You are on page 1of 10

Heart Fail Rev (2012) 17:1726

DOI 10.1007/s10741-010-9193-3

Significance of hyponatremia in heart failure


Luca Bettari Mona Fiuzat Gary M. Felker

Christopher M. OConnor

Published online: 14 September 2010


Springer Science+Business Media, LLC 2010

Abstract Heart failure is one of the most common, within several aspects: incidence in clinical trials and
costly, disabling and growing diseases (McMurray and registries, prognostic value, underlying mechanisms, ther-
Pfeffer in Lancet 365(9474):18771889, 2005). Hypona- apeutic options, and possible future perspectives.
tremia, conventionally defined as a serum-sodium con-
centration equal or less than 135 mmol/l (American Heart Keywords Heart failure  Hyponatremia  Clinical trials 
Association in Heart disease and stroke statistics2007 Outcomes  Arginine vasopressin antagonists
update. American Heart Association, Dallas, 2007; Stewart
et al. in Eur J Heart Fail 4:361371, 2002), is a common
phenomenon in patients with heart failure, with an inci- Introduction
dence of 2025% (Krumholz et al. in Arch Intern Med
157:e99e104, 1997; Rosamond et al. in Circulation Heart failure (HF) is one of the most common, costly,
117(4):e25e146, 2008; Adrogue and Madias in N Engl J disabling and growing diseases [1]. About 5 millions of
Med 342:15811589, 2000) and seems to be of prognostic Americans are affected by HF, and more than 5,00,000 new
importance in patients with heart failure (Luca et al. in Am cases are diagnosed each year [2]. HF mortality rate still
J Cardiol 96:19L23L, 2005; Gheorghiade et al. in Eur high, over 60% within 5 years of diagnosis [3], and within
Heart J 28:980988, 2007; Gheorghiade et al. in Arch 6 months of discharge 50% of patients require readmission
Intern Med 167:19982005, 2007). So far treatment strat- [4]. The estimated costs in the United States amount to
egies have been limited and burdened by side effects. The more than $35 billion [5].
development of hyponatremia in the setting of heart failure Hyponatremia (conventionally defined as a serum-
is related to the arginine vasopressin (AVP) dysregulation. sodium concentration [Na] B135 mmol/l) [6, 7], is a
Thus, AVP receptor antagonists are a promising approach common phenomenon in HF patients, with an incidence of
to treatment. However, several questions remain: whether 2025% [810], and it is known to be associated with
there is a cause-and-effect mechanism, if the correction of increased morbidity and mortality in these patients
hyponatremia improves outcomes, and defining the specific [1113]. So far treatment strategies have been limited and
cut-off level of serum-sodium that should be used to define burdened by side effects. The development of hyponatremia
hyponatremia. In this review, we aim to summarize the in the setting of HF is related to the arginine vasopressin
literature on hyponatremia in patients with heart failure (AVP) dysregulation. Thus, AVP receptor antagonists are a
promising approach to treatment.
Hyponatremia seems to be of prognostic importance in
L. Bettari (&)
University of the Studies of Brescia, Piazzale Spedali Civili 1, HF patients. However, several questions remain: whether
Brescia, Italy there is a cause-and-effect mechanism, whether the cor-
e-mail: luca_bettari@yahoo.it rection of hyponatremia improves outcomes, and defining
the specific cut-off level of serum-sodium ([Na]) that
M. Fiuzat  G. M. Felker  C. M. OConnor
Duke University Medical Center, Duke Clinical Research should be used to define hyponatremia. In this review,
Institute, 2400 Pratt street, Durham, NC, USA we aim to summarize the literature on hyponatremia in HF

