Michigan Hospital over a 6‐month period, and the protocol was approved by the Institutional Review Board. We performed a quasi‐experimental study comparing 3 patient groups. The intervention group (IG) was subject to all of the interventions discussed below (physician education, nurse education, and the standardized order form). The concurrent control group (CCG) was hospitalized during the same time period as the IG, but was only subject to 1 of the interventions (physician education). These patients were cared for by the same physicians as the IG, but on a different unit where the nurses had not received the education and where the standardized insulin order form was not available. Patients were admitted to the IG unit or the CCG unit via the institution's usual admission process. In addition, we examined an historic control group (HCG) which was hospitalized during the same months of the year, but 2 years prior. The HCG was not subject to any of the interventions.

Interventions

Standardized Subcutaneous Insulin Order Form

This form (Supporting Information Appendix 1) was designed to encourage physicians to prescribe insulin in a physiologic way, providing basal, nutritional, and correctional insulin. The form is based on best practice guidelines,1 and is in agreement with the principles of the inpatient management of diabetes and hyperglycemia endorsed by several professional organizations.16, 17 The form was engineered by a multidisciplinary team, including an endocrinologist, several hospitalists, several nurses, a certified diabetes educator, a pharmacist, and others. It is derived from the extensive experience of the University of Michigan Hospital Intensive Insulin Program (HIIP) in the Division of Endocrinology, and on work done by the Society of Hospital Medicine (SHM) Glycemic Control Task Force.1719 This form was only used in the care of patients in the IG. The form, which was not approved for use on other floors, did not creep to other units. The standardized order form was the only way to order insulin or to modify the insulin regimen on the IG unit. The frequency of review or revision of the insulin orders was left to the discretion of the inpatient physicians.

Physician/Midlevel Provider Education

Physicians and midlevel providers caring for patients in the IG and the CCG were given specific education about the best practice recommendations for the management of diabetes and hyperglycemia in hospitalized patients. This education was based on the principles of anticipatory, physiologic insulin use. On nonhouse staff services, the education was provided to the attending physicians and midlevel providers, and on house staff services, the education was provided to the residents. All physician education was provided by the physician authors (D.W. and R.G.). A summary of the content of the physician education is provided in Supporting Information Appendix 2.

Nurse Education

Nurses caring for patients in the IG were given education similar to that which was provided to the physicians (see above), with an emphasis on practical issues related to delivering physiologic insulin. It included topics such as blood glucose monitoring, and the real‐time manipulation of nutritional insulin doses in accordance with the clinical situation (decision‐making that was specifically delegated to the nursing staff by the order set).

Patients

Patients were eligible for inclusion in the analysis if they met the following inclusion criteria: they were admitted to the inpatient General Internal Medicine Services; subcutaneous insulin was provided to the patient during the hospitalization; they had at least 2 blood glucose values >180 mg/dL; they were discharged from the hospital on a pharmacologic glucose lowering agent (insulin or oral); and their total length‐of‐stay was 3 days to 14 days. Patients were excluded from the analysis if they were admitted with a primary diagnosis of diabetic ketoacidosis, diabetic hyperosmolar state, or hypoglycemia. Up to 10 consecutive days of glucose data were recorded for each patient, and the first day on which blood glucose information was available from the admitting floor was excluded from the analysis. Also, specific patient‐days were not analyzed if there were no bedside glucoses recorded, or if the patient was treated with an IV insulin infusion on that day.

Outcomes

The primary outcome was glycemic control. The primary unit of measure was the patient‐day (ie, all of the information for 1 patient on a single qualifying day). This was done to correct for the phenomenon of frequent repeat testing in response to abnormal values. It also allows for a more clinically relevant description of the actual glycemic control on a given day. Specifically, each patient‐day was categorized as in‐range (70‐180 mg/dL), hyperglycemic (>180 mg/dL), severely hyperglycemic (>250 mg/dL), hypoglycemic (<70 mg/dL), and/or severely hypoglycemic (<50 mg/dL). The primary endpoint was glycemic control in‐range. For a patient‐day to be in‐range, all readings for that particular day were within 70 mg/dL to180 mg/dL. For the readings that were not in the desired range, a minimum of 1 deviant reading in a particular day constituted classification into that category, and a single out‐of‐range patient‐day could be included in 1 or more of the out‐of‐range categories (eg, a patient‐day could be categorized as both severely hyperglycemic and hypoglycemic if it contained glucose readings in both of those ranges).

The day‐weighted mean blood glucose value was also calculated for each of the groups. This calculation utilized the mean blood glucose for each patient‐day, and then averaged these values for each group. These metrics have been endorsed as appropriate measures of glycemic control by the SHM Glycemic Control Task Force.20

Other Data

Several other clinical features were also examined, including the following: primary diagnoses listed in the hospital discharge summary for each patient (3 maximum); possible confounders including patient weight, length‐of‐stay, days receiving tube feeds, days receiving parenteral nutrition, and days during which patients were treated with high‐dose glucocorticoids (>10 mg/day of prednisone, or its equivalent) or oral diabetes medications; and the composition of the insulin regimen on each hospital day. Definitions of insulin regimens are provided in Table 1.

Definitions of the Insulin Regimens Prescribed for Each Patient‐Day
  • Correctional insulin (also known as sliding scale or as needed insulin) was allowed as part of any insulin regimen above. Correctional insulin was only recorded when it was unaccompanied by a scheduled insulin.

Any basal insulin dayAny day in which intermediate‐acting or long‐acting, scheduled insulin was given.
Basal insulin alone dayA day in which intermediate‐acting or long‐acting insulin was the only scheduled insulin given.
Any nutritional insulin dayAny day in which rapid‐acting or short‐acting, scheduled insulin was given.
Nutritional insulin alone dayA day in which rapid‐acting or short‐acting insulin was the only scheduled insulin given.
Basal plus nutritional dayA day in which both scheduled, intermediate‐acting or long‐acting insulin and scheduled, rapid‐acting or short‐acting insulin were given.
Pre‐mixed insulin dayAny day in which a pre‐mixed combination insulin was given.
Basal plus nutritional or pre‐mixed insulin dayA composite of the basal plus nutritional day category and the mixed insulin day category described above. This group includes any day in which either a pre‐mixed combination insulin was given OR a day in which both: (a) scheduled, intermediate‐acting or long‐acting and (b) scheduled, rapid‐acting or short‐acting insulin were given.
Sliding scale insulin alone day*Any day when only correctional (as needed) insulin was given.

Statistical Analysis

Bivariate analyses (chi‐square, and t‐tests) were carried out to compare demographic characteristics of the intervention and control populations. Since there were multiple glucose readings nested within individuals, multilevel mixed‐effects logistic regression was used to evaluate the association between the intervention and outcomes. A 2‐level hierarchical model was developed in which patient‐days were nested within patients; this accounted for the correlation between glycemic control across days for a given patient. Patient‐day was modeled as a random intercept and the log likelihood was estimated using adaptive Gaussian quadrature with 7 integration points. Alpha was set at 0.05, 2‐tailed. The final model was adjusted for gender, age, weight, length‐of‐stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was too infrequent to be considered in the adjusted analysis. All analyses were conducted in Stata/IC 10.0 (College Station, TX).

Results

A total of 245 patients provided 1315 patient‐days. Patient demographics are shown in Table 2. The patients' weight, length‐of‐stay, and primary diagnoses were similar across the 3 groups. There was a higher percentage of males in the IG as compared to the HCG.

Demographic Characteristics by Group
 IGCCGP Value IG vs. CCGHCGP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

  • Misc/other includes: malignancies, fluid/electrolyte, hematologic, transplant‐related, cardiac, venous thromboemolism, pulmonary, and dermatologic.

Number of patients8486 75 
Age, years, mean (SD)59.3 (15.3)60.4 (15.9)0.6859.2 (17.2)0.96
Range (n = 245)18‐9420‐87 24‐92 
Weight in kg, mean (SD)92.2 (29.5)89.5 (27.2)0.5794.2 (35.4)0.69
Range (n = 237)40‐19840‐188 42‐235 
Sex, n (%) (n = 245)  0.17 0.04
Male45 (53.6)37 (43.0) 28 (37.3) 
Female39(46.4)49 (57.0) 47 (62.7) 
Length of stay, mean (SD)7.6 (3.3)7.4 (3.0)0.627.0 (2.5)0.14
Range (n = 245)4‐154‐15 4‐14 
Number of diagnoses169158 160 
Primary diagnoses, n (%)  0.56 0.10
Infections40 (23.7)45 (28.5) 49 (30.6) 
Gastrointestinal33 (19.5)19 (12.0) 14 (8.8) 
Rheumatologic13 (7.7)12 (7.6) 18 (11.2) 
Renal14 (8.3)10 (6.3) 16 (10.0) 
Diabetes‐related11 (6.5)11 (7.0) 10 (6.2) 
Neurologic8 (4.7)11 (7.0) 11 (6.9) 
*Misc/other50 (29.6)50 (31.6) 42 (26.3) 

Table 3 shows the insulin regimens used in the different groups. The use of basal insulin was similar between groups. Congruent with the goals of the education session and the order set, patients in the IG were more likely to be treated with a combination of basal and nutritional insulin than patients in the other groups. Patients in the HCG were more likely to be treated with a premixed insulin than patients in the other groups. However, even when premixed insulin was categorized as a form of basal plus nutritional insulin and combined into a composite group with the combined basal and nutritional days, this type of regimen remained more common in the IG than in the HCG. The rate of sliding scale insulin use alone (ie, without any scheduled insulin) was similar in the 3 groups.

Insulin Regimen, Oral Diabetes Agent Use and Nutritional Information by Group
Patient‐days on the followingIG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions of the insulin regimens are shown in Table 1.

  • Abbreviations: IG, intervention group; CCG, concurrent control group; HCG, historic control group.

Sliding scale alone, n (%)105 (23.2)130 (27.6)0.1289 (22.8)0.89
Basal alone, n (%)132 (29.1)231 (49.0)<0.01199 (50.9)<0.01
Nutritional alone, n (%)22 (4.9)5 (1.1)<0.018 (2.0)0.03
Basal plus nutritional, n (%)166 (36.6)71 (15.1)<0.0114 (3.6)<0.01
Pre‐mixed insulin included, n (%)27 (6.0)32 (6.8)0.6078 (20.0)<0.01
No insulin, n (%)1 (<1)2 (<1)0.593 (<1)0.28
Any basal, n (%)325 (71.7)334 (70.9)0.78291 (74.4)0.38
Any nutritional, n (%)215 (47.5)108 (22.9)<0.01100 (25.6)<0.01
Basal plus nutritional or pre‐mixed, n (%)193 (42.6)103 (21.9)<0.0192 (23.5)<0.01
Oral diabetes agents, n (%)79 (17.4)83 (17.6)0.9474 (18.9)0.58
Sulfonylureas, n (%)40 (8.8)63 (13.4)0.0337 (9.5)0.75
Parenteral nutrition/tube feeds, n (%)0 (0)18 (3.8) 8 (2.0) 
High dose corticosteroids, n (%)66 (14.6)93 (19.8)0.0451 (13.0)0.52

Other relevant measures are also shown in Table 3. The use of oral diabetes agents was similar in the 3 groups. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was infrequent.

A comparison of glycemic control in the three groups is shown in Table 4. In contrast to the CCG, patients in the IG experienced more days within the target glucose range (17% vs. 10.6%, P < 0.01), fewer days with severe hyperglycemia (48.3% vs. 59.2%, P < 0.01), and had a lower day‐weighted average blood glucose (195.9 vs. 212.6, P < 0.01). Compared to the HCG, patients in the IG experienced similar rates of hyperglycemia, but fewer hypoglycemic days (5.1% vs. 9.2%, P = 0.02).

Glycemic Control by Group
 IG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions for glycemic control are provided in the text.

  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

Patient‐days     
In range, n (%)77 (17.0)50 (10.6)<0.0166 (16.9)0.98
Out of range, n (%)376 (83.0)421 (89.4)<0.01325 (83.1)0.98
Hyperglycemic, n (%)289 (63.8)310 (65.8)0.52248 (63.4)0.91
Severely hyperglycemic, n (%)219 (48.3)279 (59.2)<0.01176 (45.0)0.32
Hypoglycemic, n (%)23 (5.1)36 (7.6)0.1136 (9.2)0.02
Severely hypoglycemic, n (%)13 (2.9)10 (2.1)0.4715 (3.8)0.44
Day weighted average blood glucose (SD)195.9 (66.8)212.6 (73.4)<0.01190.5 (63.1)0.25

The percentages of patients with severe hyperglycemia in each group are shown in Figure 1 by hospital day. Severe hyperglycemia was common, but there was a trend towards a decrease in the prevalence of severe hyperglycemia with increasing hospital days for all study groups, although it was consistently higher in the CCG than in the IG. Figure 2 shows the types of insulin regimens used by hospital day (composite for all groups). The use of basal plus nutritional insulin (the recommended regimen) increased gradually with increasing hospital days. When taken together, the information in both figures support the hypothesis that the use of the recommended insulin regimen may have contributed to the modest improvements in glycemic control seen in the IG.

Figure 1
Severe Hyperglycemia by hospital day.
Figure 2
Regimen by hospital day.

In the final adjusted regression model, the intervention had a positive impact on glycemic control (Table 5). Subjects in the IG had a 72% increase in the odds of being in the target glucose range when compared to subjects in the CCG (P = 0.01). In addition, subjects in the IG had a 35% reduction in the odds of being severely hyperglycemic when compared to those in the CCG (P < 0.01). Finally, the odds ratio (OR) for being hypoglycemic among intervention subjects was 0.59 (P = 0.06) when compared to subjects in the CCG and 0.48 (P = 0.01) when compared to subjects in the HCG.

Multivariable Analysis of Glycemic Control
 Adjusted OR* IG vs. CCG95% CIP value IG vs. CCGAdjusted OR* IG vs. HCG95% CIP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; CI, confidence interval; HCG, historic control group; IG, intervention group; OR, odds ratio.

  • Adjusted for gender, age, weight, length of stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids.

  • This odds ratio reflects the unadjusted analysis as this model failed to converge after adjusting for the covariates.

In range1.721.16,2.550.011.080.74,1.580.68
Hyperglycemic0.930.70,1.220.580.950.71,1.280.74
Severely Hyperglycemic0.650.49,0.85<0.011.100.82,1.470.52
Hypoglycemic0.590.34,1.020.060.480.27,0.850.01
Severely Hypoglycemic1.360.59,3.140.470.970.29,1.440.28

Discussion

In this study, we investigated the effects of a standardized insulin order set, coupled with physician and nurse education, on glycemic control in hyperglycemic hospitalized patients. These interventions were designed to encourage a standardized approach to the treatment of hyperglycemia in hospitalized patients, based on the principles of physiologic insulin use, as described above. Our data suggest that the interventions did, indeed, alter the way insulin was prescribed, as more patients in the IG received a combination of basal plus nutritional insulin (the recommended regimen) than in the other groups. These interventions were associated with improved glycemic outcomes in the IG as compared to the CCG. The IG experienced a higher percentage of days in the target range and a trend toward fewer hypoglycemic days than the CCG. Although the IG experienced a similar percentage of days in the target range, it had significantly fewer hypoglycemic days than the HCG.

