Você está na página 1de 11

Vasopressin Excess and Hyponatremia

Phuong-Chi T. Pham, MD, Phuong-Mai T. Pham, MD, and Phuong-Thu T. Pham, MD

● Hyponatremia is a common electrolyte disorder that frequently is overlooked and undertreated. Although the
pathophysiological process of hyponatremia is complex, arginine vasopressin (AVP) is a common etiologic factor.
Excess AVP release by osmotic or nonosmotic stimuli or both can lead to sodium and water imbalance. Conventional
treatment options for hyponatremia, including water restriction and administration of sodium chloride with or without
loop diuretics, do not directly address the underlying water retention induced by excess AVP in many cases.
Clinical trials showed that AVP-receptor antagonists, including lixivaptan, tolvaptan, and conivaptan, produce
aquaresis, the electrolyte-sparing excretion of free water, to correct serum sodium concentration. We review results
from recent clinical trials involving AVP-receptor antagonists in the treatment of hyponatremia associated with AVP
excess. Am J Kidney Dis 47:727-737.
© 2006 by the National Kidney Foundation, Inc.

INDEX WORDS: Hyponatremia; arginine vasopressin–receptor antagonist; syndrome of inappropriate secretion of


antidiuretic hormone; cirrhosis; congestive heart failure.

H YPONATREMIA, USUALLY defined as a


serum sodium concentration less than 136
mEq/L (⬍136 mmol/L), is a common, yet fre-
and prone to complications. The use of loop
diuretics and water restriction, for example, may
lead to volume depletion–induced renal failure
quently overlooked and undertreated, condi- and unpleasant lifestyle changes, respectively.
tion.1,2 Depending on the definition used, the In the treatment of significant or life-threaten-
prevalence of hyponatremia in hospitalized pa- ing hyponatremia in patients with edematous
tients was reported to range from 3% to 15%.3-5 states, another challenge arises when free-water
Hyponatremia per se has been well documented as restriction cannot provide a timely improvement.
a potentially life-threatening condition, as well as In such cases, a loop diuretic may be adminis-
an independent predictor of death in intensive care tered, but this approach can induce profound
unit and geriatric patients and patients with heart hypotension, hypokalemia, potentially worsen-
failure, acute ST elevation myocardial infarction, ing hyponatremia, and renal failure. In cases of
and cirrhosis.6-12 severe hyponatremia, sodium infusion may be
Despite the high prevalence of hyponatremia considered at the expense of exacerbating the
in hospitalized patients and its association with preexisting hypervolemic state.
adverse overall outcomes, many clinicians fail to Until recently, clinicians had not been able to
address this condition at its early stage because more directly treat one of the most common
of its nonspecific symptoms and often complex causes of hyponatremia, namely, excessive secre-
management options. tion of antidiuretic hormone, otherwise known as
For many years, sodium replacement, rela- arginine vasopressin (AVP).13,14 AVP-receptor
tively arbitrary free-water restriction, or both,
with or without the use of loop diuretics, com-
prised the major therapeutic options for hypona- From the Nephrology Division, Olive View-UCLA Medi-
tremia. The management of hyponatremia is a cal Center, Sylmar; Department of Medicine, Kidney and
Pancreas Transplantation, David Geffen School of Medicine
multistep process that demands great focus from at UCLA, Los Angeles, CA; and Central Maine Medical
the clinician because he or she must determine the Center, Lewiston, ME.
appropriate rate of correction according to the Received September 13, 2005; accepted in revised form
chronicity of the disorder, select the best therapeu- January 24, 2006.
Originally published online as doi:10.1053/j.ajkd.2006.01.020
tic option based on the cause of hyponatremia, on March 23, 2006.
predict dynamic changes in salt and water bal- Support: None. Potential conflicts of interest: None.
ance according to the underlying cause and ongo- Address reprint requests to Phuong-Chi T. Pham, MD,
ing acute illnesses, closely monitor changes in Olive View-UCLA Medical Center, Department of Medicine,
serum sodium levels, and adjust the patient’s Nephrology Division, 14445 Olive View Dr, 2B-182 Sylmar,
CA 91342. E-mail: pctp@ucla.edu
therapy or rate of sodium infusion accordingly. © 2006 by the National Kidney Foundation, Inc.
To further challenge the clinician, currently avail- 0272-6386/06/4705-0002$32.00/0
able therapeutic options may be unpredictable doi:10.1053/j.ajkd.2006.01.020

American Journal of Kidney Diseases, Vol 47, No 5 (May), 2006: pp 727-737 727
728 PHAM, PHAM, AND PHAM

antagonists are a new class of drugs designed


specifically to promote aquaresis, the electrolyte-
sparing excretion of free water, and subsequently
increase serum sodium levels.
AVP IN WATER AND ELECTROLYTE BALANCE
Overview of AVP Release
AVP is produced predominantly in magnocel-
lular neurons in the paraventricular and supraop-
tic nuclei in the hypothalamus, subsequently
transported down axon terminals through the
supraoptic hypophyseal tract, and eventually
stored in the posterior pituitary as inactive com-
plexes with neurophysins.15,16 AVP is released
quantally by exocytosis from secretory granules
in response to osmotic and nonosmotic stimuli.15
An increase in serum osmolality leads to extra-
cellular water shift in the hypothalamus osmore-
ceptor cells. The resultant cell-volume shrinkage
or increased intracellular osmolality in osmore-
ceptor cells initiates a cascade of events leading
to enhanced AVP synthesis and release.17 Os- Fig 1. Physiological relations between plasma os-
motic changes that stimulate AVP release also molality, plasma AVP concentrations, urine volume,
enhance its production. In humans, the osmotic and urine osmolality in healthy humans. To convert
AVP in pg/mL to pmol/L, multiply by 0.923; osmolality
threshold for AVP release is approximately 280 in mOsm/kg to mmol/kg, multiply by 1. (Adapted from
to 290 mOsm/kg (280 to 290 mmol/kg).18 With Best Pract Res Clin Endocrinol Metab 17; Verbalis JG,
serum osmolality greater than the osmotic thresh- Disorders of body water homeostasis, pp 471-503,
200314; and Clin Endocrinol Metab 14, Robinson AG,
old, AVP release is increased linearly. A 2- to Disorders of antidiuretic hormone secretion, pp 55-88,
4-fold increase in AVP level may be observed 1985,23 with permission from Elsevier.)
with each 1% increase in serum osmolality.19
The exact threshold for AVP release decreases pattern observed in approximately 36% of pa-
with age and during pregnancy.20,21 The slope of tients in whom a normal linear AVP response is
the AVP response to plasma osmolality tends to preserved, but is initiated at a lower osmotic
vary among individuals and may be genetically threshold than that observed in most of the popu-
determined; however, the slope of AVP response lation. Type C, observed in 32% of patients, is
within each individual may alter in response to characterized by normal AVP release when se-
blood volume and pressure.22 Renal water han- rum osmolality is normal or elevated, but AVP
dling also is linear in response to AVP. An release is not inhibited when serum osmolality is
increasing systemic AVP level results in a propor- low. Type D, a pattern observed in 8% of pa-
tional decrease in urine flow and increase in tients, is associated with normal osmoregulated
urine osmolality (Fig 1).23 AVP release, but with an inability to dilute urine
In patients with hyponatremia associated with or excrete a normal water load. This condition
the syndrome of inappropriate antidiuretic hor- could be caused by enhanced AVP sensitivity or
mone release (SIADH), AVP release may follow the presence of other antidiuretic factors.24
1 of 4 distinct patterns and not necessarily reflect Nonosmotic stimuli of AVP release include
osmotic or volume changes (Fig 2).13,24 Type A, hypovolemia, a decrease in arterial blood pres-
found in 24% of patients from a small study sure, neural regulation by various neurotransmit-
cohort involving 25 patients with SIADH, is ters and neuropeptides in the hypothalamus, and
characterized by erratic fluctuations in AVP re- various pharmacological agents. Decreases in
lease that are independent of serum osmolality. plasma volume exceeding 8% may exponentially
Type B, also known as “reset osmostat,” is a stimulate the release of AVP, presumably in part
VASOPRESSIN EXCESS AND HYPONATREMIA 729