123
18 Heart Fail Rev (2012) 17:1726

patients within several aspects: incidence in clinical trials IV, LVEF B40%), 8% of the patients had hyponatremia at
and registries, prognostic value, underlying mechanisms, admission ([Na] \134 mEq/l). No mortality outcome
therapeutic options, and possible future perspectives. benefit was seen with treatment in this subgroup, and there
was no association of improved hyponatremia with better
outcomes [18].
Scope of the problem The secondary analyses of RCTs evaluating hyponatre-
mia in HF are summarized in Table 1.
Published estimates of the prevalence of hyponatremia in
HF patients vary according to the [Na] value used in the
different studies [14]. Moreover, the incidence and out- Hyponatremia in prognostic models
comes vary according to the severity of HF [11].
In the Acute Decompensated HEart failure REgistry Risk assessment based on any single factor has limited
(ADHERE) of 1,58,168 HF patients, hyponatremia ([Na] accuracy and reproducibility. By combining individual
\130 mmol/l) was found on admission in only 5% of the clinical characteristics into a multivariable predictive
patients and was considered to be a risk factor for mortality index, frequently discordant implications of univariable
and morbidity [15, 16]. analyses may be made meaningful. Prognostic models are
In the Outcomes of a Prospective Trial of Intravenous important tools that can be used to estimate outcomes,
Milrinone for Exacerbation of Chronic Heart Failure enhance compliance and increase the use of life-saving
(OPTIME-CHF) Study of 949 patients hospitalized for medications and devices.
worsening HF, hyponatremia ([Na] \135 mmol/l) was Hyponatremia has been found to be an important pre-
present on admission in 27% of the patients, and it was an dictor of survival in several risk models in HF patients.
important predictor of increased number of days of hos- These models have been developed in different HF cohorts:
pitalization for cardiovascular causes and mortality within ambulatory patients, hospitalized patients, and cardiac
60 days of discharge (P = 0.018) [10]. transplant patients, and have been used to predict both
Likewise in the Acute and Chronic Therapeutic Impact of short-term and long-term outcomes.
a Vasopressin Antagonist in Congestive Heart Failure The Heart Failure Survival Score (HFSS) is the first
(ACTIVinCHF) trial of 319 patients with left ventricular predictive model in HF developed (1997) [19]. It was
ejection fraction (LVEF)\40% and hospitalized for HF with developed in a prospective study for pretransplant risk
persistent signs and symptoms of systemic congestion stratification in ambulatory patients (age B70 years) with
despite standard therapy, hyponatremia ([Na]\136 mmol/l) advanced HF (LVEF B40%) referred for cardiac transplant
was found on admission in 21% of patients. However, no evaluation. A predictive model based on 268 patients
differences in worsening HF at 60 days between the groups (derivation sample) was generated, obtaining 7 significant
were observed [17]. variables after stabilization with maximal medical therapy:
In the Evaluation Study of Congestive Heart Failure and ischemic cardiomyopathy, resting heart rate (HR), LVEF,
Pulmonary Artery Effectiveness (ESCAPE) trial of 433 HF QRS duration C0.12 s, mean resting blood pressure (BP),
patients (NYHA IV, LVEF \30%), 23.8% of patients had peak VO2, and [Na] (for patients with vs. patients without
hyponatremia on admission ([Na] B134 mmol/l). Notably, the characteristic for dichotomous variables or per unit
68.9% of these patients had persistent hyponatremia, which increase for continuous variables). Between the variables,
was found to be an independent predictor of mortality, HF the ischemic etiology of HF was found to be the strongest
hospitalization, and death or rehospitalization at 6 months predictor (HR 2.00, 95% CI 1.352.97), while [Na] was the
(P = 0.01) [9]. weakest, associated with a 5% increased mortality risk (per
In the Organized Program to Initiate Lifesaving Treat- 1U decrease) (HR 0.95, 95% CI 0.921.00).
ment in Hospitalized Patients with Heart Failure (OPTI- The EFFECT model is another clinical model that was
MIZE-HF) registry, which included 48,162 HF patients, developed to identify predictors of mortality using infor-
19.7% of these patients had hyponatremia on admission mation available at hospital presentation. In this retrospec-
([Na] \135 mmol/l). It was associated with longer hospital tive study of 4,031 patients presenting to the hospital with
stays and higher in-hospital and early (6090 days) post- HF, the main outcome measures were all-cause 30-day and
discharge mortality (P \ 0.0001); however, no difference 1-year mortality. The multivariable predictors of mortality
was found in the readmission rates. Notably, the risk of at both 30 days and 1 year were found to be: older age (per
mortality significantly increased with [Na]\138 mmol/l [8]. 10U increase), lower systolic BP (per 10U decrease), higher
In a subgroup analysis of the Efficacy of Vasopressin respiratory rate (per 5U increase), higher blood urea nitrogen
Antagonism in Heart Failure Outcome Study With Tol- levels (per 10U increase), and baseline hyponatremia.
vaptan (EVEREST) trial of 4,133 HF patients (NYHA III/ Hyponatremia was defined as [Na] \136 mEq/l, and it was