It is useful to consider the results of our study in the context of 2 other similar studies performed by Schnipper et al.14 and Maynard et al.15 Although each of these 3 studies have different study designs, they are similar in intent (to test the effects of simple quality improvement interventions on glycemic control in the hospital) and results (all showed significant improvements in some aspect of glycemic control). In our study, and the study by Maynard et al.,15 the interventions also led to decreases in the rates of hypoglycemia, whereas Schnipper et al.14 observed no difference in hypoglycemia. Of interest, in each of the three studies the interventions were associated with an increase in the use of some type of scheduled insulin. In our study and the study from Schnipper et al.14 the baseline use of basal insulin was quite high, and the interventions were associated with a significant increase in the addition of nutritional insulin. In the Maynard et al.15 study, the baseline use of sliding scale insulin alone was prevalent, and the interventions resulted in an increase in the use of basal insulin. The results of these studies, taken together, prompt us to conclude that the interventions employed in these studies are likely to lead to more frequent prescription of scheduled (anticipatory) insulin, and a modest improvement in glycemic control, without an increase (and perhaps with a decrease) in hypoglycemia.

A few of our study results are unexpected, or difficult to explain. In contrast to the other studies discussed above, our interventions did not affect the frequency of the use of sliding‐scale insulin alone (without any scheduled insulin), which was similar in the 3 groups. Although the reason for this is uncertain, we hypothesize that the high baseline use of basal insulin in our institution, and the lack of a hard stop preventing the use of sliding scale insulin alone explain this finding. Also, it is difficult to explain why measures of hyperglycemia were similar between the IG and the HCG despite the fact that the HCG was less often treated with a combination of basal and nutritional insulin and more often treated with mixed insulin.

There are several different mechanisms by which the interventions might have resulted in improved glycemic control in the IG compared to CCG. Our data clearly shows that insulin was prescribed differently in the IG, and the more frequent use of a combination of scheduled basal and nutritional insulin might have contributed to the differences between the groups. However, the effects of our interventions clearly went beyond physician education into the realm of true process improvement and standardization. The standardized order form was designed to prompt physicians to use a basal‐bolus insulin regimen. The order form also created nursing expectations of how insulin should be ordered, and clarified the roles of the different insulins that were prescribed.

On the medication administration record, each insulin was labeled as basal insulin (to be given even when fasting) or nutritional insulin (to be given along with the meal). The nurses caring for the IG also attended an education program that reinforced the role of the nurse in the bedside management of insulin administration. Specifically, nurses were taught to assess the premeal blood glucose and the patient's nutritional situation before giving the nutritional insulin (ie, Does the patient have food available? Will he tolerate eating the food?). In situations where is was not clear if the patient would be able to tolerate the ordered nutrition, the order set empowered the nurse to give the nutritional insulin after the meal, and to reduce the dose to match the patient's actual intake. These interventions resulted in some fundamental improvements in the nursing process of delivering insulin to the patient, and these changes might have resulted in improvements via mechanisms that are difficult to directly measure. Since the same physicians cared for both the IG and the CCG, interventions other than physician education clearly contributed to the observed improvements in the IG.

This study was not a randomized study, and there could be important undetected differences between the groups. However, all of the patients were admitted to the General Medicine Inpatient Services and the comparison of the general patient demographics and primary diagnoses between the groups do not suggest major differences.

Although the improvements in glycemic control seen in this study were statistically significant, they were quantitatively modest. The rates of hyperglycemia seen in this study, on the other hand, are quite remarkable. Both the American Diabetes Association and the American College of Endocrinology have recommended that blood glucoses in hospitalized patients not exceed a maximum value of 180 mg/dL, but the day‐weighted average blood glucose in this study was above that for each group. Even in the IG, over 80% of all patient‐days included at least 1 blood glucose value outside of the target range. These data suggest that better strategies for achieving metabolic control in hospitalized patients are needed.

It is worth mentioning that our interventions were not aggressively enforced. While the use of the order set was mandatory for the IG, it was flexible enough to allow for substantial practice variation, especially with respect to the dose of insulin prescribed. Although the education sessions discussed the specifics of insulin dosing in hospitalized patients, the order form did not offer dosing guidelines. It is possible that our interventions may have had a larger impact if a starting dose of insulin had been specified on the form. Although the insulin order form prompted physicians to act, there were no forced functions. Also, not all house staff attended the education sessions for physicians, and there was no feedback provided to physicians related to how they might improve their adherence to the recommendations presented in the educational module. Therefore, it is likely that more aggressive interventions could have led to greater changes in physician practice.

In conclusion, this study demonstrates that interventions including physician and nurse education and a standardized insulin order set can lead to improvement in glycemic control and patient safety in hospitalized patients treated with subcutaneous insulin. However, the observed improvements are modest, and poor metabolic control remains common, despite these interventions. These data suggest that standardization of the process of ordering and delivering subcutaneous insulin in the hospital may lead to a reduction in both hyperglycemia and hypoglycemia. However, it is clear that the interventions used in this study were not potent enough to achieve the recommended glycemic targets for the majority of patients. Additional research is needed to determine the best strategy for achieving safe and effective metabolic control in hospitalized, hyperglycemic, noncritically ill patients.

Acknowledgements

The authors thank David Conway for his work in data collection and management.

Hyperglycemia is common in hospitalized patients, and hyperglycemia has been associated with poor hospital outcomes. The adverse physiologic effects of acute hyperglycemia are well established1 and several clinical studies have linked hyperglycemia with poor clinical outcomes in certain patient populations.28 Although the optimal target range for inpatient glycemic control has not yet been defined, these studies support the goal of metabolic control for hospital patients. However, there are many barriers to achieving adequate glycemic control in the hospital, and blood glucoses in the hospital are often far from recommended targets.9, 10 One barrier appears to be the low priority given to glycemic control in the hospital. Hyperglycemia in the hospital is often ignored,11 and insulin regimens are often chosen for simplicity as opposed to effectiveness.12 Other barriers to glycemic control in the hospital include the physiologic effects (stress) of acute illness, and the frequent nutritional changes and interruptions that occur.

Most hyperglycemic patients on a general medicine unit are treated with subcutaneous insulin, but the optimal strategy for prescribing insulin in the hospital remains uncertain. A technical review of the literature on the management of diabetes in the hospital setting from 2004 recommends prescribing insulin in a way that mimics physiologic insulin secretion (ie, physiologic or basal‐bolus insulin).1 This approach has been promulgated by experts, but there has been very little research to support these recommendations. One small, randomized trial concluded that a basal‐bolus approach achieved better glycemic control than the use of sliding‐scale insulin alone,13 and 2 quality improvement studies using a before/after design have demonstrated improvements in glycemic control after the implementation of interventions designed to encourage physiologic insulin use.14, 15

In this study we hypothesized that a few simple interventions (education for physicians and nurses, and a standardized insulin order form) would lead to a higher rate of basal‐bolus insulin use and simultaneously improve glycemic control and patient safety.

Methods

Study Design

This study was performed at the University of Michigan Hospital over a 6‐month period, and the protocol was approved by the Institutional Review Board. We performed a quasi‐experimental study comparing 3 patient groups. The intervention group (IG) was subject to all of the interventions discussed below (physician education, nurse education, and the standardized order form). The concurrent control group (CCG) was hospitalized during the same time period as the IG, but was only subject to 1 of the interventions (physician education). These patients were cared for by the same physicians as the IG, but on a different unit where the nurses had not received the education and where the standardized insulin order form was not available. Patients were admitted to the IG unit or the CCG unit via the institution's usual admission process. In addition, we examined an historic control group (HCG) which was hospitalized during the same months of the year, but 2 years prior. The HCG was not subject to any of the interventions.

Interventions

Standardized Subcutaneous Insulin Order Form

This form (Supporting Information Appendix 1) was designed to encourage physicians to prescribe insulin in a physiologic way, providing basal, nutritional, and correctional insulin. The form is based on best practice guidelines,1 and is in agreement with the principles of the inpatient management of diabetes and hyperglycemia endorsed by several professional organizations.16, 17 The form was engineered by a multidisciplinary team, including an endocrinologist, several hospitalists, several nurses, a certified diabetes educator, a pharmacist, and others. It is derived from the extensive experience of the University of Michigan Hospital Intensive Insulin Program (HIIP) in the Division of Endocrinology, and on work done by the Society of Hospital Medicine (SHM) Glycemic Control Task Force.1719 This form was only used in the care of patients in the IG. The form, which was not approved for use on other floors, did not creep to other units. The standardized order form was the only way to order insulin or to modify the insulin regimen on the IG unit. The frequency of review or revision of the insulin orders was left to the discretion of the inpatient physicians.

Physician/Midlevel Provider Education

Physicians and midlevel providers caring for patients in the IG and the CCG were given specific education about the best practice recommendations for the management of diabetes and hyperglycemia in hospitalized patients. This education was based on the principles of anticipatory, physiologic insulin use. On nonhouse staff services, the education was provided to the attending physicians and midlevel providers, and on house staff services, the education was provided to the residents. All physician education was provided by the physician authors (D.W. and R.G.). A summary of the content of the physician education is provided in Supporting Information Appendix 2.

Nurse Education

Nurses caring for patients in the IG were given education similar to that which was provided to the physicians (see above), with an emphasis on practical issues related to delivering physiologic insulin. It included topics such as blood glucose monitoring, and the real‐time manipulation of nutritional insulin doses in accordance with the clinical situation (decision‐making that was specifically delegated to the nursing staff by the order set).

Patients

Patients were eligible for inclusion in the analysis if they met the following inclusion criteria: they were admitted to the inpatient General Internal Medicine Services; subcutaneous insulin was provided to the patient during the hospitalization; they had at least 2 blood glucose values >180 mg/dL; they were discharged from the hospital on a pharmacologic glucose lowering agent (insulin or oral); and their total length‐of‐stay was 3 days to 14 days. Patients were excluded from the analysis if they were admitted with a primary diagnosis of diabetic ketoacidosis, diabetic hyperosmolar state, or hypoglycemia. Up to 10 consecutive days of glucose data were recorded for each patient, and the first day on which blood glucose information was available from the admitting floor was excluded from the analysis. Also, specific patient‐days were not analyzed if there were no bedside glucoses recorded, or if the patient was treated with an IV insulin infusion on that day.

Outcomes

The primary outcome was glycemic control. The primary unit of measure was the patient‐day (ie, all of the information for 1 patient on a single qualifying day). This was done to correct for the phenomenon of frequent repeat testing in response to abnormal values. It also allows for a more clinically relevant description of the actual glycemic control on a given day. Specifically, each patient‐day was categorized as in‐range (70‐180 mg/dL), hyperglycemic (>180 mg/dL), severely hyperglycemic (>250 mg/dL), hypoglycemic (<70 mg/dL), and/or severely hypoglycemic (<50 mg/dL). The primary endpoint was glycemic control in‐range. For a patient‐day to be in‐range, all readings for that particular day were within 70 mg/dL to180 mg/dL. For the readings that were not in the desired range, a minimum of 1 deviant reading in a particular day constituted classification into that category, and a single out‐of‐range patient‐day could be included in 1 or more of the out‐of‐range categories (eg, a patient‐day could be categorized as both severely hyperglycemic and hypoglycemic if it contained glucose readings in both of those ranges).

The day‐weighted mean blood glucose value was also calculated for each of the groups. This calculation utilized the mean blood glucose for each patient‐day, and then averaged these values for each group. These metrics have been endorsed as appropriate measures of glycemic control by the SHM Glycemic Control Task Force.20

Other Data

Several other clinical features were also examined, including the following: primary diagnoses listed in the hospital discharge summary for each patient (3 maximum); possible confounders including patient weight, length‐of‐stay, days receiving tube feeds, days receiving parenteral nutrition, and days during which patients were treated with high‐dose glucocorticoids (>10 mg/day of prednisone, or its equivalent) or oral diabetes medications; and the composition of the insulin regimen on each hospital day. Definitions of insulin regimens are provided in Table 1.

Definitions of the Insulin Regimens Prescribed for Each Patient‐Day
  • Correctional insulin (also known as sliding scale or as needed insulin) was allowed as part of any insulin regimen above. Correctional insulin was only recorded when it was unaccompanied by a scheduled insulin.

Any basal insulin dayAny day in which intermediate‐acting or long‐acting, scheduled insulin was given.
Basal insulin alone dayA day in which intermediate‐acting or long‐acting insulin was the only scheduled insulin given.
Any nutritional insulin dayAny day in which rapid‐acting or short‐acting, scheduled insulin was given.
Nutritional insulin alone dayA day in which rapid‐acting or short‐acting insulin was the only scheduled insulin given.
Basal plus nutritional dayA day in which both scheduled, intermediate‐acting or long‐acting insulin and scheduled, rapid‐acting or short‐acting insulin were given.
Pre‐mixed insulin dayAny day in which a pre‐mixed combination insulin was given.
Basal plus nutritional or pre‐mixed insulin dayA composite of the basal plus nutritional day category and the mixed insulin day category described above. This group includes any day in which either a pre‐mixed combination insulin was given OR a day in which both: (a) scheduled, intermediate‐acting or long‐acting and (b) scheduled, rapid‐acting or short‐acting insulin were given.
Sliding scale insulin alone day*Any day when only correctional (as needed) insulin was given.

Statistical Analysis

Bivariate analyses (chi‐square, and t‐tests) were carried out to compare demographic characteristics of the intervention and control populations. Since there were multiple glucose readings nested within individuals, multilevel mixed‐effects logistic regression was used to evaluate the association between the intervention and outcomes. A 2‐level hierarchical model was developed in which patient‐days were nested within patients; this accounted for the correlation between glycemic control across days for a given patient. Patient‐day was modeled as a random intercept and the log likelihood was estimated using adaptive Gaussian quadrature with 7 integration points. Alpha was set at 0.05, 2‐tailed. The final model was adjusted for gender, age, weight, length‐of‐stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was too infrequent to be considered in the adjusted analysis. All analyses were conducted in Stata/IC 10.0 (College Station, TX).

Results

A total of 245 patients provided 1315 patient‐days. Patient demographics are shown in Table 2. The patients' weight, length‐of‐stay, and primary diagnoses were similar across the 3 groups. There was a higher percentage of males in the IG as compared to the HCG.

Demographic Characteristics by Group
 IGCCGP Value IG vs. CCGHCGP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

  • Misc/other includes: malignancies, fluid/electrolyte, hematologic, transplant‐related, cardiac, venous thromboemolism, pulmonary, and dermatologic.

Number of patients8486 75 
Age, years, mean (SD)59.3 (15.3)60.4 (15.9)0.6859.2 (17.2)0.96
Range (n = 245)18‐9420‐87 24‐92 
Weight in kg, mean (SD)92.2 (29.5)89.5 (27.2)0.5794.2 (35.4)0.69
Range (n = 237)40‐19840‐188 42‐235 
Sex, n (%) (n = 245)  0.17 0.04
Male45 (53.6)37 (43.0) 28 (37.3) 
Female39(46.4)49 (57.0) 47 (62.7) 
Length of stay, mean (SD)7.6 (3.3)7.4 (3.0)0.627.0 (2.5)0.14
Range (n = 245)4‐154‐15 4‐14 
Number of diagnoses169158 160 
Primary diagnoses, n (%)  0.56 0.10
Infections40 (23.7)45 (28.5) 49 (30.6) 
Gastrointestinal33 (19.5)19 (12.0) 14 (8.8) 
Rheumatologic13 (7.7)12 (7.6) 18 (11.2) 
Renal14 (8.3)10 (6.3) 16 (10.0) 
Diabetes‐related11 (6.5)11 (7.0) 10 (6.2) 
Neurologic8 (4.7)11 (7.0) 11 (6.9) 
*Misc/other50 (29.6)50 (31.6) 42 (26.3) 

Table 3 shows the insulin regimens used in the different groups. The use of basal insulin was similar between groups. Congruent with the goals of the education session and the order set, patients in the IG were more likely to be treated with a combination of basal and nutritional insulin than patients in the other groups. Patients in the HCG were more likely to be treated with a premixed insulin than patients in the other groups. However, even when premixed insulin was categorized as a form of basal plus nutritional insulin and combined into a composite group with the combined basal and nutritional days, this type of regimen remained more common in the IG than in the HCG. The rate of sliding scale insulin use alone (ie, without any scheduled insulin) was similar in the 3 groups.