peutic agents, tricyclic anticonvulsants, antide-


pressants, clofibrate, and chlorpropamide, and
such conditions as nausea, vomiting, pain, and
A hypoglycemia.27,28
20
AVP Receptors
Plasma AVP (pmol/L)

The neurohypophyseal hormone AVP exerts


its actions through G-protein–coupled mem-
brane receptors pharmacologically subtyped as
V1A (vascular), V2 (renal), and V3 (pituitary),
also denoted as V1B in the literature.
10 B
V1A receptors are found on vascular smooth
muscle, liver, kidneys, reproductive organs, spleen,
adrenal cortex, and platelets. The vasoconstrictive,
mitogenic, and possibly platelet aggregative and
hypercoagulable actions of AVP through V1A theo-
C
retically may contribute to the pathogenesis and
LD
D
progression of arterial hypertension, heart fail-
ure, and atherosclerosis.29
260 280 300 V2 receptors are expressed primarily in baso-
Plasma Osmolality (mmol/kg) lateral membranes of renal cortical and medul-
lary collecting ducts and, to a lesser extent, distal
Fig 2. Four patterns of induced AVP release in tubules, where the antidiuretic effect of AVP is
patients with SIADH. Erratic AVP release unrelated to mediated. Binding of circulating AVP to the
(A) plasma osmolality, (B) regulation of water excre-
tion around a lower “reset osmostat” value, (C) AVP G-protein–coupled V2 receptor on basolateral
release when it cannot be “switched off” at low plasma membranes of the principal cells in collecting
osmolality, and (D) normal AVP release in patients ducts induces the activation of adenylyl cyclase,
with SIADH. The shaded area represents normal re-
sponses.13,24 To convert AVP in pg/mL to pmol/L, production of cyclic adenosine monophosphate,
multiply by 0.923; osmolality in mOsm/kg to mmol/kg, and activation of protein kinase A. The result is
multiply by 1. (Reprinted from Int J Biochem Cell Biol enhanced production and trafficking of the water
35; Baylis PH, The syndrome of inappropriate antidi-
uretic hormone secretion, pp 1495-1499, 2003,13 with channel aquaporin-2 to the luminal membranes,
permission from Elsevier. Copyright 1985, The Endo- where water reabsorption through osmotic equili-
crine Society.24) bration with the hypertonic medullary intersti-
tium occurs. Physiological actions of AVP through
by decreasing tonic inhibitory impulses from the V2 receptors have a major role in plasma tonic-
left atrial and possibly pulmonary vein stretch ity, volume regulation, and blood pressure main-
receptors to the hypothalamus.19 Hypotension, tenance.29,30
particularly secondary to blood loss, serves as a V3 (or V1B) receptors are found predomi-
potent stimulus for AVP release through activa- nantly in the anterior pituitary corticotroph cells
tion of carotid and aortic baroreceptors.25 In and, to a lesser extent, the brain, kidney, pan-
addition, the decrease in arterial blood pressure creas, and adrenal medulla. Physiologically, acti-
also may induce AVP release through activation vation of V3 receptors is associated with adreno-
of the sympathetic nervous system and renin- corticotropic hormone and ␤-endorphin release.29
angiotensin-aldosterone system.26 Many neuro-
transmitters and neuropeptides in the hypothala- AVP and Water Homeostasis
mus, including acetylcholine, angiotensin II, Total-body water content in healthy individu-
histamine, bradykinin, and neuropeptide Y, also als is estimated to be between 55% to 65% of
have been implicated in the stimulation of AVP total-body weight and is dependent on various
release.27,28 Other nonosmotic stimuli also may factors, including age, sex, and fat content.22
have a role, including various pharmacological Two thirds of the total-body water volume is
agents, such as morphine, nicotine, chemothera- distributed in the intracellular fluid, and one
730 PHAM, PHAM, AND PHAM