123
Heart Fail Rev (2012) 17:1726 19

Table 1 Secondary analyses of randomized clinical trials evaluating hyponatremia in heart failure
RCTs Number Hyponatremia on admission Results
of patients

OPTIME-CHF [10] 949 [Na] \135 mmol/l (27% of the Lowest [Na] quartile associated with more
(exacerbation of HF, patients) days hospitalized for CV causes
LVEF \40%, not (P \ 0.015) ? trend toward higher
requiring iv inotrope mortality/rehospitalization
support) Lower [Na] associated with higher in-
hospital and 60-day mortality (P \ 0.015,
P = 0.002, respectively)
Baseline [Na] predicted increased 60-day
mortality (for 3 mEq/l decrease, HR 1.18)
ACTIVinCHF [17] 319 [Na] \136 mmol/l (21.6% of the Baseline hyponatremia: significant predictor
(persistent HF, LVEF patients) of 60-day mortality (P = 0.0016)
\40%) Change in [Na]: significant predictor of
60-day mortality (for 1 mmol/l increase,
HR 0.736, P = 0.018)
ESCAPE [9] (persistent HF, 433 [Na] B134 mmol/l (23.8% of the Hyponatremia associated with higher
LVEF \30%, NYHA IV) patients) 6-month mortality (for each 3 mEq/l
decrease, HR 1.23, P = 0.01)
Persistent hyponatremia (68.9% of Persistent hyponatremia associated with
the patients) increased risk of all-cause mortality, HF
rehospitalization, and death/
rehospitalization (HR 1.82, 1.52, 1.54;
P = 0.04, 0.03, 0.01 respectively)

significantly associated with increased risk of 30-day mor- associated with [Na] \138 mEq/l (HR 1.050, 95% CI
tality (HR 1.53, 95% CI 1.142.05) and 1-year mortality 1.0051.097). Subsequently, 3 other variables were forced
(HR 1.46, 95% CI 1.191.80) [20]. into the model: age and gender as important demographic
Subsequently, a prognostic index for long-term mortal- variables, and potassium-sparing diuretics [22].
ity (over a 5-year period) in patients with mild to moderate A model to predict outcomes in patients with decom-
chronic HF (CHF) was developed (Area Under the Curve pensated HF was developed from the OPTIME-
[AUC] 0.74, 95% CI 0.700.78) [21]. Five hundred and CHF study. The variables at admission that predicted death
fifty-three HF outpatients from the UK-HEART study were at 60 days were found to be increasing age (per 10U
enrolled. Eight non-invasive prognostic measurements increase), lower systolic BP (per 10U decrease), NYHA
were identified to be significantly associated with long- class IV symptoms, elevated BUN (per 5U increase) and
term mortality: baseline hyponatremia ([Na] B140 mmol/ lower [Na], which resulted to be associated with a 25%
l), serum-creatinine, cardiothoracic ratio, standard devia- increase in mortality risk each 5U decrease (HR 0.75, 95%
tion of normal-to-normal RR intervals, maximum corrected CI 0.600.95) [23].
QT, QRS dispersion, presence of non-sustained ventricular The MUSIC model was developed to predict mortality
tachycardia (NSVT) and voltage criteria for LV hypertro- in an independent, non-clinical trial, outpatient CHF pop-
phy on 12-lead ECG. In this model, baseline hyponatremia ulation easily assessable in clinical practice. The MUerte
was associated with an increased mortality risk of 13% Subita en Insuficiencia Cardiaca (MUSIC) study was a
(HR 1.13, 95% CI 1.051.19). prospective, multicentre, longitudinal study designed to
The Seattle Heart Failure Model was derived in a cohort assess risk predictors of cardiac mortality and sudden
of 1,125 HF patients and validated in 5 additional cohorts cardiac death (SCD) in either systolic dysfunction (75%) or
totaling 9,942 HF patients. Eleven prognostic variables were preserved EF (25%) ambulatory HF patients. Four final
identified as predictors of increased mortality: the calculated models (total mortality, cardiac mortality, pump failure
daily diuretic dose per kg of body weight (BW), NYHA death (PFD), SCD (c-indices of 0.76, 0.78, 0.80, and 0.77
class, ischemic etiology, 1/x transformation of EF, choles- respectively) were developed including several combina-
terol, hemoglobin (\ or [16 g/dl), lymphocytes ([47%), tions of the same discovered 10 independent predictors:
uric acid (\3.4 mg/dl), systolic BP ([160 mmHg), statin prior atherosclerotic vascular event, left atrial size
use, and baseline [Na]. There was increased mortality risk ([26 mm/m2), EF (B35%), AF, left bundle branch block