Insulin Regimen, Oral Diabetes Agent Use and Nutritional Information by Group
Patient‐days on the followingIG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions of the insulin regimens are shown in Table 1.

  • Abbreviations: IG, intervention group; CCG, concurrent control group; HCG, historic control group.

Sliding scale alone, n (%)105 (23.2)130 (27.6)0.1289 (22.8)0.89
Basal alone, n (%)132 (29.1)231 (49.0)<0.01199 (50.9)<0.01
Nutritional alone, n (%)22 (4.9)5 (1.1)<0.018 (2.0)0.03
Basal plus nutritional, n (%)166 (36.6)71 (15.1)<0.0114 (3.6)<0.01
Pre‐mixed insulin included, n (%)27 (6.0)32 (6.8)0.6078 (20.0)<0.01
No insulin, n (%)1 (<1)2 (<1)0.593 (<1)0.28
Any basal, n (%)325 (71.7)334 (70.9)0.78291 (74.4)0.38
Any nutritional, n (%)215 (47.5)108 (22.9)<0.01100 (25.6)<0.01
Basal plus nutritional or pre‐mixed, n (%)193 (42.6)103 (21.9)<0.0192 (23.5)<0.01
Oral diabetes agents, n (%)79 (17.4)83 (17.6)0.9474 (18.9)0.58
Sulfonylureas, n (%)40 (8.8)63 (13.4)0.0337 (9.5)0.75
Parenteral nutrition/tube feeds, n (%)0 (0)18 (3.8) 8 (2.0) 
High dose corticosteroids, n (%)66 (14.6)93 (19.8)0.0451 (13.0)0.52

Other relevant measures are also shown in Table 3. The use of oral diabetes agents was similar in the 3 groups. The use of a nonoral feeding route (eg, tube feeding or parenteral nutrition) was infrequent.

A comparison of glycemic control in the three groups is shown in Table 4. In contrast to the CCG, patients in the IG experienced more days within the target glucose range (17% vs. 10.6%, P < 0.01), fewer days with severe hyperglycemia (48.3% vs. 59.2%, P < 0.01), and had a lower day‐weighted average blood glucose (195.9 vs. 212.6, P < 0.01). Compared to the HCG, patients in the IG experienced similar rates of hyperglycemia, but fewer hypoglycemic days (5.1% vs. 9.2%, P = 0.02).

Glycemic Control by Group
 IG (n = 453)CCG (n = 471)P Value IG vs. CCGHCG (n = 391)P Value IG vs. HCG
  • NOTE: Definitions for glycemic control are provided in the text.

  • Abbreviations: CCG, concurrent control group; HCG, historic control group; IG, intervention group; SD, standard deviation.

Patient‐days     
In range, n (%)77 (17.0)50 (10.6)<0.0166 (16.9)0.98
Out of range, n (%)376 (83.0)421 (89.4)<0.01325 (83.1)0.98
Hyperglycemic, n (%)289 (63.8)310 (65.8)0.52248 (63.4)0.91
Severely hyperglycemic, n (%)219 (48.3)279 (59.2)<0.01176 (45.0)0.32
Hypoglycemic, n (%)23 (5.1)36 (7.6)0.1136 (9.2)0.02
Severely hypoglycemic, n (%)13 (2.9)10 (2.1)0.4715 (3.8)0.44
Day weighted average blood glucose (SD)195.9 (66.8)212.6 (73.4)<0.01190.5 (63.1)0.25

The percentages of patients with severe hyperglycemia in each group are shown in Figure 1 by hospital day. Severe hyperglycemia was common, but there was a trend towards a decrease in the prevalence of severe hyperglycemia with increasing hospital days for all study groups, although it was consistently higher in the CCG than in the IG. Figure 2 shows the types of insulin regimens used by hospital day (composite for all groups). The use of basal plus nutritional insulin (the recommended regimen) increased gradually with increasing hospital days. When taken together, the information in both figures support the hypothesis that the use of the recommended insulin regimen may have contributed to the modest improvements in glycemic control seen in the IG.

Figure 1
Severe Hyperglycemia by hospital day.
Figure 2
Regimen by hospital day.

In the final adjusted regression model, the intervention had a positive impact on glycemic control (Table 5). Subjects in the IG had a 72% increase in the odds of being in the target glucose range when compared to subjects in the CCG (P = 0.01). In addition, subjects in the IG had a 35% reduction in the odds of being severely hyperglycemic when compared to those in the CCG (P < 0.01). Finally, the odds ratio (OR) for being hypoglycemic among intervention subjects was 0.59 (P = 0.06) when compared to subjects in the CCG and 0.48 (P = 0.01) when compared to subjects in the HCG.

Multivariable Analysis of Glycemic Control
 Adjusted OR* IG vs. CCG95% CIP value IG vs. CCGAdjusted OR* IG vs. HCG95% CIP Value IG vs. HCG
  • Abbreviations: CCG, concurrent control group; CI, confidence interval; HCG, historic control group; IG, intervention group; OR, odds ratio.

  • Adjusted for gender, age, weight, length of stay, use of oral diabetes agents, use of sulfonylureas, and use of high‐dose corticosteroids.

  • This odds ratio reflects the unadjusted analysis as this model failed to converge after adjusting for the covariates.

In range1.721.16,2.550.011.080.74,1.580.68
Hyperglycemic0.930.70,1.220.580.950.71,1.280.74
Severely Hyperglycemic0.650.49,0.85<0.011.100.82,1.470.52
Hypoglycemic0.590.34,1.020.060.480.27,0.850.01
Severely Hypoglycemic1.360.59,3.140.470.970.29,1.440.28

Discussion

In this study, we investigated the effects of a standardized insulin order set, coupled with physician and nurse education, on glycemic control in hyperglycemic hospitalized patients. These interventions were designed to encourage a standardized approach to the treatment of hyperglycemia in hospitalized patients, based on the principles of physiologic insulin use, as described above. Our data suggest that the interventions did, indeed, alter the way insulin was prescribed, as more patients in the IG received a combination of basal plus nutritional insulin (the recommended regimen) than in the other groups. These interventions were associated with improved glycemic outcomes in the IG as compared to the CCG. The IG experienced a higher percentage of days in the target range and a trend toward fewer hypoglycemic days than the CCG. Although the IG experienced a similar percentage of days in the target range, it had significantly fewer hypoglycemic days than the HCG.

It is useful to consider the results of our study in the context of 2 other similar studies performed by Schnipper et al.14 and Maynard et al.15 Although each of these 3 studies have different study designs, they are similar in intent (to test the effects of simple quality improvement interventions on glycemic control in the hospital) and results (all showed significant improvements in some aspect of glycemic control). In our study, and the study by Maynard et al.,15 the interventions also led to decreases in the rates of hypoglycemia, whereas Schnipper et al.14 observed no difference in hypoglycemia. Of interest, in each of the three studies the interventions were associated with an increase in the use of some type of scheduled insulin. In our study and the study from Schnipper et al.14 the baseline use of basal insulin was quite high, and the interventions were associated with a significant increase in the addition of nutritional insulin. In the Maynard et al.15 study, the baseline use of sliding scale insulin alone was prevalent, and the interventions resulted in an increase in the use of basal insulin. The results of these studies, taken together, prompt us to conclude that the interventions employed in these studies are likely to lead to more frequent prescription of scheduled (anticipatory) insulin, and a modest improvement in glycemic control, without an increase (and perhaps with a decrease) in hypoglycemia.

A few of our study results are unexpected, or difficult to explain. In contrast to the other studies discussed above, our interventions did not affect the frequency of the use of sliding‐scale insulin alone (without any scheduled insulin), which was similar in the 3 groups. Although the reason for this is uncertain, we hypothesize that the high baseline use of basal insulin in our institution, and the lack of a hard stop preventing the use of sliding scale insulin alone explain this finding. Also, it is difficult to explain why measures of hyperglycemia were similar between the IG and the HCG despite the fact that the HCG was less often treated with a combination of basal and nutritional insulin and more often treated with mixed insulin.

There are several different mechanisms by which the interventions might have resulted in improved glycemic control in the IG compared to CCG. Our data clearly shows that insulin was prescribed differently in the IG, and the more frequent use of a combination of scheduled basal and nutritional insulin might have contributed to the differences between the groups. However, the effects of our interventions clearly went beyond physician education into the realm of true process improvement and standardization. The standardized order form was designed to prompt physicians to use a basal‐bolus insulin regimen. The order form also created nursing expectations of how insulin should be ordered, and clarified the roles of the different insulins that were prescribed.

On the medication administration record, each insulin was labeled as basal insulin (to be given even when fasting) or nutritional insulin (to be given along with the meal). The nurses caring for the IG also attended an education program that reinforced the role of the nurse in the bedside management of insulin administration. Specifically, nurses were taught to assess the premeal blood glucose and the patient's nutritional situation before giving the nutritional insulin (ie, Does the patient have food available? Will he tolerate eating the food?). In situations where is was not clear if the patient would be able to tolerate the ordered nutrition, the order set empowered the nurse to give the nutritional insulin after the meal, and to reduce the dose to match the patient's actual intake. These interventions resulted in some fundamental improvements in the nursing process of delivering insulin to the patient, and these changes might have resulted in improvements via mechanisms that are difficult to directly measure. Since the same physicians cared for both the IG and the CCG, interventions other than physician education clearly contributed to the observed improvements in the IG.

This study was not a randomized study, and there could be important undetected differences between the groups. However, all of the patients were admitted to the General Medicine Inpatient Services and the comparison of the general patient demographics and primary diagnoses between the groups do not suggest major differences.

Although the improvements in glycemic control seen in this study were statistically significant, they were quantitatively modest. The rates of hyperglycemia seen in this study, on the other hand, are quite remarkable. Both the American Diabetes Association and the American College of Endocrinology have recommended that blood glucoses in hospitalized patients not exceed a maximum value of 180 mg/dL, but the day‐weighted average blood glucose in this study was above that for each group. Even in the IG, over 80% of all patient‐days included at least 1 blood glucose value outside of the target range. These data suggest that better strategies for achieving metabolic control in hospitalized patients are needed.

It is worth mentioning that our interventions were not aggressively enforced. While the use of the order set was mandatory for the IG, it was flexible enough to allow for substantial practice variation, especially with respect to the dose of insulin prescribed. Although the education sessions discussed the specifics of insulin dosing in hospitalized patients, the order form did not offer dosing guidelines. It is possible that our interventions may have had a larger impact if a starting dose of insulin had been specified on the form. Although the insulin order form prompted physicians to act, there were no forced functions. Also, not all house staff attended the education sessions for physicians, and there was no feedback provided to physicians related to how they might improve their adherence to the recommendations presented in the educational module. Therefore, it is likely that more aggressive interventions could have led to greater changes in physician practice.

In conclusion, this study demonstrates that interventions including physician and nurse education and a standardized insulin order set can lead to improvement in glycemic control and patient safety in hospitalized patients treated with subcutaneous insulin. However, the observed improvements are modest, and poor metabolic control remains common, despite these interventions. These data suggest that standardization of the process of ordering and delivering subcutaneous insulin in the hospital may lead to a reduction in both hyperglycemia and hypoglycemia. However, it is clear that the interventions used in this study were not potent enough to achieve the recommended glycemic targets for the majority of patients. Additional research is needed to determine the best strategy for achieving safe and effective metabolic control in hospitalized, hyperglycemic, noncritically ill patients.

Acknowledgements

The authors thank David Conway for his work in data collection and management.

References
  1. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  2. Van den Berghe G,Wouters P,Weekers F, et al.Intensive insulin therapy in the critically ill patients.N Engl J Med.2001;345:13591367.
  3. Van den Berghe G,Wilmer A,Hermans G, et al.Intensive insulin therapy in the medical ICU.N Engl J Med.2006;354:449461.
  4. Furnary AP,Zerr KJ,Grunkemeier GL,Starr A.Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures.Ann Thorac Surg.1999;67:352360; discussion360–352.
  5. Furnary AP,Gao G,Grunkemeier GL, et al.Continuous insulin infusion reduces mortality in patients with diabetes undergoing coronary artery bypass grafting.J Thorac Cardiovasc Surg.2003;125:10071021.
  6. Umpierrez GE,Isaacs SD,Bazargan N,You X,Thaler LM,Kitabchi AE.Hyperglycemia: an independent marker of in‐hospital mortality in patients with undiagnosed diabetes.J Clin Endocrinol Metab.2002;87:978982.
  7. Pomposelli JJ,Baxter JK,Babineau TJ, et al.Early postoperative glucose control predicts nosocomial infection rate in diabetic patients.JPEN J Parenter Enteral Nutr.1998;22:7781.
  8. McAlister FA,Majumdar SR,Blitz S,Rowe BH,Romney J,Marrie TJ.The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community‐acquired pneumonia.Diabetes Care.2005;28:810815.
  9. Umpierrez G,Maynard G.Glycemic chaos (not glycemic control) still the rule for inpatient care: how do we stop the insanity?J Hosp Med.2006;1:141144.
  10. Boord JB,Greevy RA,Braithwaite SS, et al.Evaluation of hospital glycemic control at US Academic Medical Centers.J Hosp Med.2009;4:3544.
  11. Knecht LA,Gauthier SM,Castro JC, et al.Diabetes care in the hospital: is there clinical inertia?J Hosp Med.2006;1:151160.
  12. Schnipper JL,Barsky EE,Shaykevich S,Fitzmaurice G,Pendergrass ML.Inpatient management of diabetes and hyperglycemia among general medicine patients at a large teaching hospital.J Hosp Med.2006;1:145150.
  13. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  14. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  15. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  16. Garber AJ,Moghissi ES,Bransome ED, et al.American College of Endocrinology position statement on inpatient diabetes and metabolic control.Endocr Pract.2004;10:7782.
  17. Society of Hospital Medicine. Glycemic Control Resource Room. Available at: http://www.hospitalmedicine.org/AM/Template.cfm?Section= Quality_Improvement_Resource_Rooms3(5 Suppl):1728.
  18. Maynard G,Wesorick DH,O'Malley C,Inzucchi SE.Subcutaneous insulin order sets and protocols: effective design and implementation strategies.J Hosp Med.2008;3(5 Suppl):2941.
  19. Schnipper JL,Magee M,Larsen K,Inzucchi SE,Maynard G.Society of Hospital Medicine Glycemic Control Task Force summary: practical recommendations for assessing the impact of glycemic control efforts.J Hosp Med.2008;3(5 Suppl):6675.
References
  1. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  2. Van den Berghe G,Wouters P,Weekers F, et al.Intensive insulin therapy in the critically ill patients.N Engl J Med.2001;345:13591367.
  3. Van den Berghe G,Wilmer A,Hermans G, et al.Intensive insulin therapy in the medical ICU.N Engl J Med.2006;354:449461.
  4. Furnary AP,Zerr KJ,Grunkemeier GL,Starr A.Continuous intravenous insulin infusion reduces the incidence of deep sternal wound infection in diabetic patients after cardiac surgical procedures.Ann Thorac Surg.1999;67:352360; discussion360–352.
  5. Furnary AP,Gao G,Grunkemeier GL, et al.Continuous insulin infusion reduces mortality in patients with diabetes undergoing coronary artery bypass grafting.J Thorac Cardiovasc Surg.2003;125:10071021.
  6. Umpierrez GE,Isaacs SD,Bazargan N,You X,Thaler LM,Kitabchi AE.Hyperglycemia: an independent marker of in‐hospital mortality in patients with undiagnosed diabetes.J Clin Endocrinol Metab.2002;87:978982.
  7. Pomposelli JJ,Baxter JK,Babineau TJ, et al.Early postoperative glucose control predicts nosocomial infection rate in diabetic patients.JPEN J Parenter Enteral Nutr.1998;22:7781.
  8. McAlister FA,Majumdar SR,Blitz S,Rowe BH,Romney J,Marrie TJ.The relation between hyperglycemia and outcomes in 2,471 patients admitted to the hospital with community‐acquired pneumonia.Diabetes Care.2005;28:810815.
  9. Umpierrez G,Maynard G.Glycemic chaos (not glycemic control) still the rule for inpatient care: how do we stop the insanity?J Hosp Med.2006;1:141144.
  10. Boord JB,Greevy RA,Braithwaite SS, et al.Evaluation of hospital glycemic control at US Academic Medical Centers.J Hosp Med.2009;4:3544.
  11. Knecht LA,Gauthier SM,Castro JC, et al.Diabetes care in the hospital: is there clinical inertia?J Hosp Med.2006;1:151160.
  12. Schnipper JL,Barsky EE,Shaykevich S,Fitzmaurice G,Pendergrass ML.Inpatient management of diabetes and hyperglycemia among general medicine patients at a large teaching hospital.J Hosp Med.2006;1:145150.
  13. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  14. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  15. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  16. Garber AJ,Moghissi ES,Bransome ED, et al.American College of Endocrinology position statement on inpatient diabetes and metabolic control.Endocr Pract.2004;10:7782.
  17. Society of Hospital Medicine. Glycemic Control Resource Room. Available at: http://www.hospitalmedicine.org/AM/Template.cfm?Section= Quality_Improvement_Resource_Rooms3(5 Suppl):1728.
  18. Maynard G,Wesorick DH,O'Malley C,Inzucchi SE.Subcutaneous insulin order sets and protocols: effective design and implementation strategies.J Hosp Med.2008;3(5 Suppl):2941.
  19. Schnipper JL,Magee M,Larsen K,Inzucchi SE,Maynard G.Society of Hospital Medicine Glycemic Control Task Force summary: practical recommendations for assessing the impact of glycemic control efforts.J Hosp Med.2008;3(5 Suppl):6675.
Issue
Journal of Hospital Medicine - 5(8)
Issue
Journal of Hospital Medicine - 5(8)
Page Number
438-445
Page Number
438-445
Article Type
Display Headline
Effects of an educational program and a standardized insulin order form on glycemic outcomes in non‐critically ill hospitalized patients
Display Headline
Effects of an educational program and a standardized insulin order form on glycemic outcomes in non‐critically ill hospitalized patients
Legacy Keywords
diabetes mellitus, hospitalization, hyperglycemia, inpatient, insulin
Legacy Keywords
diabetes mellitus, hospitalization, hyperglycemia, inpatient, insulin
Sections
Article Source