third, in the extracellular fluid. Two thirds of the ment, the thick ascending loop of Henle, by salt
extracellular fluid volume is in interstitial tis- extractions from the lumen through the sodium-
sues, and one third is in the intravascular space, potassium-2 chloride cotransporter. Intraluminal
of which 85% occupies the venous circulation free water is produced with this process because
and 15% occupies the arterial circulation.22 water cannot diffuse across the luminal surface
Maintenance of these intricate water partitions in parallel with salt uptake due to the unique
involves the movement of osmotically active water-impermeable characteristic of this diluting
solutes and passive water diffusion between the segment. Reabsorption of salt without water con-
intracellular and extracellular fluid. In normal tinues to occur in the distal convoluted tubule,
human physiological states, sodium, potassium, where dilution of urine is maximized. While free
and glucose are the major osmotically active or water is produced within the lumen, salt uptake
effective solutes. Intercompartmental movement simultaneously enhances and maintains the greater
of any of these osmotically effective solutes tonicity of the medullary interstitium. Subse-
necessitates either an active/facilitated reverse- quently, the free water formed may be either reab-
transport process or the obligatory parallel diffu- sorbed or excreted into urine at the collecting ducts,
sion of water across the 2 compartments to main- as dictated by AVP. In the presence of AVP and
tain equivalent osmotic pressures.31 Solutes, such an intact hypertonic medullary interstitium, AVP,
as urea, are considered ineffective because they through V2 receptors, upregulates the expression
can move freely across cell membranes bidirec- and trafficking of aquaporin-2 to the luminal side
tionally without necessitating any other process of the collecting tubules and allows water to be
to maintain the osmolality of either compart- reabsorbed efficiently down a more tonic medul-
ment. lary milieu. AVP-facilitated free-water reabsorp-
Whereas intracellular water homeostasis is tion decreases urine volume and increases urine
dependent on osmotically active solutes, total- concentration.14 In the absence of AVP, the free
body water balance is determined and main- water produced from the diluting segment is
tained predominantly by the thirst center and excreted and is reflected clinically as high urine
renal water excretion.14 Thirst, a physiological output with low osmolality.
response to increase free-water uptake, may be
stimulated by hyperosmolality, profound volume AVP Excess and Hyponatremia
contraction, and gastric salt loading.32 With expo- Although AVP is the major regulatory factor in
sure to hyperosmolality, osmoreceptor cells in the maintenance of total-body water, its exces-
the subfornical organ and organum vasculosum sive production and release can result in pro-
laminae terminalis activate neurons projecting to found water retention and hyponatremia.13 In
the paraventricular and supraoptic nuclei in the hypovolemia, volume loss or decrease in arterial
hypothalamus to stimulate thirst. In addition, blood pressure promotes AVP release appropri-
profound volume contraction caused by a signifi- ately to enhance both thirst and renal water
cant decrease in arterial blood pressure or vol- retention. Hyponatremia occurs when free water
ume depletion may induce an increase in levels is replaced in excess of salt.
of intracerebroventricular angiotensin II, a strong In hypervolemia caused by congestive heart
dipsogenic factor.33 Finally, thirst may be stimu- failure (CHF), blood volume is expanded, but
lated by gastric sodium loading, presumably cannot be translocated effectively from the ve-
through sodium receptors in the abdominal vis- nous to arterial circulation to support adequate
cera (such as hepatic portal osmoreceptors), inde- cardiac output. As a result, effective arterial
pendent of any increase in systemic plasma osmo- volume, pressure, or both are decreased in a
lality.34 manner similar to that in a hypovolemic state. To
Renal free-water excretion is dependent on the expand the effective arterial volume, AVP release
integrity of nephron-diluting segments, mainte- is enhanced to increase thirst, water intake, and
nance of a hypertonic medullary interstitium, renal water retention.35 With normal renal perfu-
and intact AVP activity at the collecting ducts. sion, as much as 70% of glomerular filtrate is
For renal water excretion to occur, free water reabsorbed proximally, leaving the remaining
first is produced in the nephron-diluting seg- volume to reach the loop of Henle, where the
VASOPRESSIN EXCESS AND HYPONATREMIA 731

upper limit on urine flow and output is deter- tion; and (5) high renal sodium excretion (ie,
mined. As renal perfusion decreases in associa- ⬎40 mEq/L [⬎40 mmol/L]).41
tion with low cardiac output, glomerular filtrate The major categories of illnesses associated
volume decreases, increased proximal filtrate re- with SIADH include neoplasm, neurological dis-
absorption occurs, and thus a smaller remaining ease or trauma, pulmonary disorders, and various
filtrate volume is delivered to the diluting seg- stress states, such as nausea, vomiting, pain, and
ment for free-water production and subsequent anxiety. In addition, SIADH may be caused by a
excretion. Any free water that can be produced in number of medications, particularly tricyclic an-
the subsequent diluting segment will be reab- tidepressants and anticonvulsants.1,13,40,42
sorbed avidly in the collecting ducts through the
action of AVP on V2 receptors. The severe de- Impact of Age on AVP
crease in urine volume (salt and water retention) The pathophysiological processes of age-
and free-water clearance (FWC; water retention) related disturbances in water homeostasis and
induced by the decreased glomerular filtrate vol- the resultant hyponatremia are complex and most
ume and secondary excessive AVP release per- likely multifactorial. In older individuals, total-
petuate the edematous state, while exacerbating body water content is as much as 20% less than
hyponatremia. In addition to volume expansion that in younger individuals.43 A change in salt
and hyponatremia, the elevated AVP levels in and water balance therefore may result in a more
patients with CHF secondarily may increase sys- pronounced effect on body fluid osmolality and
temic vascular resistance through V1A vasocon- various salt concentrations. The development of
strictive activity and decrease cardiac output.30 hyponatremia specifically may be attributed to
Accordingly, the excess AVP-induced V1A vaso- the decreased capacity for free-water handling
constrictive effect theoretically may be harmful because of various factors, including a decrease
to some patients with CHF. in glomerular filtration rate, decrease in solute
As in patients with CHF, those with cirrhosis load caused by poor nutritional intake, and en-
or nephrosis who have poor arterial pressure hanced osmotic and nonosmotic AVP sensitiv-
because of peripheral vasodilation, low intravas- ity.20,44 In addition, a blunted aldosterone re-
cular oncotic pressure, or both may present with sponse was observed in healthy elderly
decreased urine output, enhanced free-water re- individuals.45 In patients in whom water balance
absorption, and significant hyponatremia caused is simultaneously altered, hyponatremia may oc-
by poor renal perfusion and enhanced AVP re- cur.
lease.21,36 Of interest, prostaglandin synthesis
inhibitors, such as nonsteroidal anti-inflamma- PHARMACOLOGICAL OPTIONS
tory drugs, were observed to potentiate the anti- FOR HYPONATREMIA
diuretic effect of AVP, presumably through stimu- Current treatment options for patients with
lation of AVP-induced generation of cyclic hyponatremia, including sodium supplementa-
adenosine monophosphate, and thereby may ex- tion, water restriction, and concomitant diuretic
acerbate free-water retention and hyponatremia therapy, usually are effective, but not without
in patients with preexisting hypervolemia and limitations. Although sodium supplementation
low intravascular effective volume or pres- through saline infusion is effective in patients
sure.37-39 with hypovolemic hyponatremia, its use may
Euvolemic hyponatremia accounts for 60% be problematic in patients with hypervolemic
of all forms of chronic hyponatremia, of which hyponatremia because of the acute volume
SIADH, a disorder in which AVP release is expansion. Water restriction with or without
independent of volume, blood pressure, and os- concomitant diuretic administration in patients
motic control, predominates.13,40 SIADH is de- with euvolemic and hypervolemic hyponatremia
fined by 5 major criteria: (1) hypotonic hypona- may lead to a slow response, less predictable
tremia; (2) euvolemia; (3) inappropriately high outcomes, and, sometimes, serious complica-
urine osmolality (ie, ⬎70 to 100 mOsm/kg [⬎70 tions. For example, water restriction in patients
to 100 mmol/kg]) in the presence of hyponatre- with underlying malignancy or various other
mia; (4) normal renal, thyroid, and adrenal func- SIADH-associated conditions may lead to anxi-
732 PHAM, PHAM, AND PHAM