123
20 Heart Fail Rev (2012) 17:1726

(LBBB) or intraventricular conduction delay, NSVT and Classification and pathophysiology


frequent ventricular premature beats, estimated glomerular
filtration rate (eGFR \60 ml/min/1.73 m2), hyponatremia Hyponatremia results from an excess of body water com-
([Na] B138 mEq/l), NT-proBNP ([1,000 ng/l), and pared to serum-sodium. It can be classified into 3 categories:
positive troponin (values greater than the 99th reference hypertonic, in which osmotically active non-electrolyte
percentile). On the basis of Cox models, the MUSIC Risk solutes accumulate (e.g., glucose); isotonic, due to labora-
scores were calculated. A cardiac mortality score [20 tory artifact (e.g., hyperproteinemia, hyperlipidemia,
points identified a high-risk subgroup with a fourfold hyperglycemia); hypotonic, which can either be hypovole-
increased cardiac mortality risk. Hyponatremia was not mic caused by the depletion of water and sodium (e.g.,
associated with SCD mortality, but significantly associ- diarrhea, diuretics) or euvolemic caused by water retention
ated with total mortality (3 points), cardiac mortality (3 (e.g., syndrome of inappropriate secretion of antidiuretic
pointsHR 1.38, 95% CI 1.031.77) and particularly hormone [SIADH]) [26]; hypervolemic, in which the
PFD mortality (4 pointsHR 1.60, 95% CI 1.122.29) increase in body water overtakes that of sodium (e.g., HF,
[24]. cirrhosis) [6].
At last, the ESCAPE risk model and discharge score was The pathogenesis of hyponatremia in HF is multifacto-
derived from the namesake study, which examined the rial: the reduction of cardiac output (CO) decreases renal
efficacy of pulmonary artery catheter (PAC)-guided ther- perfusion and GFR and enhances the reabsorption of
apy versus clinical assessment on the resolution of symp- sodium and water compromising their delivery to the distal
toms and signs of congestion, with days alive out of the diluting segment of the nephron [11]. The increase in AVP
hospital during the first 6 months as primary endpoint. In binding to the vasopressin-2 receptor (V2R) increases the
this study, the patients discharged with complete data number of aquaporin-2 water channels, enhancing free-
(n = 423) had 6-month mortality and combined death and water retention [27, 28]. Increased sympathetic nervous
rehospitalization rates of 18.7 and 64%, respectively. system (SNS) activity [29] causes renal vasoconstriction,
Discharge risk factors significantly associated with mor- decreasing GFR [30] and stimulates the reninangiotensin
tality were found to be BNP per doubling, cardiopulmonary aldosterone system (RAAS), increasing sodium and water
resuscitation or mechanical ventilation during hospitaliza- retention, thirst center activity and release of AVP [31, 32].
tion, BUN per 20U increase, serum-sodium per 1U Hyponatremia may also result from diuretic therapy [11,
increase, age [70 years, daily loop diuretic furosemide 14] with thiazides due to increased sodium and potassium
equivalents[240 mg, lack of beta-blocker, and 6-min walk excretion in a setting of AVP-enhanced water reabsorption,
per 100-foot increase. The c-index for the model was 0.76. resulting in the excretion of hypertonic urine. Loop
Discharge serum-sodium was found to be an independent diuretics induce isotonic diuresis with salt loss, which in
predictor of mortality, associated with a 7% decreased risk turn can worsen hyponatremia [14, 33].