Copyright © 2010 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
3119 Taubman Center, 1500 E. Medical Center Drive, Box 0376, Ann Arbor, MI 48109‐0376
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

A Resting Place

Article Type
Changed
Fri, 09/14/2018 - 12:30
Display Headline
A Resting Place

When his hospital’s board of trustees was considering a palliative-care program seven years ago, hospitalist Stephen Bekanich, MD, wasn’t sure what to expect. What he did know was that his hospitalist group could provide the University of Utah Health System in Salt Lake City answers to staffing and financial issues surrounding the addition of a palliative-care service.

Dr. Bekanich

“They looked around and decided the hospitalist group would be the best place to house [the service], based on our experience with a range of medical management issues and the fact that we’re around 24 hours, seven days a week,” says Dr. Bekanich, who in 2006 became the first medical director of the palliative-care service at University of Utah Hospital.

The hospital board eventually selected palliative care as one of its annual projects, Dr. Bekanich says, not just because it was the right thing to do, but also because palliative care increasingly is used as a quality marker for hospitals. Dr. Bekanich says he took the assignment because it provides a nice buffer and change of pace from the stress of full-time HM service. Several colleagues joined him in rotating through palliative care coverage, although he continued to carry a pager most days and nights to support the physicians, advanced practice nurses, social worker, and chaplain with challenging cases.

After six months of operation, Dr. Bekanich went before his hospital board to discuss the program. He presented hospital data that showed the service had helped save the hospital $600,000, along with “thank-you letters” from grateful families and mentions in obituaries.

A few years ago, it was cutting-edge for hospitals to just have a palliative-care program, but now the focus is on quality and the qualifications of the palliative-care physicians and other professionals.

—Steven Pantilat, MD, SFHM, director, Univ, of California at San Francisco palliative care service, SHM past president

“A couple of months later, I realized that we needed another nurse practitioner to staff the growing caseload,” he explains. “I went to the chief medical officer and he said to me, ‘I don’t need to see the numbers. I know you’re doing a great job. Just tell me what you need.’ ”

Widely extolled for relieving the physical suffering and emotional distress of seriously ill patients, palliative care has seen rapid advancement in recent years, not only as a humanitarian impulse, but also as a legitimate and recognized medical subspecialty and career choice. Palliative care has its own board certification, fellowships, and training opportunities. For working hospitalists, this subspecialty can complement a career path and enhance job satisfaction. For HM groups, it represents diversification and an additional, albeit modest, income stream, as well as opportunities to improve the quality of hospital care.

“Palliative medicine is recognized by the American Board of Medical Specialties [ABMS] and nine of its medical specialty boards, which is very significant,” says Steven Pantilat, MD, SFHM, a hospitalist at the University of California at San Francisco (UCSF) Medical Center, medical director of UCSF’s palliative-care service. “Along with that come fellowships.”

Before You Build It, Do Your Homework

John Harney, COO at University of Colorado Hospital, moved west in 2008 after working at New York University Hospitals Center. The East Coast hospital had used a grant to establish a palliative-care program and witnessed immediate results.

“We truly believed it resulted in reductions in length of stay, as well as humanistic benefits,” Harney says. “When I came out to Colorado, I was pleasantly surprised at the breadth and depth of the programs here.”

Harney says HM is a logical place to advance palliative care to the next level, as most HM groups already possess an in-house presence and commitment to efficient throughput. Hospital administrators will be concerned with consistency, routines, and protocols, he says, as well as the palliative-care service’s commitment to quality improvement. Those same administrators appreciate the need for program and salary support, although he advises palliative-care advocates do their homework and develop a viable business plan.

“Hospital administrators will quickly figure out the math,” Harney says. “If you’re coming to speak to us, you need to have your numbers in order. You also need some monitoring in place.”

The initial conversation should include confirmation that HM group leaders have done their homework: Survey their own HM staff and discuss the idea with oncologists and other specialists. “It’s also helpful to have real champions in nursing and social work,” Harney says. “It’s never easy to get financial support for a new program, but if you have those ducks lined up, it goes better.”—LB

 

 

The Basics

Palliative care’s focus is managing patients’ symptoms, maximizing quality of life, and clarifying treatment goals—regardless of diagnosis or other treatments they might be receiving. It is not hospice care, which is defined by Medicare as treatment for patients with a terminal prognosis of six months or less (see “Hospice and Palliative: End-of-Life Care Siblings,” p. 21). Palliative care and hospice care utilize many of the same techniques, and are combined in the ABMS program for certifying subspecialist physicians.

The interdisciplinary consultation service, where a palliative care consultant rounds with a team that might include physicians, nurses, social workers, pharmacists, and chaplains, is the most common palliative-care model in the hospital setting, but other approaches include dedicated units and community-based programs.

The latest data from the American Hospital Association (AHA) and the Center to Advance Palliative Care (CAPC) count 1,486 operational palliative-care programs in U.S. acute-care hospitals, more than twice as many as a decade before.1 Currently, the demand for physicians certified in hospice and palliative medicine outstrips the supply, which poses challenges to those trying to hire as well as bona fide opportunities for qualified physicians hoping to pursue their dream jobs in the field, says Dr. Pantilat, a past president of SHM.

“A few years ago, it was cutting-edge for hospitals to just have a palliative care program,” Dr. Pantilat says, “but now the focus is on quality and the qualifications of the palliative care physicians and other professionals. Expectations for what palliative care will deliver will only go up.”

UCSF’s palliative care service “lives” within its HM division. Five of the six palliative care attending physicians are hospitalists. They divide weeklong assignments on the service into seven-day commitments at the hospital; each shift includes an on-call pager for night coverage.

A palliative-care shift can be just as emotionally demanding as an HM shift, although usually with fewer patients. One big difference: More time is needed for each palliative care consult, Dr. Pantilat says. A typical consult consists of an intense conversation with the patient and family to explore the patient’s prognosis, family values, and goals for treatment and pain relief.

Additionally, palliative care physicians routinely discuss the psychosocial and spiritual distress that the patient and family normally encounter.

Know When to Call for Help

Hospitalist involvement in palliative care varies by service, individual experience, and institution guidelines. Generally, though, it starts with an understanding of what the service provides and determining when is the right time to call a palliative-care consultant for help (see “Your Page Is Welcomed,” p. 22).

Hospitalists can obtain basic training and incorporate palliative-care principles and practices into the care of all hospitalized patients (see “Training Opportunities,” p. 22). If your hospital has a palliative-care service, hospitalists could join an advisory committee or provide backup coverage. If no such service exists, hospitalists could advocate with other physicians and hospital administrators to start one, Dr. Pantilat says.

Some hospitalists go deeper, developing subspecialty expertise and board certification in palliative medicine.

For HM groups, integration with a palliative-care service could mean taking on medical management of the service. If your group chooses to go this route, experts suggest you research how busy the service could be and gauge the interest of physicians in your group. Also check on the willingness of hospitalists in the group who are not interested in working on the palliative care service; they could help free up time for those who want to do it.

Dr. Bekanich
click for large version

What Every Hospitalist Should Know

 

 

The basic clinical skills needed to perform palliative medicine include:

  • Titrating opioid analgesics;
  • Using adjuvant pain medications;
  • Managing nonpain-related symptoms, including nausea, vomiting, constipation, dyspnea, seizures, and anorexia;
  • Managing delirium, anxiety, and depression;
  • Communicating sensitive information;
  • Working with cultural issues and differences; and
  • Bereavement support for families.

“Every hospitalist should know how to elicit a patient’s goals of care and incorporate them into routine treatment, be fluid and comfortable discussing advance-care planning, and possess basic skills in pain management,” says Jeanie Youngwerth, MD, hospitalist and director of the palliative-care service at the University of Colorado Denver. “Unfortunately, we’re not there yet as a field, given current residency training in internal medicine. Our center has a hospitalist residency training track, and those residents all get dedicated, palliative care experience.”

Hospice and Palliative: End-of-Life Siblings

Hospice care and palliative care share the same subspecialty medical board certification (in hospice and palliative medicine—HPM), similar approaches to relieving patients’ pain and suffering, and a philosophy emphasizing quality of life and personal empowerment for seriously ill patients and their families.

The main distinction between the two is that hospice care was recognized by Medicare as a covered benefit starting in 1983, allowing terminally ill patients to die in relative peace and comfort, without unwanted aggressive medical treatments, often in their own homes. Under Medicare coverage, hospice has grown into a $12 billion industry serving an estimated 1.4 million patients per year.

To qualify for hospice coverage under Medicare (along with Medicaid and many private health plans), a patient must be certified by two physicians as having a terminal illness with a prognosis of six months or less to live, assuming the disease follows its expected course.

Palliative care, as practiced in many hospitals, shares with hospice the commitment to supporting patients and their families emotionally and spiritually and helping them make treatment decisions that reflect their hopes and values. But palliative care does not require a terminal diagnosis or prognosis. Many palliative-care guidelines do not even mention the word “terminal.”

Medicare does not have a palliative care benefit, although consults provided by palliative care physicians and advanced practice nurses can be billed the same as for other specialists.—LB

Knowing when to refer a patient to a palliative-care specialist is another important skill, Dr. Youngwerth explains. The CARING criteria, developed by Dr. Youngwerth’s colleagues at UC Denver, are a simple set of prognostic markers that identify patients with limited life expectancy at the time of hospital admission. The CARING criteria are a set of prognostic criteria that incorporate cancer diagnosis, repeated hospital admissions, ICU stays with multi-organ failure, residence in a nursing home, and meeting non-cancer hospice guidelines developed by the National Hospice Organization, which collectively correlate with the need for a palliative-care consultation (see Table 1, above).2

A simpler way to initially assess a patient’s need for palliative care is to ask yourself: Would you be surprised if you found out this patient had died within a year? “If physicians don’t think the patient is going to be alive in a year, then they should incorporate palliative care into the care plan,” Dr. Youngwerth says. “The next question is: Should I do it myself, or refer for a palliative-care consultation?”

Dr. Bekanich, who starting this month will head a new palliative care program at the University of Miami that features a 10-bed inpatient unit, encourages hospitalists to avoid focusing only on terminally ill patients when considering a palliative consult. Any seriously ill patient with unmet needs could benefit from a referral, he says.

“Lots of hospitalists are good at controlling nausea and vomiting, but if the symptoms are refractory or have uncommon presentations, I would like to get on board as the palliative care consultant,” Dr. Bekanich says. “I have also tried to emphasize to my group the importance of timely family meetings.

 

 

“If they don’t have the time or the skills, or if they expect a difficult meeting, for example, due to religious or cultural differences, send these patients our way. And when there are ethical issues that need to be addressed, or a particular need for educating patients and families about the disease process and what to expect, I like consultations like that.”

Bad Business or New Revenue Stream?

The traditional business model for palliative-care services has focused on the potential contributions to the hospital’s bottom line through reduced length of stay and cost avoidance for a group of patients who can be among the hospital’s most challenging and expensive. Palliative care saves time and money by working with patients and their families to clarify their values and treatment preferences, which routinely differ from standard treatment modes.

A recent multisite study of palliative care by Morrison et al found that the use of palliative care services saved from $1,700 to $4,900 per admission in direct costs, compared with similar patients who did not receive palliative care.3 The savings were realized primarily through reduced laboratory, pharmacy, and ICU costs.

Cost avoidance, combined with palliative care’s contributions to quality and patient satisfaction, is essential to the field’s growth. Even though physician consultation visits are billable, a palliative-care service rarely covers its staffing costs solely with billing revenue. A service requires nonbillable support from administration and midlevel providers, including nurses and social workers.

“Integrating palliative care into the work of hospitalists is a great idea,” says Jean Kutner, MD, head of the division of general internal medicine at the University of Colorado Denver. However, there are important issues related to scheduling, availability, and commitment that need to be explored before a group launches a new service. “I’d want to have discussions about how the palliative-care business model fits with our hospital medicine model and an agreement with the hospital on goals and metrics,” she says.

Your Page Is Welcomed

When faced with a potential palliative-care case, never hesitate to call, says Dr. Bekanich. Palliative specialists and teams expect to receive pages for such matters, and actually welcome them. The following are basic guidelines for when residents, early-career hospitalists and other providers should call the palliative-care service:

  • Patient has a serious medical condition that you are unfamiliar or uncomfortable with;
  • Consults for pain, dyspnea, or other distressing symptoms AND the patient is medically complex (tenuous cardiopulmonary status, chronic renal or liver disease, etc.);
  • Patients who have symptoms resistant to standard care;
  • Patients, family, or the referring team is unsatisfied with the consult;
  • Red flags are identified for patient safety (including disposition);
  • Ethical issues;
  • The case involves risk management;
  • Emotional involvement is becoming a strain;
  • Patient has a recent history of substance abuse; or
  • Patient volume increases and other patients are not receiving normal quality of care.

Hospitalists Fill a Need

Whether a full-fledged palliative-care service fits your group’s dynamic or not, hospitalists as a whole should be competent in basic palliative care. Community and rural hospitals need HM to bridge this gap and deliver quality care to seriously ill patients.