Table 1. Overview of AVP-Receptor Antagonists ics are associated with neurohormonal activation
involving the renin-angiotensin-aldosterone sys-
Receptor Disorders Studied in Clinical
Agent Target Trials tem and sympathetic nervous system and may
be particularly harmful to elderly and cardiac
Lixivaptan V2 CHF,49 SIADH,49 patients.31
cirrhosis48,49
Tolvaptan V2 CHF (NYHA class I-IV),50
(LVEF ⬍ 40%)51 Overview of AVP-Receptor Antagonists
Conivaptan V1A/V2 CHF NYHA class III and IV35 The role of AVP-receptor antagonists in the
Euvolemic or hypervolemic
treatment of patients with hyponatremia was
hyponatremia52,54,55
evaluated in clinical trials. This novel class of
Abbreviations: NYHA, New York Heart Association; agents was shown to promote aquaresis, the
LVEF, left ventricular ejection fraction. electrolyte-sparing excretion of free water.47 Cur-
rently, AVP-receptor antagonists directed at V2 re-
ety, malnourishment, and symptomatic volume ceptors (lixivaptan and tolvaptan) are in develop-
depletion. ment for clinical use in patients with euvolemic and
In cases of prolonged hyponatremia in which hypervolemic hyponatremia. Conivaptan, a V1A/V2-
fluid restriction is not tolerated, demeclocycline, receptor antagonist, recently was approved in the
a tetracycline antibiotic, may be an option. Nev- United States for use in hospitalized patients
ertheless, the use of demeclocycline is limited by with euvolemic hyponatremia (Tables 1 and 2).
poor results; troublesome adverse events, includ- Lixivaptan. The efficacy and tolerability of
ing azotemia, photosensitivity, and nausea; and lixivaptan (VPA-985), an oral V2-receptor antag-
contraindications in patients with cirrhosis or onist, was evaluated in patients with hyponatre-
renal failure.22,46 Conventional diuretics are ef- mia secondary to CHF, SIADH, and cirrhosis
fective in decreasing volume overload, but may with ascites. In a randomized, double-blind, mul-
lead to excessive salt and water excretion and ticenter trial, 60 patients with dilutional hypona-
result in or worsen effective arterial volume tremia (serum sodium levels between 115 and
depletion and hyponatremia. In addition, diuret- 132 mEq/L [115 and 132 mmol/L]) secondary to

Table 2. Effects of AVP-Receptor Antagonists in Patients With AVP Excess

Agent/Study No. of Urine Urine Serum Body Blood Heart Renal


Duration Reference Patients Output* FWC† Osmolality Sodium Weight Pressure Rate Thirst Function

Lixivaptan
7d Gerbes et al48‡ 60 1 1 2 1 2 NC NC 1 ND
Wong et al49§ 44 1 1 2 1 2 NC NC 1 NC
Tolvaptan
25 d Gheorghiade et al50储 254 1 NA 2 1 2 NC NC 1 NC
60 d Gheorghiade et al51¶ 319 1 NA NA 1 2 NC NC 1 NC
Conivaptan
1d Udelson et al35# 142 1 NA 2 1 NA NC NC NA NA
4d Verbalis et al52¶ 84 NA 1 NA 1 NA NA NA NA NA
5d Ghali et al54¶ 74 NA 1 NA 1 NA NA NA NA NA
Gross et al55¶ 83 NA 1 NA 1 NA NA NA NA NA

Abbreviations: 1, increase; 2, decrease (see text for actual statistical significance); NC, no change from baseline in all
groups; ND, no difference— glomerular filtration rate decreased approximately 10% from baseline in all active-treatment
groups and the placebo group, but the change was similar in all groups; NA, data not available.
*Urine output or net fluid balance (defined as urine output ⫺ fluid intake).
†In the 4- and 5-day conivaptan studies, effective water clearance was measured instead of FWC.
‡One liter per day fluid restriction.
§Fluid restriction of 1.5 L/d, but may be higher for those with high urine output (⬎3 L/d).
储No fluid restriction.
¶Freedom of fluid access not stated by investigators.
#Fluid restriction of 250 mL/2 h from time of insertion of pulmonary artery catheter throughout the treatment period.
VASOPRESSIN EXCESS AND HYPONATREMIA 733