(per 1U increase) of all-cause death (HR 0.93, 95% CI The mechanisms leading to hypotonic hypervolemic
0.870.99). Baseline [Na] was significantly different hyponatremia in HF are summarized in Table 3.
between those who died or survived (P = 0.001). A Sim-
plified Discharge Score Model of Mortality was developed AVP: role in HF
(c-index 0.739), and in this model hyponatremia at dis-
charge ([Na] \130 mEq/l) was found to be the 4th most AVP dysregulation is the most common cause of hypotonic
important predictor out of 8 variables. The simplified dis- hypervolemic hyponatremia [34]. AVP is a non-peptide
charge score discriminated mortality risk from 5% (lowest hormone synthesized and regulated by 2 mechanisms:
score) to 94% (highest score). This discharge risk model osmotical (increase in serum osmolality increases AVP
allows for risk assessment after in-hospital therapy for release) and non-osmotical (decrease in plasma circulating
advanced decompensated systolic HF [25]. volume increases AVP release). In HF patients, AVP pro-
Thus, hyponatremia has been demonstrated to be an duction is increased, and AVP levels do not fall to the
important prognostic predictor of mortality in HF patients, expected levels but often remain elevated despite the
ranging from hospitalized patients to ambulatory patients presence of volume overload, atrial distension, hyponatre-
and from short to long-term mortality. However, there is mia, and low plasma osmolality [35, 36].
no consistency between these models on the value of AVP has 3 different receptor subtypes. V1A receptors,
serum-sodium level to use, or the incremental value of found on vascular smooth muscle and cardiac myocytes,
hyponatremia as a predictor compared to other prognostic may cause vasoconstriction, positive inotropic effect, and
markers. increase in the synthesis of contractile protein in myocytes,
The HF prognostic models in which hyponatremia is potentially leading to increased afterload, left ventricular
included are summarized in Table 2. hypertrophy and remodeling [3739]. V2 receptors, on the

123
Heart Fail Rev (2012) 17:1726

Table 2 Hyponatremia in HF prognostic models


Models Number Cohort of patients Model endpoints [Na] value cutoff Significance of C-Index
of patients hyponatremia in model
(HR, 95% CI)

HFSS 268 Outpatients with systolic HF 1-year mortality [Na] per 1 mEq/l change 1.05, 1.001.08 0.74
(NYHA IIIIV) (value after medical
therapy optimization)
EFFECT 2,624 Hospitalized patients with 30-day and 1-year mortality [Na] \136 mEq/l (baseline 1.53, 1.142.05 (for 30-day 0.77
systolic and diastolic HF value) mortality) 1.46, 1.191.80
(NYHA IIIII) (for 1-year mortality)
UK-HEART 553 Outpatients with systolic HF 5-year mortality [Na] per 2 mmol/l change 1.13, 1.051.19 0.78
(NYHA IIIII) [Na] B140 mmol/l
(baseline value)
Seattle heart 1,125 Hospitalized patients with 1-, 2-, 3-year mortality [Na] \138 mEq/l (baseline 1.05, 1.0051.097 0.73
failure model systolic HF (NYHA IIIV) value)
MUSIC 992 Outpatients with systolic and 4-year mortality for total, [Na] B138 mEq/l (baseline 1.35, 1.031.77 (for cardiac 0.76
diastolic HF (NYHA IIIII) cardiac, PFD mortality value) mortality) 1.60, 1.122.29 0.78
(for PFD mortality)
0.80
ESCAPE 423 Hospitalized patients with 6-month mortality [Na] per 1U change [Na] 0.93, 0.870.99 0.74
systolic HF (NYHA IIIIV) \130 mEq/l (discharge
value)
OPTIME-CHF 949 Hospitalized patients with 60-day mortality [Na] per 5 mmol/l change 0.75, 0.600.95 0.77
systolic HF (NYHA IIIIV) (baseline value)
21