Dr. Harris

“I started at a community hospital, Eden Medical Center in Castro Valley, California. I had a personal interest in palliative care and realized there’s a tremendous need for it in community hospitals,” says Heather A. Harris, MD, a hospitalist at San Francisco General Hospital who previously worked with Dr. Pantilat’s palliative care service at UCSF. “We deal with end-of-life issues on a regular basis—whether recognized or not—based on our caseloads and requests for consultations.

“I got a little perspective about palliative care while a resident at UCSF. But as I’ve gotten further into this, I have come to realize that there is an actual skill set that needs to be learned to do it properly.”

 

 

Dr. Harris says there is a big difference between physicians helping patients with end-of-life issues the best they can and being part of a “dedicated, interdisciplinary team.”

“Palliative care is a wonderful opportunity for hospitalists,” she says. “It’s already part of your practice. Why not do it in a more organized fashion?” TH

Larry Beresford is a freelance medical writer based in Oakland, Calif.

References

  1. Palliative care programs continue rapid growth in U.S. hospitals. Center to Advance Palliative Care website. Available at: www.capc.org/news-and-events/releases/04-05-10. Accessed July 15, 2010.
  2. Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292.
  3. Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with US hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

Training Opportunities

Resources available to hospitalists interested in integrating palliative care into their programs:

The American Academy of Hospice and Palliative Medicine (AAHPM), a 4,000-member professional society based in Glenview, Ill., offers annual educational meetings (the next is Feb. 16-19 in Vancouver), a self-study course, summaries of medical literature, and a Clinical Scholars Program with 40 hours of mentorship at one of eight AAHPM training sites. The website (www.aahpm.org) is home to the Hospice and Palliative Medicine (HPM) Fellowship Program Directory, which lists 74 active programs that offer 181 fellowships. The fellowships generally are for one year and include 27 research slots.

Starting in 2014, physicians who want to become board-certified in HPM must complete an American College of Graduate Medical Education-accredited HPM fellowship (www.aahpm.org/certification/abms.html). Sitting for the boards based on work experience is still an option for the 2010 and 2012 exams. Palliative care leaders encourage interested working hospitalists to take advantage of this window of opportunity and join the more than 2,000 physicians who already are HPM certified.

The Center to Advance Palliative Care at Mount Sinai School of Medicine in New York City offers a variety of resources focused on palliative-care program development, including nine regional palliative care leadership centers, annual national training sessions, and financial models. For more info, visit www.capc.org.

Twice a year, Harvard Medical School offers the Program in Palliative Care Education and Practice (www.hms.harvard.edu/cdi/pallcare/pcep.htm) for physician and nurse educators who want to become experts in comprehensive, interdisciplinary palliative care.—LB

What Can Palliative Care Do to Hospitals’ Mortality Rates?

By Larry Beresford

Severity-adjusted hospital mortality rates are the cornerstone of a proliferating number of public and private hospital quality initiatives—and thus a quality focus for hospitalists. Yet some hospital-connected deaths are unavoidable, predictable, and even appropriate when palliative support focused on maximizing comfort and quality of life replaces medical efforts to stave off death.

Where hospice and palliative care fit in hospital mortality rates, how they are defined and counted, and how predictable deaths are either included or excluded from hospitals’ risk-adjusted mortality tallies vary between the reporting programs, according to J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University. He presented on mortality rates at the National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C. in April 2009.

“How hospital mortality rates are determined can be quite complex,” with varied data sources and various methods of adjusting for severity and balancing mortality with other quality metrics, says J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University who presented on mortality rates at the 2009 National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C.

Typically, the risk-adjusted mortality rate is for selected diagnoses but counts deaths from all causes, either during the index hospitalization or within 30 days of that admission, Cassel says. He reviewed three quality programs that use mortality data: the Centers for Medicare and Medicaid Services’ Hospital Compare, which publicly reports data on patient satisfaction and hospital processes and outcomes, including mortality; U.S. News & World Report’s “Best Hospitals”; and HealthGrades (www.healthgrades.com), a Golden, Colo.-based company that ranks hospitals and other health providers. He started studying the subject because of concerns that an acute palliative-care unit at VCU might be hurting the medical center’s overall mortality scores; it turned out not to be the case.

An ICD-9 (International Classifications of Disease) billing code, V66.7 for “palliative care encounter,” can flag the involvement of palliative- care consultants on a hospital case, although this code often goes unused and needs to be among the top nine listed diagnoses in order to turn up in most quality calculations. Palliative care consultants can help promote the use and higher positioning of this code in hospital billing, along with more complete documentation of co-morbidities and symptoms. It is also possible that involving hospice and palliative care teams with seriously ill patients earlier in their disease progression could help manage their care in community settings, avoiding hospitalizations when death is likely in the next few months.

Some hospitals might choose to refer patients known to be close to death to contracted hospice programs—although this should be based on the best interests of the patient, not on improving the facility’s mortality rates. Cassel’s advice for hospice and palliative care advocates trying to stake their claim in hospital quality measurement: provide the best possible care to patients and families, but meanwhile, know which quality measurement systems the hospital’s leadership follows, and what these look for.

Issue
The Hospitalist - 2010(08)
Publications
Sections

When his hospital’s board of trustees was considering a palliative-care program seven years ago, hospitalist Stephen Bekanich, MD, wasn’t sure what to expect. What he did know was that his hospitalist group could provide the University of Utah Health System in Salt Lake City answers to staffing and financial issues surrounding the addition of a palliative-care service.

Dr. Bekanich

“They looked around and decided the hospitalist group would be the best place to house [the service], based on our experience with a range of medical management issues and the fact that we’re around 24 hours, seven days a week,” says Dr. Bekanich, who in 2006 became the first medical director of the palliative-care service at University of Utah Hospital.

The hospital board eventually selected palliative care as one of its annual projects, Dr. Bekanich says, not just because it was the right thing to do, but also because palliative care increasingly is used as a quality marker for hospitals. Dr. Bekanich says he took the assignment because it provides a nice buffer and change of pace from the stress of full-time HM service. Several colleagues joined him in rotating through palliative care coverage, although he continued to carry a pager most days and nights to support the physicians, advanced practice nurses, social worker, and chaplain with challenging cases.

After six months of operation, Dr. Bekanich went before his hospital board to discuss the program. He presented hospital data that showed the service had helped save the hospital $600,000, along with “thank-you letters” from grateful families and mentions in obituaries.

A few years ago, it was cutting-edge for hospitals to just have a palliative-care program, but now the focus is on quality and the qualifications of the palliative-care physicians and other professionals.

—Steven Pantilat, MD, SFHM, director, Univ, of California at San Francisco palliative care service, SHM past president

“A couple of months later, I realized that we needed another nurse practitioner to staff the growing caseload,” he explains. “I went to the chief medical officer and he said to me, ‘I don’t need to see the numbers. I know you’re doing a great job. Just tell me what you need.’ ”

Widely extolled for relieving the physical suffering and emotional distress of seriously ill patients, palliative care has seen rapid advancement in recent years, not only as a humanitarian impulse, but also as a legitimate and recognized medical subspecialty and career choice. Palliative care has its own board certification, fellowships, and training opportunities. For working hospitalists, this subspecialty can complement a career path and enhance job satisfaction. For HM groups, it represents diversification and an additional, albeit modest, income stream, as well as opportunities to improve the quality of hospital care.

“Palliative medicine is recognized by the American Board of Medical Specialties [ABMS] and nine of its medical specialty boards, which is very significant,” says Steven Pantilat, MD, SFHM, a hospitalist at the University of California at San Francisco (UCSF) Medical Center, medical director of UCSF’s palliative-care service. “Along with that come fellowships.”

Before You Build It, Do Your Homework

John Harney, COO at University of Colorado Hospital, moved west in 2008 after working at New York University Hospitals Center. The East Coast hospital had used a grant to establish a palliative-care program and witnessed immediate results.

“We truly believed it resulted in reductions in length of stay, as well as humanistic benefits,” Harney says. “When I came out to Colorado, I was pleasantly surprised at the breadth and depth of the programs here.”

Harney says HM is a logical place to advance palliative care to the next level, as most HM groups already possess an in-house presence and commitment to efficient throughput. Hospital administrators will be concerned with consistency, routines, and protocols, he says, as well as the palliative-care service’s commitment to quality improvement. Those same administrators appreciate the need for program and salary support, although he advises palliative-care advocates do their homework and develop a viable business plan.

“Hospital administrators will quickly figure out the math,” Harney says. “If you’re coming to speak to us, you need to have your numbers in order. You also need some monitoring in place.”

The initial conversation should include confirmation that HM group leaders have done their homework: Survey their own HM staff and discuss the idea with oncologists and other specialists. “It’s also helpful to have real champions in nursing and social work,” Harney says. “It’s never easy to get financial support for a new program, but if you have those ducks lined up, it goes better.”—LB

 

 

The Basics

Palliative care’s focus is managing patients’ symptoms, maximizing quality of life, and clarifying treatment goals—regardless of diagnosis or other treatments they might be receiving. It is not hospice care, which is defined by Medicare as treatment for patients with a terminal prognosis of six months or less (see “Hospice and Palliative: End-of-Life Care Siblings,” p. 21). Palliative care and hospice care utilize many of the same techniques, and are combined in the ABMS program for certifying subspecialist physicians.

The interdisciplinary consultation service, where a palliative care consultant rounds with a team that might include physicians, nurses, social workers, pharmacists, and chaplains, is the most common palliative-care model in the hospital setting, but other approaches include dedicated units and community-based programs.

The latest data from the American Hospital Association (AHA) and the Center to Advance Palliative Care (CAPC) count 1,486 operational palliative-care programs in U.S. acute-care hospitals, more than twice as many as a decade before.1 Currently, the demand for physicians certified in hospice and palliative medicine outstrips the supply, which poses challenges to those trying to hire as well as bona fide opportunities for qualified physicians hoping to pursue their dream jobs in the field, says Dr. Pantilat, a past president of SHM.

“A few years ago, it was cutting-edge for hospitals to just have a palliative care program,” Dr. Pantilat says, “but now the focus is on quality and the qualifications of the palliative care physicians and other professionals. Expectations for what palliative care will deliver will only go up.”

UCSF’s palliative care service “lives” within its HM division. Five of the six palliative care attending physicians are hospitalists. They divide weeklong assignments on the service into seven-day commitments at the hospital; each shift includes an on-call pager for night coverage.

A palliative-care shift can be just as emotionally demanding as an HM shift, although usually with fewer patients. One big difference: More time is needed for each palliative care consult, Dr. Pantilat says. A typical consult consists of an intense conversation with the patient and family to explore the patient’s prognosis, family values, and goals for treatment and pain relief.

Additionally, palliative care physicians routinely discuss the psychosocial and spiritual distress that the patient and family normally encounter.

Know When to Call for Help

Hospitalist involvement in palliative care varies by service, individual experience, and institution guidelines. Generally, though, it starts with an understanding of what the service provides and determining when is the right time to call a palliative-care consultant for help (see “Your Page Is Welcomed,” p. 22).

Hospitalists can obtain basic training and incorporate palliative-care principles and practices into the care of all hospitalized patients (see “Training Opportunities,” p. 22). If your hospital has a palliative-care service, hospitalists could join an advisory committee or provide backup coverage. If no such service exists, hospitalists could advocate with other physicians and hospital administrators to start one, Dr. Pantilat says.

Some hospitalists go deeper, developing subspecialty expertise and board certification in palliative medicine.

For HM groups, integration with a palliative-care service could mean taking on medical management of the service. If your group chooses to go this route, experts suggest you research how busy the service could be and gauge the interest of physicians in your group. Also check on the willingness of hospitalists in the group who are not interested in working on the palliative care service; they could help free up time for those who want to do it.

Dr. Bekanich
click for large version

What Every Hospitalist Should Know

 

 

The basic clinical skills needed to perform palliative medicine include:

  • Titrating opioid analgesics;
  • Using adjuvant pain medications;
  • Managing nonpain-related symptoms, including nausea, vomiting, constipation, dyspnea, seizures, and anorexia;
  • Managing delirium, anxiety, and depression;
  • Communicating sensitive information;
  • Working with cultural issues and differences; and
  • Bereavement support for families.

“Every hospitalist should know how to elicit a patient’s goals of care and incorporate them into routine treatment, be fluid and comfortable discussing advance-care planning, and possess basic skills in pain management,” says Jeanie Youngwerth, MD, hospitalist and director of the palliative-care service at the University of Colorado Denver. “Unfortunately, we’re not there yet as a field, given current residency training in internal medicine. Our center has a hospitalist residency training track, and those residents all get dedicated, palliative care experience.”

Hospice and Palliative: End-of-Life Siblings

Hospice care and palliative care share the same subspecialty medical board certification (in hospice and palliative medicine—HPM), similar approaches to relieving patients’ pain and suffering, and a philosophy emphasizing quality of life and personal empowerment for seriously ill patients and their families.

The main distinction between the two is that hospice care was recognized by Medicare as a covered benefit starting in 1983, allowing terminally ill patients to die in relative peace and comfort, without unwanted aggressive medical treatments, often in their own homes. Under Medicare coverage, hospice has grown into a $12 billion industry serving an estimated 1.4 million patients per year.

To qualify for hospice coverage under Medicare (along with Medicaid and many private health plans), a patient must be certified by two physicians as having a terminal illness with a prognosis of six months or less to live, assuming the disease follows its expected course.

Palliative care, as practiced in many hospitals, shares with hospice the commitment to supporting patients and their families emotionally and spiritually and helping them make treatment decisions that reflect their hopes and values. But palliative care does not require a terminal diagnosis or prognosis. Many palliative-care guidelines do not even mention the word “terminal.”

Medicare does not have a palliative care benefit, although consults provided by palliative care physicians and advanced practice nurses can be billed the same as for other specialists.—LB

Knowing when to refer a patient to a palliative-care specialist is another important skill, Dr. Youngwerth explains. The CARING criteria, developed by Dr. Youngwerth’s colleagues at UC Denver, are a simple set of prognostic markers that identify patients with limited life expectancy at the time of hospital admission. The CARING criteria are a set of prognostic criteria that incorporate cancer diagnosis, repeated hospital admissions, ICU stays with multi-organ failure, residence in a nursing home, and meeting non-cancer hospice guidelines developed by the National Hospice Organization, which collectively correlate with the need for a palliative-care consultation (see Table 1, above).2

A simpler way to initially assess a patient’s need for palliative care is to ask yourself: Would you be surprised if you found out this patient had died within a year? “If physicians don’t think the patient is going to be alive in a year, then they should incorporate palliative care into the care plan,” Dr. Youngwerth says. “The next question is: Should I do it myself, or refer for a palliative-care consultation?”

Dr. Bekanich, who starting this month will head a new palliative care program at the University of Miami that features a 10-bed inpatient unit, encourages hospitalists to avoid focusing only on terminally ill patients when considering a palliative consult. Any seriously ill patient with unmet needs could benefit from a referral, he says.

“Lots of hospitalists are good at controlling nausea and vomiting, but if the symptoms are refractory or have uncommon presentations, I would like to get on board as the palliative care consultant,” Dr. Bekanich says. “I have also tried to emphasize to my group the importance of timely family meetings.

 

 

“If they don’t have the time or the skills, or if they expect a difficult meeting, for example, due to religious or cultural differences, send these patients our way. And when there are ethical issues that need to be addressed, or a particular need for educating patients and families about the disease process and what to expect, I like consultations like that.”

Bad Business or New Revenue Stream?

The traditional business model for palliative-care services has focused on the potential contributions to the hospital’s bottom line through reduced length of stay and cost avoidance for a group of patients who can be among the hospital’s most challenging and expensive. Palliative care saves time and money by working with patients and their families to clarify their values and treatment preferences, which routinely differ from standard treatment modes.