cirrhosis were randomly assigned to administra- dietary sodium restriction determined by each
tion of placebo (n ⫽ 20); lixivaptan, 100 mg/d investigator and fluid restriction (1.5 L/d). The
(n ⫽ 22); or lixivaptan, 200 mg/d (n ⫽ 18).48 last condition was not strictly enforced because
Treatment was continued for as long as 7 days or patients with urine output greater than 3 L/d were
until the primary end point of normalization of allowed to have greater fluid intake. The actual
serum sodium levels (ⱖ136 mEq/L [ⱖ136 mmol/ amount of fluid intake in each group was not
L]) was reached. All patients continued to take reported in the study. Nevertheless, lixivaptan
their usual medications throughout the study increased net fluid volume (urine output ⫺ fluid
period. Normalization of serum sodium levels intake) and FWC significantly more than placebo
was achieved in 27% and 50% of patients admin- (P ⬍ 0.05), in accordance with the dosage admin-
istered lixivaptan, 100 and 200 mg/d, but in no istered. At the end of the 7-day study, FWC was
patient in the placebo group, respectively. Accord- ⫺0.3 ⫾ 0.2 mL/min in patients administered
ingly, lixivaptan increased serum sodium levels lixivaptan, 25 mg (P ⫽ not significant), 0.5 ⫾
from baseline more than placebo at study end 0.2 mL/min in those administered 125 mg (P ⬍
(128.3 ⫾ 4.1 to 130.4 ⫾ 6.5 mEq/L [128.3 ⫾ 4.1 0.01), 0.3 ⫾ 0.3 mL/min in those administered
to 130.4 ⫾ 6.5 mmol/L] in patients administered 250 mg (P ⬍ 0.01), and ⫺0.7 ⫾ 0.3 mL/min in
100 mg/d [P ⫽ 0.053], 126.4 ⫾ 4.4 to 132.3 ⫾ patients administered placebo.
6.9 mEq/L [126.4 ⫾ 4.4 to 132.3 ⫾ 6.9 mmol/L] In a separate analysis of patients who had
in those administered 200 mg/d [P ⬍ 0.001], and cirrhosis, serum sodium levels increased signifi-
127.3 ⫾ 3.0 to 127.7 ⫾ 4.0 mEq/L [127.3 ⫾ 3.0 cantly more between day 1 and the end of the
to 127.7 ⫾ 4.0 mmol/L] in those administered study in patients administered lixivaptan, 25 mg
placebo). Mean changes in serum sodium levels (126 ⫾ 1 mEq/L on day 1 versus 129 ⫾ 2 mEq/L
per day were 0.8 ⫾ 0.4/L/d, 1.8 ⫾ 0.5/L/d, and on day 7 [126 ⫾ 1 versus 129 ⫾ 2 mmol/L]; P ⬍
0.1 ⫾ 0.2/L/d in the 100-mg/d, 200-mg/d, and 0.05), 125 mg (122 ⫾ 2 versus 127 ⫾ 3 mEq/L,
placebo groups, respectively. The maximum in- respectively; P ⬍ 0.05), or 250 mg (125 ⫾ 1
crease in serum sodium levels with either dose of versus 132 ⫾ 1 mEq/L; P ⬍ 0.003) than in those
lixivaptan was 7/L/d. Both doses of lixivaptan administered placebo (127 ⫾ 1 versus 126 ⫾ 1
increased FWC significantly more than placebo mEq/L; P ⬍ 0.05). Accordingly, there was a
(lixivaptan, 100 mg/d, 0.3 ⫾ 0.3 mL/min [P ⬍ significant decrease in urine osmolality in pa-
0.01]; lixivaptan, 200 mg/d, 1.2 ⫾ 0.4 mL/min tients administered lixivaptan.
[P ⬍ 0.001]; and placebo, ⫺0.7 ⫾ 0.2 mL/min). Serum creatinine levels did not change signifi-
By the end of the study, glomerular filtration cantly in any of the patient groups throughout the
rate decreased by approximately 10% in both study period. However, the group administered
lixivaptan groups and the placebo group. Treat- the highest dose of lixivaptan (250 mg twice
ment with lixivaptan did not have an additional daily) consistently reported increased thirst and
negative impact on renal function. Use of either had significantly increased thirst scores. With
dose of lixivaptan was associated with increased respect to the vasoactive hormonal profile, includ-
thirst compared with placebo.48 ing vasopressin, norepinephrine, plasma renin
In another multicenter, randomized, placebo- activity, and aldosterone, lixivaptan only in-
controlled trial, lixivaptan was evaluated for its creased AVP levels significantly by study end.49
efficacy and safety in the correction of hyponatre- Tolvaptan. The efficacy and tolerability of
mia. The study involved 44 patients with hypona- tolvaptan (OPC-41061), an oral V2-receptor an-
tremia (serum sodium ⬍130 mEq/L [⬍130 mmol/ tagonist, were evaluated in patients with CHF. In
L]) secondary to CHF (n ⫽ 6), SIADH (n ⫽ 5), a randomized double-blind trial designed to inves-
and cirrhosis with ascites (n ⫽ 33).49 Patients tigate the effects of 3 doses of tolvaptan, 254
were randomly assigned to administration of patients with a “diagnosis of CHF” were ran-
placebo (n ⫽ 11) or oral lixivaptan at 25 mg domly assigned to administration of tolvaptan,
twice daily (n ⫽ 12), 125 mg twice daily (n ⫽ 11), 30 mg/d (n ⫽ 64), 45 mg/d (n ⫽ 64), and 60
or 250 mg twice daily (n ⫽ 10) during a 7-day mg/d (n ⫽ 63) or placebo (n ⫽ 63) in combina-
period. All patients continued to take their usual tion with standard furosemide therapy for 25
medications, including diuretics, and followed days.50 Tolvaptan was effective in increasing
734 PHAM, PHAM, AND PHAM