123
22 Heart Fail Rev (2012) 17:1726

Table 3 Mechanisms leading to hypotonic hypervolemic hyponatremia in heart failure


Direct hemodynamic effects Chronic neurohormonal activation

Reduction of CO SNS
Reduction of GF pressure ? reduction of GFR Peripheral and renal vasoconstriction ? decrease of renal blood flow and
Reabsorption of sodium and water in the proximal GFR ? decrease of the rate of solute and water delivery to the distal
tubule ? reduction of the delivery to the distal diluting segment of diluting segment of the nephron
the nephron Excretion of renin
Reduction of atrial-renal reflexes SRAA
The increase in left atrial pressure does not decrease anymore the Sodium and water reabsorption in the proximal tubule
release of AVP and the renal adrenergic tone Increase in the efferent arteriolar tone ? enhancing the
GFR ? promoting sodium and water absorption
Activation of the thirst center
Release of AVP
ANP
Renal sodium loss (direct effect ? inhibition of aldosterone release)
AVP
Increase the expression of aquaporin-2 water channels ? enhancing free-
water retention

renal collecting ducts, may cause free-water reabsorption, urea is linked with toxicity making these options
which leads to increased water retention, volume expansion unpleasant. Diuretic therapy can be associated with wors-
and potentially hyponatremia [40]. Finally, V1B receptors, ening of hyponatremia. All these treatment may not be
located in the anterior pituitary gland, determine ACTH effective in all patients and can result in significant adverse
and b-endorphin release. This subtype does not seem to be events [6].
related to HF, however, recent studies have demonstrated AVP secretion is the leading mechanism of hyponatre-
that AVP is involved in the regulation of glucose homeo- mia in HF. So far there are no therapies aimed to reduce
stasis through V1A and V1B receptors. Indeed V1A receptor AVP production. The blockade of AVP receptors is a
deficiency results in decreased insulin sensitivity, whereas rational therapeutic approach [49, 50]. The antagonism of
V1B receptor deficiency results in increased insulin sensi- V2 receptors induces the elimination of free-water
tivity [41]. (aquaresis) without depleting electrolytes or activating
AVP levels are increased in HF patients [4244] neurohormonal systems, common side effects of diuretic
and seem to correlate with severity and adverse outcome therapy [51]. Furthermore, the antagonism of V1A/V2
[4547]; however, a cause-and-effect relationship between receptors provides aquaresis induced by the V2 receptor
inappropriate AVP levels and HF progression has not yet stimulation and should suppress the deleterious cardiac
been proven. Due to the number of different mechanisms effects caused by the activation of the V1A receptor
that may underlie the pathophysiology of hyponatremia, [49, 52]. Experimental models and clinical trials have
blocking either or both the V1A and V2 receptors may be shown that AVP receptor antagonists represent a promising
useful. Evidence is needed that interfering with hormone approach for the treatment of hyponatremia in HF [53, 54].
secretion or effect has a clinically important benefit. So far 3 vasopressin antagonists have been studied for the
treatment of hyponatremia in HF: conivaptan (V1A/V2
receptor antagonist), lixivaptan, and tolvaptan (both
Treatment and AVP antagonism selective V2 antagonists) [55]. Of these, conivaptan is
approved as intravenous formulation for the treatment of
Treatment options are limited. Dietary sodium restriction euvolemic and hypervolemic hyponatremia (for up to
and diuretic therapy are the mainstays [48]. The purpose is 4 days) [56]; tolvaptan is approved as an oral formulation
to generate a negative solute-free water balance, thus fluid for euvolemic and hypervolemic hyponatremia [57]; while
restriction to 1 l/day is recommended. However, it induces lixivaptan is in phase 3 evaluation [58].
thirst and therefore may have lack of compliance. Another Hyponatremia seems to be effectively managed by the
possible treatment is the use of intravenous (iv) hypertonic vasopressin receptor antagonists, but it is not yet clear
saline infusion; however, this may cause worsening of whether normalization of [Na] leads to an improved
congestion symptoms. The use of demeclocycline and/or prognosis.