A recent multisite study of palliative care by Morrison et al found that the use of palliative care services saved from $1,700 to $4,900 per admission in direct costs, compared with similar patients who did not receive palliative care.3 The savings were realized primarily through reduced laboratory, pharmacy, and ICU costs.

Cost avoidance, combined with palliative care’s contributions to quality and patient satisfaction, is essential to the field’s growth. Even though physician consultation visits are billable, a palliative-care service rarely covers its staffing costs solely with billing revenue. A service requires nonbillable support from administration and midlevel providers, including nurses and social workers.

“Integrating palliative care into the work of hospitalists is a great idea,” says Jean Kutner, MD, head of the division of general internal medicine at the University of Colorado Denver. However, there are important issues related to scheduling, availability, and commitment that need to be explored before a group launches a new service. “I’d want to have discussions about how the palliative-care business model fits with our hospital medicine model and an agreement with the hospital on goals and metrics,” she says.

Your Page Is Welcomed

When faced with a potential palliative-care case, never hesitate to call, says Dr. Bekanich. Palliative specialists and teams expect to receive pages for such matters, and actually welcome them. The following are basic guidelines for when residents, early-career hospitalists and other providers should call the palliative-care service:

  • Patient has a serious medical condition that you are unfamiliar or uncomfortable with;
  • Consults for pain, dyspnea, or other distressing symptoms AND the patient is medically complex (tenuous cardiopulmonary status, chronic renal or liver disease, etc.);
  • Patients who have symptoms resistant to standard care;
  • Patients, family, or the referring team is unsatisfied with the consult;
  • Red flags are identified for patient safety (including disposition);
  • Ethical issues;
  • The case involves risk management;
  • Emotional involvement is becoming a strain;
  • Patient has a recent history of substance abuse; or
  • Patient volume increases and other patients are not receiving normal quality of care.

Hospitalists Fill a Need

Whether a full-fledged palliative-care service fits your group’s dynamic or not, hospitalists as a whole should be competent in basic palliative care. Community and rural hospitals need HM to bridge this gap and deliver quality care to seriously ill patients.

Dr. Harris

“I started at a community hospital, Eden Medical Center in Castro Valley, California. I had a personal interest in palliative care and realized there’s a tremendous need for it in community hospitals,” says Heather A. Harris, MD, a hospitalist at San Francisco General Hospital who previously worked with Dr. Pantilat’s palliative care service at UCSF. “We deal with end-of-life issues on a regular basis—whether recognized or not—based on our caseloads and requests for consultations.

“I got a little perspective about palliative care while a resident at UCSF. But as I’ve gotten further into this, I have come to realize that there is an actual skill set that needs to be learned to do it properly.”

 

 

Dr. Harris says there is a big difference between physicians helping patients with end-of-life issues the best they can and being part of a “dedicated, interdisciplinary team.”

“Palliative care is a wonderful opportunity for hospitalists,” she says. “It’s already part of your practice. Why not do it in a more organized fashion?” TH

Larry Beresford is a freelance medical writer based in Oakland, Calif.

References

  1. Palliative care programs continue rapid growth in U.S. hospitals. Center to Advance Palliative Care website. Available at: www.capc.org/news-and-events/releases/04-05-10. Accessed July 15, 2010.
  2. Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292.
  3. Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with US hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

Training Opportunities

Resources available to hospitalists interested in integrating palliative care into their programs:

The American Academy of Hospice and Palliative Medicine (AAHPM), a 4,000-member professional society based in Glenview, Ill., offers annual educational meetings (the next is Feb. 16-19 in Vancouver), a self-study course, summaries of medical literature, and a Clinical Scholars Program with 40 hours of mentorship at one of eight AAHPM training sites. The website (www.aahpm.org) is home to the Hospice and Palliative Medicine (HPM) Fellowship Program Directory, which lists 74 active programs that offer 181 fellowships. The fellowships generally are for one year and include 27 research slots.

Starting in 2014, physicians who want to become board-certified in HPM must complete an American College of Graduate Medical Education-accredited HPM fellowship (www.aahpm.org/certification/abms.html). Sitting for the boards based on work experience is still an option for the 2010 and 2012 exams. Palliative care leaders encourage interested working hospitalists to take advantage of this window of opportunity and join the more than 2,000 physicians who already are HPM certified.

The Center to Advance Palliative Care at Mount Sinai School of Medicine in New York City offers a variety of resources focused on palliative-care program development, including nine regional palliative care leadership centers, annual national training sessions, and financial models. For more info, visit www.capc.org.

Twice a year, Harvard Medical School offers the Program in Palliative Care Education and Practice (www.hms.harvard.edu/cdi/pallcare/pcep.htm) for physician and nurse educators who want to become experts in comprehensive, interdisciplinary palliative care.—LB

What Can Palliative Care Do to Hospitals’ Mortality Rates?

By Larry Beresford

Severity-adjusted hospital mortality rates are the cornerstone of a proliferating number of public and private hospital quality initiatives—and thus a quality focus for hospitalists. Yet some hospital-connected deaths are unavoidable, predictable, and even appropriate when palliative support focused on maximizing comfort and quality of life replaces medical efforts to stave off death.

Where hospice and palliative care fit in hospital mortality rates, how they are defined and counted, and how predictable deaths are either included or excluded from hospitals’ risk-adjusted mortality tallies vary between the reporting programs, according to J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University. He presented on mortality rates at the National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C. in April 2009.

“How hospital mortality rates are determined can be quite complex,” with varied data sources and various methods of adjusting for severity and balancing mortality with other quality metrics, says J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University who presented on mortality rates at the 2009 National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C.

Typically, the risk-adjusted mortality rate is for selected diagnoses but counts deaths from all causes, either during the index hospitalization or within 30 days of that admission, Cassel says. He reviewed three quality programs that use mortality data: the Centers for Medicare and Medicaid Services’ Hospital Compare, which publicly reports data on patient satisfaction and hospital processes and outcomes, including mortality; U.S. News & World Report’s “Best Hospitals”; and HealthGrades (www.healthgrades.com), a Golden, Colo.-based company that ranks hospitals and other health providers. He started studying the subject because of concerns that an acute palliative-care unit at VCU might be hurting the medical center’s overall mortality scores; it turned out not to be the case.

An ICD-9 (International Classifications of Disease) billing code, V66.7 for “palliative care encounter,” can flag the involvement of palliative- care consultants on a hospital case, although this code often goes unused and needs to be among the top nine listed diagnoses in order to turn up in most quality calculations. Palliative care consultants can help promote the use and higher positioning of this code in hospital billing, along with more complete documentation of co-morbidities and symptoms. It is also possible that involving hospice and palliative care teams with seriously ill patients earlier in their disease progression could help manage their care in community settings, avoiding hospitalizations when death is likely in the next few months.

Some hospitals might choose to refer patients known to be close to death to contracted hospice programs—although this should be based on the best interests of the patient, not on improving the facility’s mortality rates. Cassel’s advice for hospice and palliative care advocates trying to stake their claim in hospital quality measurement: provide the best possible care to patients and families, but meanwhile, know which quality measurement systems the hospital’s leadership follows, and what these look for.

When his hospital’s board of trustees was considering a palliative-care program seven years ago, hospitalist Stephen Bekanich, MD, wasn’t sure what to expect. What he did know was that his hospitalist group could provide the University of Utah Health System in Salt Lake City answers to staffing and financial issues surrounding the addition of a palliative-care service.

Dr. Bekanich

“They looked around and decided the hospitalist group would be the best place to house [the service], based on our experience with a range of medical management issues and the fact that we’re around 24 hours, seven days a week,” says Dr. Bekanich, who in 2006 became the first medical director of the palliative-care service at University of Utah Hospital.

The hospital board eventually selected palliative care as one of its annual projects, Dr. Bekanich says, not just because it was the right thing to do, but also because palliative care increasingly is used as a quality marker for hospitals. Dr. Bekanich says he took the assignment because it provides a nice buffer and change of pace from the stress of full-time HM service. Several colleagues joined him in rotating through palliative care coverage, although he continued to carry a pager most days and nights to support the physicians, advanced practice nurses, social worker, and chaplain with challenging cases.

After six months of operation, Dr. Bekanich went before his hospital board to discuss the program. He presented hospital data that showed the service had helped save the hospital $600,000, along with “thank-you letters” from grateful families and mentions in obituaries.

A few years ago, it was cutting-edge for hospitals to just have a palliative-care program, but now the focus is on quality and the qualifications of the palliative-care physicians and other professionals.

—Steven Pantilat, MD, SFHM, director, Univ, of California at San Francisco palliative care service, SHM past president

“A couple of months later, I realized that we needed another nurse practitioner to staff the growing caseload,” he explains. “I went to the chief medical officer and he said to me, ‘I don’t need to see the numbers. I know you’re doing a great job. Just tell me what you need.’ ”

Widely extolled for relieving the physical suffering and emotional distress of seriously ill patients, palliative care has seen rapid advancement in recent years, not only as a humanitarian impulse, but also as a legitimate and recognized medical subspecialty and career choice. Palliative care has its own board certification, fellowships, and training opportunities. For working hospitalists, this subspecialty can complement a career path and enhance job satisfaction. For HM groups, it represents diversification and an additional, albeit modest, income stream, as well as opportunities to improve the quality of hospital care.

“Palliative medicine is recognized by the American Board of Medical Specialties [ABMS] and nine of its medical specialty boards, which is very significant,” says Steven Pantilat, MD, SFHM, a hospitalist at the University of California at San Francisco (UCSF) Medical Center, medical director of UCSF’s palliative-care service. “Along with that come fellowships.”

Before You Build It, Do Your Homework

John Harney, COO at University of Colorado Hospital, moved west in 2008 after working at New York University Hospitals Center. The East Coast hospital had used a grant to establish a palliative-care program and witnessed immediate results.

“We truly believed it resulted in reductions in length of stay, as well as humanistic benefits,” Harney says. “When I came out to Colorado, I was pleasantly surprised at the breadth and depth of the programs here.”

Harney says HM is a logical place to advance palliative care to the next level, as most HM groups already possess an in-house presence and commitment to efficient throughput. Hospital administrators will be concerned with consistency, routines, and protocols, he says, as well as the palliative-care service’s commitment to quality improvement. Those same administrators appreciate the need for program and salary support, although he advises palliative-care advocates do their homework and develop a viable business plan.

“Hospital administrators will quickly figure out the math,” Harney says. “If you’re coming to speak to us, you need to have your numbers in order. You also need some monitoring in place.”

The initial conversation should include confirmation that HM group leaders have done their homework: Survey their own HM staff and discuss the idea with oncologists and other specialists. “It’s also helpful to have real champions in nursing and social work,” Harney says. “It’s never easy to get financial support for a new program, but if you have those ducks lined up, it goes better.”—LB

 

 

The Basics

Palliative care’s focus is managing patients’ symptoms, maximizing quality of life, and clarifying treatment goals—regardless of diagnosis or other treatments they might be receiving. It is not hospice care, which is defined by Medicare as treatment for patients with a terminal prognosis of six months or less (see “Hospice and Palliative: End-of-Life Care Siblings,” p. 21). Palliative care and hospice care utilize many of the same techniques, and are combined in the ABMS program for certifying subspecialist physicians.

The interdisciplinary consultation service, where a palliative care consultant rounds with a team that might include physicians, nurses, social workers, pharmacists, and chaplains, is the most common palliative-care model in the hospital setting, but other approaches include dedicated units and community-based programs.

The latest data from the American Hospital Association (AHA) and the Center to Advance Palliative Care (CAPC) count 1,486 operational palliative-care programs in U.S. acute-care hospitals, more than twice as many as a decade before.1 Currently, the demand for physicians certified in hospice and palliative medicine outstrips the supply, which poses challenges to those trying to hire as well as bona fide opportunities for qualified physicians hoping to pursue their dream jobs in the field, says Dr. Pantilat, a past president of SHM.

“A few years ago, it was cutting-edge for hospitals to just have a palliative care program,” Dr. Pantilat says, “but now the focus is on quality and the qualifications of the palliative care physicians and other professionals. Expectations for what palliative care will deliver will only go up.”

UCSF’s palliative care service “lives” within its HM division. Five of the six palliative care attending physicians are hospitalists. They divide weeklong assignments on the service into seven-day commitments at the hospital; each shift includes an on-call pager for night coverage.

A palliative-care shift can be just as emotionally demanding as an HM shift, although usually with fewer patients. One big difference: More time is needed for each palliative care consult, Dr. Pantilat says. A typical consult consists of an intense conversation with the patient and family to explore the patient’s prognosis, family values, and goals for treatment and pain relief.

Additionally, palliative care physicians routinely discuss the psychosocial and spiritual distress that the patient and family normally encounter.

Know When to Call for Help

Hospitalist involvement in palliative care varies by service, individual experience, and institution guidelines. Generally, though, it starts with an understanding of what the service provides and determining when is the right time to call a palliative-care consultant for help (see “Your Page Is Welcomed,” p. 22).

Hospitalists can obtain basic training and incorporate palliative-care principles and practices into the care of all hospitalized patients (see “Training Opportunities,” p. 22). If your hospital has a palliative-care service, hospitalists could join an advisory committee or provide backup coverage. If no such service exists, hospitalists could advocate with other physicians and hospital administrators to start one, Dr. Pantilat says.

Some hospitalists go deeper, developing subspecialty expertise and board certification in palliative medicine.

For HM groups, integration with a palliative-care service could mean taking on medical management of the service. If your group chooses to go this route, experts suggest you research how busy the service could be and gauge the interest of physicians in your group. Also check on the willingness of hospitalists in the group who are not interested in working on the palliative care service; they could help free up time for those who want to do it.

Dr. Bekanich
click for large version

What Every Hospitalist Should Know

 

 

The basic clinical skills needed to perform palliative medicine include:

  • Titrating opioid analgesics;
  • Using adjuvant pain medications;
  • Managing nonpain-related symptoms, including nausea, vomiting, constipation, dyspnea, seizures, and anorexia;
  • Managing delirium, anxiety, and depression;
  • Communicating sensitive information;
  • Working with cultural issues and differences; and
  • Bereavement support for families.

“Every hospitalist should know how to elicit a patient’s goals of care and incorporate them into routine treatment, be fluid and comfortable discussing advance-care planning, and possess basic skills in pain management,” says Jeanie Youngwerth, MD, hospitalist and director of the palliative-care service at the University of Colorado Denver. “Unfortunately, we’re not there yet as a field, given current residency training in internal medicine. Our center has a hospitalist residency training track, and those residents all get dedicated, palliative care experience.”

Hospice and Palliative: End-of-Life Siblings

Hospice care and palliative care share the same subspecialty medical board certification (in hospice and palliative medicine—HPM), similar approaches to relieving patients’ pain and suffering, and a philosophy emphasizing quality of life and personal empowerment for seriously ill patients and their families.

The main distinction between the two is that hospice care was recognized by Medicare as a covered benefit starting in 1983, allowing terminally ill patients to die in relative peace and comfort, without unwanted aggressive medical treatments, often in their own homes. Under Medicare coverage, hospice has grown into a $12 billion industry serving an estimated 1.4 million patients per year.

To qualify for hospice coverage under Medicare (along with Medicaid and many private health plans), a patient must be certified by two physicians as having a terminal illness with a prognosis of six months or less to live, assuming the disease follows its expected course.

Palliative care, as practiced in many hospitals, shares with hospice the commitment to supporting patients and their families emotionally and spiritually and helping them make treatment decisions that reflect their hopes and values. But palliative care does not require a terminal diagnosis or prognosis. Many palliative-care guidelines do not even mention the word “terminal.”