urine output, decreasing total-body weight, and Mean urine output at 24 hours also was less with
improving edema despite no fluid restriction. placebo (2,296.5 ⫾ 1,134.1 mL) than with each
Although fluid intake increased in patients admin- dose of tolvaptan (30 mg, 4,056.2 ⫾ 2,310.2 mL;
istered tolvaptan, 30, 45, or 60 mg, mean net P ⫽ 0.02; 60 mg, 4,175.2 ⫾ 2,695.4 mL; P ⬍
fluid loss (urine output ⫺ fluid intake) was signifi- 0.001; and 90 mg, 4,127.3 ⫾ 2,050.8 mL; P ⬍
cantly greater with each of these tolvaptan doses 0.001). This effect was maintained throughout
(1,337, 988, and 1,286 mL/d, respectively) than the hospitalization period.
with placebo (98 mL/d; P ⬍ 0.05). Mean change Patients recruited into the study had only mild
in urine osmolality from baseline to end of study hyponatremia (average serum sodium level,
also was significantly less with each tolvaptan 138.65 mEq/L [138.65 mmol/L]). Nonetheless,
dose (30 mg, 7.13 ⫾ 164.49 mOsm/kg [7.13 ⫾ at 1 day after randomization, there was a trend
164.49 mmol/kg]; 45 mg, ⫺19.38 ⫾ 157.03 for a slight increase in mean serum sodium levels
mOsm/kg; and 60 mg, ⫺49.51 ⫾ 152.98 mOsm/ from baseline in patients administered tolvaptan,
kg) than with placebo (168.55 ⫾ 204.2 mOsm/ 30 mg (2.77 ⫾ 3.56 mEq/L), 60 mg (3.38 ⫾ 4.84
kg; P ⬍ 0.05). mEq/L), and 90 mg (3.50 ⫾ 3.63 mEq/L), but
Of 254 patients, 28% had hyponatremia (se- a decrease in patients administered placebo
rum sodium ⬍136 mEq/L [⬍136 mmol/L]) at (⫺0.20 ⫾ 3.12 mEq/L). In addition, the 68 patients
baseline. In this subset, significantly more pa- with hyponatremia (serum sodium level ⬍136
tients showed normalized serum sodium levels at mEq/L) at baseline were reported to have a rapid
day 1 in the tolvaptan-treated group compared improvement and often normalization of serum
with placebo (80% versus 40%, respectively; sodium levels. The improvement in serum so-
P ⬍ 0.05). Correction of hyponatremia was dium levels was maintained throughout the study
maintained throughout the study period (82% in all treated groups.
versus 40%; P ⬍ 0.05). As in the previous clinical trial, no significant
Serum creatinine levels did not change in any changes in heart rate, blood pressure, serum
group by the end of the study. Thirst was re- potassium level, or renal function were observed.
ported at a significantly greater frequency in Thirst was a common adverse event, observed in
tolvaptan-treated groups compared with placebo 11.9% of patients administered tolvaptan versus
(31.3%, 40.3%, and 24.6% in the 30-, 45-, and 1.3% of the placebo group. Although there was
60-mg groups compared with 4.8% in the pla- no difference between the tolvaptan and placebo
cebo group). Nevertheless, only 2 patients from groups in the rate of worsening heart failure at 60
the 60-mg group were reported to discontinue days, post hoc analysis showed lower mortality
the medication because of polyuria. No signifi- for patients with renal dysfunction or severe
cant changes in heart rate, blood pressure, serum systemic congestion (defined as dyspnea, jugular
potassium level, or renal function were ob- venous distension, and edema) administered
served.50 tolvaptan.51
In the phase 2 clinical trial Acute and Chronic Conivaptan. The acute efficacy of intrave-
Therapeutic Impact of a Vasopressin Antagonist nous conivaptan, the first AVP-receptor antago-
in Congestive Heart Failure, 319 patients hospi- nist with activity at both the V1A and V2 recep-
talized with CHF and a left ventricular ejection tors, was evaluated in patients with New York
fraction less than 40% were assigned to placebo Heart Association classes III and IV heart failure.
(n ⫽ 80) or tolvaptan, 30 mg/d (n ⫽ 78), 60 mg/d In this randomized double-blind study, 142 pa-
(n ⫽ 84), or 90 mg/d (n ⫽ 77), plus routine tients were assigned to administration of a single
therapy, including diuretics, for as long as 60 intravenous dose of conivaptan, 10 mg (n ⫽ 37),
days.51 Median body-weight decrease from base- 20 mg (n ⫽ 32), or 40 mg (n ⫽ 35), or placebo
line associated with placebo (⫺1.90 kg; range, (n ⫽ 38).35 During the 4-hour period immedi-
⫺4.20 to ⫺0.50 kg) was significantly smaller ately after drug infusion, urine output was signifi-
than that with tolvaptan, 30 mg (⫺3.30 kg; cantly greater in patients administered conivaptan
range,⫺7.30 to ⫺1.35; P ⫽ 0.006), 60 mg (⫺2.80 (10 mg, 68.9 ⫾ 17.4 mL/h; 20 mg, 152.2 ⫾ 19.2
kg; range, ⫺5.90 to ⫺1.80; P ⫽ 0.002), and mL/h; 40 mg, 176.2 ⫾ 17.8 mL/h) than those
90 mg (⫺3.20 kg; range, ⫺5.80 to ⫺1.60; P ⫽ 0.06). administered placebo (11.2 ⫾ 17.3 mL/h; P ⬍
VASOPRESSIN EXCESS AND HYPONATREMIA 735

0.001 versus all conivaptan doses). The in- 23.4 hours in the group administered 80 mg/d. In
crease in urine output was associated with a addition, a greater percentage of patients admin-
decrease in urine osmolality: ⫺249.8 ⫾ 26.9 istered conivaptan, 40 mg/d (69%) or 80 mg/d
mOsm/kg (⫺249.8 ⫾ 26.9 mmol/kg) with (88%), than those administered placebo (21%;
conivaptan, 10 mg; ⫺279.6 ⫾ 31.7 mOsm/kg P ⬍ 0.01 and P ⬍ 0.001, respectively) showed
with conivaptan, 20 mg; ⫺319.3 ⫾ 25.8 an increase in serum sodium levels to 6 mEq/L or
mOsm/kg with conivaptan, 40 mg; and ⫺2.2 ⫾ greater or normalized serum sodium level at
27.9 mOsm/kg with placebo (P ⫽ 0.0001 versus study end. At day 1, effective water clearance
all conivaptan doses). Serum sodium concentra- also was significantly greater in patients adminis-
tions were not significantly different from pla- tered conivaptan, 40 mg/d (1,984 ⫾ 1,559 mL)
cebo at 4 hours. Nevertheless, there was a trend or 80 mg/d (1,759 ⫾ 1,748 mL), than in those
for greater serum sodium level increases in the administered placebo (⫺332 ⫾ 434 mL; P ⬍
conivaptan groups (10 mg, 0.5 ⫾ 0.5 mEq/L 0.05 versus both doses).52
[0.5 ⫾ 0.5 mmol/L]; 20 mg, 0.8 ⫾ 0.7 mEq/L; In 2 similar studies, 83 patients with eu-
and 40 mg, 1.5 ⫾ 0.4 mEq/L) compared with the volemic hyponatremia and 74 patients with hyper-
placebo group (0.4 ⫾ 0.7 mEq/L). volemic hyponatremia were administered oral
In a randomized, double-blind, multicenter, conivaptan, 40 or 80 mg/d, or placebo for 5 days.
placebo-controlled, parallel-group study, intrave- As in the other studies, conivaptan increased
nous conivaptan (a 20-mg loading dose followed serum sodium levels and effective water clear-
by continuous infusion of 40 [n ⫽ 29] or 80 mg/d ance significantly more than placebo.54,55
[n ⫽ 26] for 4 days) was compared with placebo Because in vivo and in vitro studies indicated
(n ⫽ 29) in patients with euvolemic or hypervo- that conivaptan is a substrate and potent inhibitor
lemic hyponatremia (serum sodium level, 115 to of cytochrome P-450 3A4, the liver and small-
⬍130 mEq/L [115 to ⬍130 mmol/L]).52 The intestine isoenzyme responsible for drug metab-
primary efficacy measure was change in serum olism, only the intravenous formulation of
sodium level from baseline to end of study, conivaptan was developed and approved in the
measured by the area under the sodium level– United States for the treatment of patients with
time curve. Secondary measures included euvolemic hyponatremia.
change in serum sodium level from baseline at
day 4, time from first dose to achieve a 4-mEq/L CONCLUSION
or greater change in serum sodium level, num-
ber of patients achieving a 6-mEq/L or greater Hyponatremia is a common and potentially
change in sodium level or normal sodium serious condition most often caused by excessive
level, and effective water clearance (defined as AVP secretion. With the exception of hypovole-
electrolyte-free water clearance as opposed to mic hyponatremia, for which saline infusion is
solute-free water clearance for FWC—the appropriate and the outcome generally is easily
former was suggested to more accurately ex- predictable, current therapeutic options for pa-
plain the “physiology of the renal role in the tients with euvolemic and hypervolemic hypona-
variations in natremia”).53 Changes in serum tremia may result in unpredictable or undesirable
sodium levels from baseline to end of study, outcomes. Because excessive AVP release is the
measured by the area under the sodium level– key etiologic factor in perpetuating the hypona-
time curve, were significantly greater in pa- tremia observed in patients with such common
tients administered conivaptan than those ad- clinical conditions as CHF, cirrhosis, nephrosis,
ministered placebo. Serum sodium levels and SIADH, therapy that directly antagonizes
increased significantly more with both AVP receptors potentially may offer better out-
conivaptan doses (6.8 and 9.0 mEq/L, respec- comes. Current AVP-receptor antagonists were
tively; P ⬍ 0.001) than placebo (2.0 mEq/L). shown to increase serum sodium levels effec-
Mean time required for serum sodium level to tively at a safe dose-dependent rate and may
reach 4 mEq/L or greater was not “estimable” in become a promising therapeutic option in the
the placebo group, but was 23.7 hours in the treatment of patients with euvolemic and hyper-
group administered conivaptan, 40 mg/d, and volemic hyponatremia.47
736 PHAM, PHAM, AND PHAM