123
Heart Fail Rev (2012) 17:1726 23

RCTs on AVP receptors antagonists patients given tolvaptan, compared with 40% of those given
placebo (P \ 0.05); a similar percentage of patients main-
V1A/V2 receptor antagonism tained normal [Na] throughout the study. Plasma AVP levels
tended to be higher in patients given tolvaptan, but this
The only V1A/V2 receptor antagonist available is finding was not significant [62].
conivaptan. It has been evaluated in phase 3, double-blin- The ACTIVinCHF trial was a phase 2, dose-ranging,
ded, placebo-controlled, multicenter RCTs in patients with randomized, double-blinded, placebo-controlled trial
euvolemic or hypervolemic hyponatremia. designed to evaluate the efficacy and safety of oral tolvaptan
Data from 3 RCTs [5961] were combined and analyzed (30, 60, or 90 mg) in 319 patients hospitalized with acute
to determine the efficacy of conivaptan in the subset of decompensated HF (ADHF) (LVEF \40%) for no longer
patients with hyponatremia and HF. Of the 241 enrolled than 10 days while hospitalized, and continued for up to
patients, 94 patients (39%) had HF (placebo31, conivap- 60 days on an outpatient basis. Treatment with tolvaptan at
tan 40 mg/day25, conivaptan 80 mg/day28 patients, all 3 doses significantly reduced median BW at 24 h com-
respectively). Compared with placebo, [Na] increased sig- pared to placebo, but had no effect on HF outcomes at
nificantly from baseline after iv and oral administration of 60 days (P = 0.002, P = 0.002, and P = 0.009, respec-
conivaptan 40 and 80 mg/day. Notably, there was a slower tively). In a post hoc analysis, tolvaptan reduced 60-day
[Na] rate of correction in patients with HF compared to those mortality in patients with renal dysfunction or severe sys-
without HF. Adverse events were similar between groups, temic congestion. 21% of the patients had hyponatremia
indicating conivaptan was well tolerated in HF patients. ([Na] \136 mEq/l) at baseline. Tolvaptan produced a rapid
Conivaptan has been approved as intravenous adminis- increase in [Na], and often a return to normal [Na], that was
tration by the Food and Drug Administration (FDA) for the maintained throughout the study (P \ 0.001). However, no
treatment of euvolemic and hypervolemic hyponatremia. differences in worsening HF (WHF) at 60 days between the
It should be initiated with a loading dose of 20 mg/100 ml groups were observed [17].
administered over 30 min, followed by a continuous infu- The EVEREST programs are multicenter, randomized,
sion of 20 mg/100 ml/24 h. If [Na] is not rising at the double-blinded, placebo-controlled trials (2 short-term and 1
desired rate, conivaptan may be titrated upward to a dose of long-term outcome studies) that evaluated the effects of
40 mg/day. The total duration of infusion (after the loading Tolvaptan among 4,133 patients admitted for ADHF (LVEF
dose) should not exceed 4 days. Moreover, patients B40%). Patients were randomized 1:1 to tolvaptan 30 mg/
receiving conivaptan must have frequent monitoring of day or placebo for a minimum of 60 days. Tolvaptan
[Na] and volume status. An overly rapid rise in [Na] determined a statistically significant improvement com-
([12 mEq/l/day) should be avoided due to possible neu- pared to placebo on the primary short-term endpoint (com-
rologic sequelae [56]. posite of patient-assessed global clinical status and BW
change) at day 7 (P = 0.0004) or discharge (P \ 0.0001)
V2 receptor antagonism [63]. However, in the long-term follow-up (FU) trial during
a mean FU of 9.9 months, tolvaptan had no effect on all-
There are 2 available V2 receptor antagonists, tolvaptan, cause mortality or the combined endpoint of cardiovascular
which has already been approved, and lixivaptan, which is mortality, or subsequent hospitalization for WHF. Adverse
still under investigation. events resulting in study drug discontinuation occurred in
The efficacy and safety of tolvaptan were evaluated in a 6.5% of tolvaptan patients. Notably, those taking tolvaptan
randomized, double-blinded, placebo-controlled study that had reductions in BW and improved [Na] with modest
enrolled 254 HF patients (NYHA Class I to III). The patients improvements in signs and symptoms of HF with no excess
were randomly assigned to tolvaptan (30, 45, or 60 mg/day) adverse events. In a subgroup analysis, *8% of patients had
or placebo for 25 days, along with stable dose of furosemide. hyponatremia ([Na] \134 mEq/l); in these patients, tol-
Tolvaptan reduced mean BW significantly more than pla- vaptan exerted a significant effect in increasing [Na] com-
cebo, regardless of the dosage given (P \ 0.001). However, pared to placebo. However, as in the larger cohort, there was
there was no further reduction beyond the first day; thus, it no associated decrease in mortality related to improvement
was largely due to fluid loss at initiation of therapy. Urine in [Na] [18]. Tolvaptan was well tolerated in these studies.
volume was also significantly greater and urinary osmolality Tolvaptan has been approved as oral administration for
significantly lower with tolvaptan than with placebo the treatment of euvolemic and hypervolemic hyponatre-
(P \ 0.05). Urinary sodium excretion also increased sig- mia. The usual starting dose for tolvaptan is 15 mg/day
nificantly during the first day of tolvaptan treatment. In 28% which can be increased to 30 mg/day, after at least 24 h, to
of the patients who had hyponatremia ([Na]\136 mEq/l) at a maximum of 60 mg/day, if [Na] does not rise. During
baseline, [Na] normalized on day 1 of treatment in 80% of initiation and titration, changes in serum electrolytes and