Medicare does not have a palliative care benefit, although consults provided by palliative care physicians and advanced practice nurses can be billed the same as for other specialists.—LB

Knowing when to refer a patient to a palliative-care specialist is another important skill, Dr. Youngwerth explains. The CARING criteria, developed by Dr. Youngwerth’s colleagues at UC Denver, are a simple set of prognostic markers that identify patients with limited life expectancy at the time of hospital admission. The CARING criteria are a set of prognostic criteria that incorporate cancer diagnosis, repeated hospital admissions, ICU stays with multi-organ failure, residence in a nursing home, and meeting non-cancer hospice guidelines developed by the National Hospice Organization, which collectively correlate with the need for a palliative-care consultation (see Table 1, above).2

A simpler way to initially assess a patient’s need for palliative care is to ask yourself: Would you be surprised if you found out this patient had died within a year? “If physicians don’t think the patient is going to be alive in a year, then they should incorporate palliative care into the care plan,” Dr. Youngwerth says. “The next question is: Should I do it myself, or refer for a palliative-care consultation?”

Dr. Bekanich, who starting this month will head a new palliative care program at the University of Miami that features a 10-bed inpatient unit, encourages hospitalists to avoid focusing only on terminally ill patients when considering a palliative consult. Any seriously ill patient with unmet needs could benefit from a referral, he says.

“Lots of hospitalists are good at controlling nausea and vomiting, but if the symptoms are refractory or have uncommon presentations, I would like to get on board as the palliative care consultant,” Dr. Bekanich says. “I have also tried to emphasize to my group the importance of timely family meetings.

 

 

“If they don’t have the time or the skills, or if they expect a difficult meeting, for example, due to religious or cultural differences, send these patients our way. And when there are ethical issues that need to be addressed, or a particular need for educating patients and families about the disease process and what to expect, I like consultations like that.”

Bad Business or New Revenue Stream?

The traditional business model for palliative-care services has focused on the potential contributions to the hospital’s bottom line through reduced length of stay and cost avoidance for a group of patients who can be among the hospital’s most challenging and expensive. Palliative care saves time and money by working with patients and their families to clarify their values and treatment preferences, which routinely differ from standard treatment modes.

A recent multisite study of palliative care by Morrison et al found that the use of palliative care services saved from $1,700 to $4,900 per admission in direct costs, compared with similar patients who did not receive palliative care.3 The savings were realized primarily through reduced laboratory, pharmacy, and ICU costs.

Cost avoidance, combined with palliative care’s contributions to quality and patient satisfaction, is essential to the field’s growth. Even though physician consultation visits are billable, a palliative-care service rarely covers its staffing costs solely with billing revenue. A service requires nonbillable support from administration and midlevel providers, including nurses and social workers.

“Integrating palliative care into the work of hospitalists is a great idea,” says Jean Kutner, MD, head of the division of general internal medicine at the University of Colorado Denver. However, there are important issues related to scheduling, availability, and commitment that need to be explored before a group launches a new service. “I’d want to have discussions about how the palliative-care business model fits with our hospital medicine model and an agreement with the hospital on goals and metrics,” she says.

Your Page Is Welcomed

When faced with a potential palliative-care case, never hesitate to call, says Dr. Bekanich. Palliative specialists and teams expect to receive pages for such matters, and actually welcome them. The following are basic guidelines for when residents, early-career hospitalists and other providers should call the palliative-care service:

  • Patient has a serious medical condition that you are unfamiliar or uncomfortable with;
  • Consults for pain, dyspnea, or other distressing symptoms AND the patient is medically complex (tenuous cardiopulmonary status, chronic renal or liver disease, etc.);
  • Patients who have symptoms resistant to standard care;
  • Patients, family, or the referring team is unsatisfied with the consult;
  • Red flags are identified for patient safety (including disposition);
  • Ethical issues;
  • The case involves risk management;
  • Emotional involvement is becoming a strain;
  • Patient has a recent history of substance abuse; or
  • Patient volume increases and other patients are not receiving normal quality of care.

Hospitalists Fill a Need

Whether a full-fledged palliative-care service fits your group’s dynamic or not, hospitalists as a whole should be competent in basic palliative care. Community and rural hospitals need HM to bridge this gap and deliver quality care to seriously ill patients.

Dr. Harris

“I started at a community hospital, Eden Medical Center in Castro Valley, California. I had a personal interest in palliative care and realized there’s a tremendous need for it in community hospitals,” says Heather A. Harris, MD, a hospitalist at San Francisco General Hospital who previously worked with Dr. Pantilat’s palliative care service at UCSF. “We deal with end-of-life issues on a regular basis—whether recognized or not—based on our caseloads and requests for consultations.

“I got a little perspective about palliative care while a resident at UCSF. But as I’ve gotten further into this, I have come to realize that there is an actual skill set that needs to be learned to do it properly.”

 

 

Dr. Harris says there is a big difference between physicians helping patients with end-of-life issues the best they can and being part of a “dedicated, interdisciplinary team.”

“Palliative care is a wonderful opportunity for hospitalists,” she says. “It’s already part of your practice. Why not do it in a more organized fashion?” TH

Larry Beresford is a freelance medical writer based in Oakland, Calif.

References

  1. Palliative care programs continue rapid growth in U.S. hospitals. Center to Advance Palliative Care website. Available at: www.capc.org/news-and-events/releases/04-05-10. Accessed July 15, 2010.
  2. Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292.
  3. Morrison RS, Penrod JD, Cassel JB, et al. Cost savings associated with US hospital palliative care consultation programs. Arch Intern Med. 2008;168(16):1783-1790.

Training Opportunities

Resources available to hospitalists interested in integrating palliative care into their programs:

The American Academy of Hospice and Palliative Medicine (AAHPM), a 4,000-member professional society based in Glenview, Ill., offers annual educational meetings (the next is Feb. 16-19 in Vancouver), a self-study course, summaries of medical literature, and a Clinical Scholars Program with 40 hours of mentorship at one of eight AAHPM training sites. The website (www.aahpm.org) is home to the Hospice and Palliative Medicine (HPM) Fellowship Program Directory, which lists 74 active programs that offer 181 fellowships. The fellowships generally are for one year and include 27 research slots.

Starting in 2014, physicians who want to become board-certified in HPM must complete an American College of Graduate Medical Education-accredited HPM fellowship (www.aahpm.org/certification/abms.html). Sitting for the boards based on work experience is still an option for the 2010 and 2012 exams. Palliative care leaders encourage interested working hospitalists to take advantage of this window of opportunity and join the more than 2,000 physicians who already are HPM certified.

The Center to Advance Palliative Care at Mount Sinai School of Medicine in New York City offers a variety of resources focused on palliative-care program development, including nine regional palliative care leadership centers, annual national training sessions, and financial models. For more info, visit www.capc.org.

Twice a year, Harvard Medical School offers the Program in Palliative Care Education and Practice (www.hms.harvard.edu/cdi/pallcare/pcep.htm) for physician and nurse educators who want to become experts in comprehensive, interdisciplinary palliative care.—LB

What Can Palliative Care Do to Hospitals’ Mortality Rates?

By Larry Beresford

Severity-adjusted hospital mortality rates are the cornerstone of a proliferating number of public and private hospital quality initiatives—and thus a quality focus for hospitalists. Yet some hospital-connected deaths are unavoidable, predictable, and even appropriate when palliative support focused on maximizing comfort and quality of life replaces medical efforts to stave off death.

Where hospice and palliative care fit in hospital mortality rates, how they are defined and counted, and how predictable deaths are either included or excluded from hospitals’ risk-adjusted mortality tallies vary between the reporting programs, according to J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University. He presented on mortality rates at the National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C. in April 2009.

“How hospital mortality rates are determined can be quite complex,” with varied data sources and various methods of adjusting for severity and balancing mortality with other quality metrics, says J. Brian Cassel, PhD, senior analyst at Virginia Commonwealth University who presented on mortality rates at the 2009 National Hospice and Palliative Care Organization’s Management and Leadership Conference in Washington, D.C.

Typically, the risk-adjusted mortality rate is for selected diagnoses but counts deaths from all causes, either during the index hospitalization or within 30 days of that admission, Cassel says. He reviewed three quality programs that use mortality data: the Centers for Medicare and Medicaid Services’ Hospital Compare, which publicly reports data on patient satisfaction and hospital processes and outcomes, including mortality; U.S. News & World Report’s “Best Hospitals”; and HealthGrades (www.healthgrades.com), a Golden, Colo.-based company that ranks hospitals and other health providers. He started studying the subject because of concerns that an acute palliative-care unit at VCU might be hurting the medical center’s overall mortality scores; it turned out not to be the case.

An ICD-9 (International Classifications of Disease) billing code, V66.7 for “palliative care encounter,” can flag the involvement of palliative- care consultants on a hospital case, although this code often goes unused and needs to be among the top nine listed diagnoses in order to turn up in most quality calculations. Palliative care consultants can help promote the use and higher positioning of this code in hospital billing, along with more complete documentation of co-morbidities and symptoms. It is also possible that involving hospice and palliative care teams with seriously ill patients earlier in their disease progression could help manage their care in community settings, avoiding hospitalizations when death is likely in the next few months.

Some hospitals might choose to refer patients known to be close to death to contracted hospice programs—although this should be based on the best interests of the patient, not on improving the facility’s mortality rates. Cassel’s advice for hospice and palliative care advocates trying to stake their claim in hospital quality measurement: provide the best possible care to patients and families, but meanwhile, know which quality measurement systems the hospital’s leadership follows, and what these look for.

Issue
The Hospitalist - 2010(08)
Issue
The Hospitalist - 2010(08)
Publications
Publications
Article Type
Display Headline
A Resting Place
Display Headline
A Resting Place
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)

ONLINE EXCLUSIVE: Audio interviews with Project BOOST Michigan principals

Article Type
Changed
Fri, 09/14/2018 - 12:30
Display Headline
ONLINE EXCLUSIVE: Audio interviews with Project BOOST Michigan principals
Audio / Podcast
Issue
The Hospitalist - 2010(08)
Publications
Sections
Audio / Podcast
Audio / Podcast
Issue
The Hospitalist - 2010(08)
Issue
The Hospitalist - 2010(08)
Publications
Publications
Article Type
Display Headline
ONLINE EXCLUSIVE: Audio interviews with Project BOOST Michigan principals
Display Headline
ONLINE EXCLUSIVE: Audio interviews with Project BOOST Michigan principals
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)

Warm Welcome

Article Type
Changed
Fri, 09/14/2018 - 12:30
Display Headline
Warm Welcome

Advocates for SHM’s Project BOOST (Better Outcomes for Older Adults through Safe Transitions) presented a standing-room-only policy briefing June 8 on Capitol Hill to explain an innovative quality-improvement (QI) initiative and new collaboration with Blue Cross Blue Shield of Michigan (BCBSM) to increase patient safety and reduce preventable hospital readmissions.

“The room was packed,” says David Share, MD, BCBSM’s executive medical director for healthcare quality. About 60 people were in attendance, mostly House and Senate legislative aides, along with a few representatives of third-party health organizations. Dr. Share, one of the presenters, says many of the staffers were well aware of the challenges of hospital-based practice. “I would say the crowd was remarkably attentive during our presentation,” he says.

SHM developed BOOST in 2008 to help hospitals and hospitalists systematically improve discharge processes through evidence-based interventions, management tools and resources, and expert mentoring. In January, BOOST was implemented in 15 Michigan hospitals with financial support from BCBSM. A 20-hospital partnership with the California HealthCare Foundation was announced in April, and more than 60 hospitals in 24 states now participate.

“These legislative staffers, who are responsible for crafting health-reform legislation, were given an in-depth understanding of how the provider community can take ownership of the challenges of transforming systems of care,” Dr. Share says. “I hope what they learned was that when payors … establish incentives for providers to transform healthcare systems, providers can do that very creatively and effectively in ways that affect patient care, patient well-being, and patient outcomes—both in terms of quality and cost.”

Hospitalist Scott Flanders, MD, SFHM, professor of medicine and director of the inpatient program at the University of Michigan in Ann Arbor, also spoke at the briefing. “I think our Michigan collaborative is a nice example of a local, provider-based, payor-supported quality initiative that will tackle an important problem and lead to a lot of collaboration and learning,” says Dr. Flanders, SHM’s immediate past president.

Also speaking at the briefing were Project BOOST principal investigator Mark Williams, MD, FHM, chief of the division of hospital medicine at Northwestern University’s Feinberg School of Medicine in Chicago, and representatives of the national Blue Cross/Blue Shield Association and the American Hospital Association, who discussed other initiatives that have successfully targeted the hospital readmission problem. “Not all readmissions are preventable. Some are necessary and important,” Dr. Flanders says, adding that the challenge is to distinguish between the necessary and the avoidable.

While there are no current legislative proposals involving Project BOOST, the initiative is aligned with a number of provisions aimed at reducing readmissions and improving care transitions, which are contained in the Patient Protection and Affordable Care Act passed in March. “Given the costs of readmissions, directly supporting demonstration projects like this would be a wise investment in improving healthcare quality,” says Dr. Flanders, adding that he heard suggestions at the briefing that the Centers for Medicare & Medicaid Services’ (CMS) Center for Innovation should consider supporting initiatives like BOOST.

Dr. Share, who calls payor support for the BOOST collaboration an example of its incentive programs with physician groups, says hospitalists are essential to partnerships with other providers, including PCPs, and the systems improvements necessary in the hospital setting.

“We’re actually bridging the gap between the hospital and the medical office,” he says. TH

Issue
The Hospitalist - 2010(08)
Publications
Sections

Advocates for SHM’s Project BOOST (Better Outcomes for Older Adults through Safe Transitions) presented a standing-room-only policy briefing June 8 on Capitol Hill to explain an innovative quality-improvement (QI) initiative and new collaboration with Blue Cross Blue Shield of Michigan (BCBSM) to increase patient safety and reduce preventable hospital readmissions.

“The room was packed,” says David Share, MD, BCBSM’s executive medical director for healthcare quality. About 60 people were in attendance, mostly House and Senate legislative aides, along with a few representatives of third-party health organizations. Dr. Share, one of the presenters, says many of the staffers were well aware of the challenges of hospital-based practice. “I would say the crowd was remarkably attentive during our presentation,” he says.

SHM developed BOOST in 2008 to help hospitals and hospitalists systematically improve discharge processes through evidence-based interventions, management tools and resources, and expert mentoring. In January, BOOST was implemented in 15 Michigan hospitals with financial support from BCBSM. A 20-hospital partnership with the California HealthCare Foundation was announced in April, and more than 60 hospitals in 24 states now participate.

“These legislative staffers, who are responsible for crafting health-reform legislation, were given an in-depth understanding of how the provider community can take ownership of the challenges of transforming systems of care,” Dr. Share says. “I hope what they learned was that when payors … establish incentives for providers to transform healthcare systems, providers can do that very creatively and effectively in ways that affect patient care, patient well-being, and patient outcomes—both in terms of quality and cost.”

Hospitalist Scott Flanders, MD, SFHM, professor of medicine and director of the inpatient program at the University of Michigan in Ann Arbor, also spoke at the briefing. “I think our Michigan collaborative is a nice example of a local, provider-based, payor-supported quality initiative that will tackle an important problem and lead to a lot of collaboration and learning,” says Dr. Flanders, SHM’s immediate past president.

Also speaking at the briefing were Project BOOST principal investigator Mark Williams, MD, FHM, chief of the division of hospital medicine at Northwestern University’s Feinberg School of Medicine in Chicago, and representatives of the national Blue Cross/Blue Shield Association and the American Hospital Association, who discussed other initiatives that have successfully targeted the hospital readmission problem. “Not all readmissions are preventable. Some are necessary and important,” Dr. Flanders says, adding that the challenge is to distinguish between the necessary and the avoidable.