REFERENCES 20. Luckey AE, Parsa CJ: Fluid and electrolytes in the
1. Adrogué HJ, Madias NE: Hyponatremia. N Engl J Med aged. Arch Surg 138:1055-1060, 2003
342:1581-1589, 2000 21. Andreoli TE: Water: Normal balance, hyponatremia,
2. Saeed BO, Beaumont D, Handley GH, Weaver JU: and hypernatremia. Ren Fail 22:711-735, 2000
Severe hyponatraemia: Investigation and management in a 22. Fried LF, Palevsky PM: Hyponatremia and hypernatre-
district general hospital. J Clin Pathol 55:893-896, 2002 mia. Med Clin North Am 81:585-609, 1997
3. Gill G, Leese G: Hyponatremia: Biochemical and 23. Robinson AG: Disorders of antidiuretic hormone se-
clinical perspectives. Postgrad Med J 74:516-523, 1998 cretion. Clin Endocrinol Metab 14:55-88, 1985
4. Tierney WM, Martin DK, Greenlee MC, Zerbe RL, 24. Robertson GL, Shelton RL, Athar S: The osmoregula-
McDonald CJ: The prognosis of hyponatremia at hospital tion of vasopressin. Kidney Int 10:25-37, 1976
admission. J Gen Intern Med 1:380-385, 1986 25. Baylis PH: Osmoregulation and control of vasopres-
5. Anderson RJ, Chung HM, Kluge R, Schrier RW: sin secretion in healthy humans. Am J Physiol 253:R671-
Hyponatremia: A prospective analysis of its epidemiology R678, 1987
and the pathogenetic role of vasopressin. Ann Intern Med 26. Goldsmith SR, Francis GS, Cowley AW Jr, Golden-
102:164-168, 1985 berg IF, Cohn JN: Hemodynamic effects of infused arginine
6. Lee CT, Guo HR, Chen JB: Hyponatremia in the vasopressin in congestive heart failure. J Am Coll Cardiol
emergency department. Am J Emerg Med 18:264-268, 2000 8:779-783, 1986
7. Bennani SL, Abouqal R, Zeggwagh AA, et al: [Inci- 27. Baylis PH: Regulation of vasopressin secretion. Bail-
dence, causes and prognostic factors of hyponatremia in lieres Clin Endocrinol Metab 3:313-330, 1989
intensive care]. Rev Med Interne 24:224-229, 2003 28. Sladek CD: Regulation of vasopressin release by
8. Terzian C, Frye EB, Piotrowski ZH: Admission hypo- neurotransmitters, neuropeptides and osmotic stimuli. Prog
natremia in the elderly: Factors influencing prognosis. J Gen Brain Res 60:71-90, 1983
Intern Med 9:89-91, 1994 29. Thibonnier M, Coles P, Thibonnier A, Shoham M:
9. Arieff AI, Llach F, Massry SG: Neurological manifes- The basic and clinical pharmacology of nonpeptide vasopres-
tations and morbidity of hyponatremia: Correlation with sin receptor antagonists. Annu Rev Pharmacol Toxicol 41:
brain water and electrolytes. Medicine (Baltimore) 55:121- 175-202, 2001
129, 1976 30. Lee CR, Watkins ML, Patterson JH, et al: Vasopres-
10. Goldberg A, Hammerman H, Petcherski S, et al: sin: A new target for the treatment of heart failure. Am
Prognostic importance of hyponatremia in acute ST-eleva- Heart J 146:9-18, 2003
tion myocardial infarction. Am J Med 117:242-248, 2004 31. Costello-Boerrigter LC, Boerrigter G, Burnett JC Jr:
11. Ruf AE, Kremers WK, Chavez LL, Descalzi VI, Revisiting salt and water retention: New diuretics, aquaret-
Podesta LG, Villamil FG: Addition of serum sodium into the ics, and natriuretics. Med Clin North Am 87:475-491, 2003
MELD score predicts waiting list mortality better than
32. Skott O: Body sodium and volume homeostasis. Am J
MELD alone. Liver Transpl 11:336-343, 2005
Physiol Regul Integr Comp Physiol 285:R14-R18, 2003
12. Borroni G, Maggi A, Sangiovanni A, Cazzaniga M,
33. Thunhorst RL, Johnson AK: Effects of hypotension
Salerno F: Clinical relevance of hyponatraemia for the
and fluid depletion on central angiotensin-induced thirst and
hospital outcome of cirrhotic patients. Dig Liver Dis 32:605-
salt appetite. Am J Physiol Regul Integr Comp Physiol
610, 2000
281:R1726-R1733, 2001
13. Baylis PH: The syndrome of inappropriate antidi-
34. Striker EM, Callahan JB, Huang W, Sved AF: Early
uretic hormone secretion. Int J Biochem Cell Biol 35:1495-
osmoregulatory stimulation of neurohypophysial hormone
1499, 2003
14. Verbalis JG: Disorders of body water homeostasis. secretion and thirst after gastric NaCl loads. Am J Physiol
Best Pract Res Clin Endocrinol Metab 17:471-503, 2003 Regul Integr Comp Physiol 282:R1710-R1717, 2002
15. Davison JM, Vallotton MB, Lindheimer MD: Plasma 35. Udelson JE, Smith WB, Hendrix GH, et al: Acute
osmolality and urinary concentration and dilution during hemodynamic effects of conivaptan, a dual V1A and V2 vaso-
and after pregnancy: Evidence that lateral recumbency inhib- pressin receptor antagonist, in patients with advanced heart
its maximal urinary concentrating ability. Br J Obstet Gynae- failure. Circulation 104:2417-2423, 2001
col 88:472-479, 1981 36. Cardenas A, Arroyo V: Mechanisms of water and
16. Ishikawa SE, Schrier RW: Pathophysiological roles sodium retention in cirrhosis and the pathogenesis of ascites.
of arginine vasopressin and aquaporin-2 in impaired water Best Pract Res Clin Endocrinol Metab 17:607-622, 2003
excretion. Clin Endocrinol 58:1-17, 2003 37. Gines P, Berl T, Bernardi M, et al: Hyponatremia in
17. Robinson AG, Roberts MM, Evron WA, Verbalis JG, cirrhosis: From pathogenesis to treatment. Hepatology 28:
Sherman TG: Hyponatremia in rats induces downregulation 851-864, 1998
of vasopressin synthesis. J Clin Invest 86:1023-1029, 1990 38. Anderson RJ, Berl T, McDonald KD, Schrier RW:
18. Robertson GL: Physiology of ADH secretion. Kidney Evidence for an in vivo antagonism between vasopressin
Int Suppl 21:S20-S26, 1987 and prostaglandin in the mammalian kidney. J Clin Invest
19. Dunn FL, Brennan TJ, Nelson AE, Robertson GL: 56:420-426, 1975
The role of blood osmolality and volume in regulating 39. Breyer MD, Jacobson HR, Hebert RL: Cellular mecha-
vasopressin secretion in the rat. J Clin Invest 52:3212-3219, nisms of prostaglandin E2 and vasopressin interactions in
1973 the collecting duct. Kidney Int 38:618-624, 1990
VASOPRESSIN EXCESS AND HYPONATREMIA 737