While there are no current legislative proposals involving Project BOOST, the initiative is aligned with a number of provisions aimed at reducing readmissions and improving care transitions, which are contained in the Patient Protection and Affordable Care Act passed in March. “Given the costs of readmissions, directly supporting demonstration projects like this would be a wise investment in improving healthcare quality,” says Dr. Flanders, adding that he heard suggestions at the briefing that the Centers for Medicare & Medicaid Services’ (CMS) Center for Innovation should consider supporting initiatives like BOOST.

Dr. Share, who calls payor support for the BOOST collaboration an example of its incentive programs with physician groups, says hospitalists are essential to partnerships with other providers, including PCPs, and the systems improvements necessary in the hospital setting.

“We’re actually bridging the gap between the hospital and the medical office,” he says. TH

Advocates for SHM’s Project BOOST (Better Outcomes for Older Adults through Safe Transitions) presented a standing-room-only policy briefing June 8 on Capitol Hill to explain an innovative quality-improvement (QI) initiative and new collaboration with Blue Cross Blue Shield of Michigan (BCBSM) to increase patient safety and reduce preventable hospital readmissions.

“The room was packed,” says David Share, MD, BCBSM’s executive medical director for healthcare quality. About 60 people were in attendance, mostly House and Senate legislative aides, along with a few representatives of third-party health organizations. Dr. Share, one of the presenters, says many of the staffers were well aware of the challenges of hospital-based practice. “I would say the crowd was remarkably attentive during our presentation,” he says.

SHM developed BOOST in 2008 to help hospitals and hospitalists systematically improve discharge processes through evidence-based interventions, management tools and resources, and expert mentoring. In January, BOOST was implemented in 15 Michigan hospitals with financial support from BCBSM. A 20-hospital partnership with the California HealthCare Foundation was announced in April, and more than 60 hospitals in 24 states now participate.

“These legislative staffers, who are responsible for crafting health-reform legislation, were given an in-depth understanding of how the provider community can take ownership of the challenges of transforming systems of care,” Dr. Share says. “I hope what they learned was that when payors … establish incentives for providers to transform healthcare systems, providers can do that very creatively and effectively in ways that affect patient care, patient well-being, and patient outcomes—both in terms of quality and cost.”

Hospitalist Scott Flanders, MD, SFHM, professor of medicine and director of the inpatient program at the University of Michigan in Ann Arbor, also spoke at the briefing. “I think our Michigan collaborative is a nice example of a local, provider-based, payor-supported quality initiative that will tackle an important problem and lead to a lot of collaboration and learning,” says Dr. Flanders, SHM’s immediate past president.

Also speaking at the briefing were Project BOOST principal investigator Mark Williams, MD, FHM, chief of the division of hospital medicine at Northwestern University’s Feinberg School of Medicine in Chicago, and representatives of the national Blue Cross/Blue Shield Association and the American Hospital Association, who discussed other initiatives that have successfully targeted the hospital readmission problem. “Not all readmissions are preventable. Some are necessary and important,” Dr. Flanders says, adding that the challenge is to distinguish between the necessary and the avoidable.

While there are no current legislative proposals involving Project BOOST, the initiative is aligned with a number of provisions aimed at reducing readmissions and improving care transitions, which are contained in the Patient Protection and Affordable Care Act passed in March. “Given the costs of readmissions, directly supporting demonstration projects like this would be a wise investment in improving healthcare quality,” says Dr. Flanders, adding that he heard suggestions at the briefing that the Centers for Medicare & Medicaid Services’ (CMS) Center for Innovation should consider supporting initiatives like BOOST.

Dr. Share, who calls payor support for the BOOST collaboration an example of its incentive programs with physician groups, says hospitalists are essential to partnerships with other providers, including PCPs, and the systems improvements necessary in the hospital setting.

“We’re actually bridging the gap between the hospital and the medical office,” he says. TH

Issue
The Hospitalist - 2010(08)
Issue
The Hospitalist - 2010(08)
Publications
Publications
Article Type
Display Headline
Warm Welcome
Display Headline
Warm Welcome
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)

The Coming Challenges—and Opportunities—of Value-Based Purchasing

Article Type
Changed
Wed, 03/27/2019 - 13:20
Display Headline
The Coming Challenges—and Opportunities—of Value-Based Purchasing

The Coming Challenges—and Opportunities—of Value-Based Purchasing

The Patient Protection and Affordable Care Act was signed into law in March, furthering the federal government’s commitment to increasing the efficiency of the U.S. healthcare system by decreasing cost and improving quality. An expansion of the “value-based purchasing” model, this law mandates that ratings and reimbursements to physicians and hospitals be increasingly tied to measured quality of care.

In this system, a physician’s quality profile will be determined by a variety of factors, including reported quality data, severity-adjusted clinical outcome measures, patient-safety indicators, and hospital-acquired conditions (HACs). Since all of these are, to a large extent, documentation issues, physicians are now forced to pay close attention to how they identify and describe diagnoses and procedures.

Medicare will, in effect, attempt to determine: “Did the clinical team correctly identify and appropriately treat all relevant patient conditions—without causing any adverse conditions—and do so safely, efficiently, and with good outcomes?”

The patient chart must “tell the story” of the episode of care in the hospital. It must accurately describe all of the patient’s conditions and demonstrate the complexity of medical decision-making and establishment of risk. Upon discharge, this risk must “match up” with the diagnoses that are being coded and the Medicare Severity Diagnostic Related Group (MS-DRGs) being assigned. Often, the information is present in the chart but is inconsistent from provider to provider, or documented in a way that is misunderstood by hospital coders.

If successful in improving our documentation skills, the reward will be a higher rating and increased reimbursement.

Medicare also is ramping up its claims denial and recovery business to help “clean up” the system. This includes the national rollout of the Recovery Audit Contactor (RAC) initiative (see “Attention to Detail,” April 2010, p. 1), as well as the new Medicare administrative contractors (MACs). Both rely on accurate documentation. The RACs will penalize hospitals and physicians financially for documentation lacking in specificity and accuracy; the MACs will deny payment for claims erroneously submitted (technical errors) or lacking documentation of medical necessity.

What makes many physicians even more uncomfortable in this new environment is that Medicare is striving to better align the priorities and financial incentives of hospitals and physicians. Alliances between the two are strongly encouraged through such programs as the Acute Care Episode (ACE) Demonstration Project (the precursor to hospital-physician bundled payments), and the establishment of accountable care organizations (ACOs).

This emerging environment presents a unique set of challenges and opportunities to HM groups. Hospitalists are historically more aligned with hospital administrations, as compared with most other specialties. Hospitalists also are asked to participate in the care of an increasingly large percentage of patients across all specialties. This could well be an opportunity to be rewarded for embracing both of these trends.

Hospitalists are uniquely positioned to function as the documentation improvement clinical team leaders, working closely with other physicians across all specialties and the administration to fulfill all documentation requirements—and to be rewarded for doing so. Though hospitalists should have a general understanding of the language and rules of documentation, a system must be in place that helps them identify and capture all the pertinent aspects of the medical record without the need to become coders themselves. To this end, a clinical documentation improvement (CDI) program is critical.

But it might not be enough.

What is needed is a clinical integration program, an enhancement of traditional CDI. This approach requires participation from ED physicians, along with clinical integration specialists, to document accurately and completely from the start. The clinical integration specialist ensures that medical necessity for inpatient admission and patient risk is being addressed and established, conditions are appropriately identified as being present on admission (POA), and all diagnoses are properly recognized and documented thoroughly and accurately. Clinical integration through collaborative documentation then continues throughout the hospitalization, with diagnostic authority and oversight from the hospitalist, all the way through discharge.

 

 

Hospitalists should welcome and champion this type of program. As documentation becomes the key to survival, a complete medical record will stand up to any and all scrutiny by Medicare or others.

As for the negatives, there are none.

Andrew H. Dombro, MD, national medical director, and Paul Weygandt, MD, JD, vice president of physician services, J.A. Thomas & Associates, Atlanta

Issue
The Hospitalist - 2010(08)
Publications
Topics
Sections

The Coming Challenges—and Opportunities—of Value-Based Purchasing

The Patient Protection and Affordable Care Act was signed into law in March, furthering the federal government’s commitment to increasing the efficiency of the U.S. healthcare system by decreasing cost and improving quality. An expansion of the “value-based purchasing” model, this law mandates that ratings and reimbursements to physicians and hospitals be increasingly tied to measured quality of care.

In this system, a physician’s quality profile will be determined by a variety of factors, including reported quality data, severity-adjusted clinical outcome measures, patient-safety indicators, and hospital-acquired conditions (HACs). Since all of these are, to a large extent, documentation issues, physicians are now forced to pay close attention to how they identify and describe diagnoses and procedures.

Medicare will, in effect, attempt to determine: “Did the clinical team correctly identify and appropriately treat all relevant patient conditions—without causing any adverse conditions—and do so safely, efficiently, and with good outcomes?”

The patient chart must “tell the story” of the episode of care in the hospital. It must accurately describe all of the patient’s conditions and demonstrate the complexity of medical decision-making and establishment of risk. Upon discharge, this risk must “match up” with the diagnoses that are being coded and the Medicare Severity Diagnostic Related Group (MS-DRGs) being assigned. Often, the information is present in the chart but is inconsistent from provider to provider, or documented in a way that is misunderstood by hospital coders.

If successful in improving our documentation skills, the reward will be a higher rating and increased reimbursement.

Medicare also is ramping up its claims denial and recovery business to help “clean up” the system. This includes the national rollout of the Recovery Audit Contactor (RAC) initiative (see “Attention to Detail,” April 2010, p. 1), as well as the new Medicare administrative contractors (MACs). Both rely on accurate documentation. The RACs will penalize hospitals and physicians financially for documentation lacking in specificity and accuracy; the MACs will deny payment for claims erroneously submitted (technical errors) or lacking documentation of medical necessity.

What makes many physicians even more uncomfortable in this new environment is that Medicare is striving to better align the priorities and financial incentives of hospitals and physicians. Alliances between the two are strongly encouraged through such programs as the Acute Care Episode (ACE) Demonstration Project (the precursor to hospital-physician bundled payments), and the establishment of accountable care organizations (ACOs).

This emerging environment presents a unique set of challenges and opportunities to HM groups. Hospitalists are historically more aligned with hospital administrations, as compared with most other specialties. Hospitalists also are asked to participate in the care of an increasingly large percentage of patients across all specialties. This could well be an opportunity to be rewarded for embracing both of these trends.

Hospitalists are uniquely positioned to function as the documentation improvement clinical team leaders, working closely with other physicians across all specialties and the administration to fulfill all documentation requirements—and to be rewarded for doing so. Though hospitalists should have a general understanding of the language and rules of documentation, a system must be in place that helps them identify and capture all the pertinent aspects of the medical record without the need to become coders themselves. To this end, a clinical documentation improvement (CDI) program is critical.

But it might not be enough.

What is needed is a clinical integration program, an enhancement of traditional CDI. This approach requires participation from ED physicians, along with clinical integration specialists, to document accurately and completely from the start. The clinical integration specialist ensures that medical necessity for inpatient admission and patient risk is being addressed and established, conditions are appropriately identified as being present on admission (POA), and all diagnoses are properly recognized and documented thoroughly and accurately. Clinical integration through collaborative documentation then continues throughout the hospitalization, with diagnostic authority and oversight from the hospitalist, all the way through discharge.

 

 

Hospitalists should welcome and champion this type of program. As documentation becomes the key to survival, a complete medical record will stand up to any and all scrutiny by Medicare or others.

As for the negatives, there are none.

Andrew H. Dombro, MD, national medical director, and Paul Weygandt, MD, JD, vice president of physician services, J.A. Thomas & Associates, Atlanta

The Coming Challenges—and Opportunities—of Value-Based Purchasing

The Patient Protection and Affordable Care Act was signed into law in March, furthering the federal government’s commitment to increasing the efficiency of the U.S. healthcare system by decreasing cost and improving quality. An expansion of the “value-based purchasing” model, this law mandates that ratings and reimbursements to physicians and hospitals be increasingly tied to measured quality of care.

In this system, a physician’s quality profile will be determined by a variety of factors, including reported quality data, severity-adjusted clinical outcome measures, patient-safety indicators, and hospital-acquired conditions (HACs). Since all of these are, to a large extent, documentation issues, physicians are now forced to pay close attention to how they identify and describe diagnoses and procedures.

Medicare will, in effect, attempt to determine: “Did the clinical team correctly identify and appropriately treat all relevant patient conditions—without causing any adverse conditions—and do so safely, efficiently, and with good outcomes?”

The patient chart must “tell the story” of the episode of care in the hospital. It must accurately describe all of the patient’s conditions and demonstrate the complexity of medical decision-making and establishment of risk. Upon discharge, this risk must “match up” with the diagnoses that are being coded and the Medicare Severity Diagnostic Related Group (MS-DRGs) being assigned. Often, the information is present in the chart but is inconsistent from provider to provider, or documented in a way that is misunderstood by hospital coders.

If successful in improving our documentation skills, the reward will be a higher rating and increased reimbursement.

Medicare also is ramping up its claims denial and recovery business to help “clean up” the system. This includes the national rollout of the Recovery Audit Contactor (RAC) initiative (see “Attention to Detail,” April 2010, p. 1), as well as the new Medicare administrative contractors (MACs). Both rely on accurate documentation. The RACs will penalize hospitals and physicians financially for documentation lacking in specificity and accuracy; the MACs will deny payment for claims erroneously submitted (technical errors) or lacking documentation of medical necessity.

What makes many physicians even more uncomfortable in this new environment is that Medicare is striving to better align the priorities and financial incentives of hospitals and physicians. Alliances between the two are strongly encouraged through such programs as the Acute Care Episode (ACE) Demonstration Project (the precursor to hospital-physician bundled payments), and the establishment of accountable care organizations (ACOs).

This emerging environment presents a unique set of challenges and opportunities to HM groups. Hospitalists are historically more aligned with hospital administrations, as compared with most other specialties. Hospitalists also are asked to participate in the care of an increasingly large percentage of patients across all specialties. This could well be an opportunity to be rewarded for embracing both of these trends.

Hospitalists are uniquely positioned to function as the documentation improvement clinical team leaders, working closely with other physicians across all specialties and the administration to fulfill all documentation requirements—and to be rewarded for doing so. Though hospitalists should have a general understanding of the language and rules of documentation, a system must be in place that helps them identify and capture all the pertinent aspects of the medical record without the need to become coders themselves. To this end, a clinical documentation improvement (CDI) program is critical.

But it might not be enough.

What is needed is a clinical integration program, an enhancement of traditional CDI. This approach requires participation from ED physicians, along with clinical integration specialists, to document accurately and completely from the start. The clinical integration specialist ensures that medical necessity for inpatient admission and patient risk is being addressed and established, conditions are appropriately identified as being present on admission (POA), and all diagnoses are properly recognized and documented thoroughly and accurately. Clinical integration through collaborative documentation then continues throughout the hospitalization, with diagnostic authority and oversight from the hospitalist, all the way through discharge.

 

 

Hospitalists should welcome and champion this type of program. As documentation becomes the key to survival, a complete medical record will stand up to any and all scrutiny by Medicare or others.

As for the negatives, there are none.

Andrew H. Dombro, MD, national medical director, and Paul Weygandt, MD, JD, vice president of physician services, J.A. Thomas & Associates, Atlanta

Issue
The Hospitalist - 2010(08)
Issue
The Hospitalist - 2010(08)
Publications
Publications
Topics
Article Type
Display Headline
The Coming Challenges—and Opportunities—of Value-Based Purchasing
Display Headline
The Coming Challenges—and Opportunities—of Value-Based Purchasing
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)