40. Zerbe R, Stropes L, Robertson G: Vasopressin func- 49. Wong F, Blei AT, Blendis LM, Thuluvath PJ: A
tion in the syndrome of inappropriate antidiuresis. Annu Rev vasopressin receptor antagonist (VPA-985) improves serum
Med 31:315-327, 1980 sodium concentration in patients with hyponatremia: A mul-
41. Bartter FC, Schwartz WB: The syndrome of inappro- ticenter, randomized, placebo-controlled trial. Hepatology
priate secretion of antidiuretic hormone. Am J Med 42:790- 37:182-191, 2003
806, 1967 50. Gheorghiade M, Niazi I, Ouyang J, et al: Vasopressin
42. Fraser CL, Arieff AI: Epidemiology, pathophysiol- V2-receptor blockade with tolvaptan in patients with chronic
ogy, and management of hyponatremic encephalopathy. Am J heart failure: Results from a double-blind, randomized trial.
Med 102:67-77, 1997 Circulation 107:2690-2696, 2003
43. Beck LH: The aging kidney. Defending a delicate 51. Gheorghiade M, Gattis WA, O’Connor CM, et al:
balance of fluid and electrolytes. Geriatrics 55:26-28, 31-22, Effects of tolvaptan, a vasopressin antagonist, in patients
2000 hospitalized with worsening heart failure: A randomized
44. Phillips PA, Rolls BJ, Ledingham JGG, et al: Re- controlled trial. JAMA 291:1963-1971, 2004
duced thirst after water deprivation in healthy elderly men. 52. Verbalis JG, Bisaha JG, Smith N: Novel vasopressin
N Engl J Med 311:753-759, 1984 V1A and V2 antagonist conivaptan increases serum sodium
45. Mulkerrin E, Epstein FH, Clark BA: Aldosterone concentration and effective water clearance in hyponatre-
responses to hyperkalemia in healthy elderly humans. J Am mia. J Am Soc Nephrol 15:356A, 2004 (abstr SA-P0254)
Soc Nephrol 6:1459-1462, 1995 53. Mallie JP, Bichet DG, Halperin ML: Effective water
46. Miller PD, Linas SL, Schrier RW: Plasma demeclocy- clearance and tonicity balance: The excretion of water revis-
cline levels and nephrotoxicity. Correlation in hyponatremic ited. Clin Invest Med 20:16-24, 1997
cirrhotic patients. JAMA 243:2513-2515, 1980 54. Ghali JK, Bisaha JG, Smith N: Efficacy of the vaso-
47. Serradeil-Le Gal C, Wagnon J, Valette G, et al: Nonpep- pressin V1A/V2 antagonist conivaptan in patients with euv-
tide vasopressin receptor antagonists: Development of selective olemic or hypervolemic hyponatremia. J Am Soc Nephrol
and orally active V1A, V2 and V1B receptor ligands. Prog Brain 15:355A, 2004 (abstr SA-P0251)
Res 139:197-210, 2002 55. Gross P, Bisaha JG, Smith N: Conivaptan, a novel
48. Gerbes AL, Gulberg V, Gines P, et al: Therapy of V1A and V2 antagonist, increased serum sodium and effec-
hyponatremia in cirrhosis with a vasopressin receptor antag- tive water clearance in patients with euvolemic or hypervo-
onist: A randomized double-blind multicenter trial. Gastroen- lemic hyponatremia. J Am Soc Nephrol 15:355A, 2004
terology 124:933-939, 2003 (abstr SA-P0243)

Você também pode gostar