123
24 Heart Fail Rev (2012) 17:1726

volume must be frequently monitored. Fluid restriction with these agents, and it failed to demonstrate a benefit
during the first 24 h of therapy must be avoided. An overly despite the correction of hyponatremia in a large random-
rapid rise in [Na] ([12 mEq/l/day) should be avoided due ized clinical trial [63]. Moreover, the cost of these new
to possible neurologic sequelae [57]. therapies, associated with the lack of positive results on
Lixivaptan has been evaluated in a randomized, double- mortality and rehospitalization endpoints, makes the cost-
blinded, placebo-controlled, ascending single-dose (oral effectiveness controversial. Future research should focus
lixivaptan 10, 30, 75, 150, 250, or 400 mg) trial on 42 on a better understanding of the cause-and-effect mecha-
HF patients (NYHA II/III) and hyponatremia (120\ [Na] nism existing between HF and hyponatremia, to further
\140 mEq/l), all but the 10-mg dose demonstrated a sig- analyze whether the correction of hyponatremia improves
nificant and dose-dependent increase in urine output (P \ long-term outcomes and to determine a consistent defini-
0.01), solute-free water clearance (P \ 0.05), and [Na] at tion of serum-sodium level that should be used to define
higher doses (250400 mg) (P \ 0.01) compared with pla- hyponatremia.
cebo. Lixivaptan demonstrated significant dose-dependent
decreases in urinary osmolality, but did not demonstrate any
significant effects on levels of plasma renin, norepinephrine, References
aldosterone, ANP and endothelin-1 [64].
The Treatment of Hyponatremia Based on Lixivaptan in 1. McMurray JJ, Pfeffer MA (2005) Heart failure. Lancet
NYHA Class III/IV Cardiac Patient Evaluation (BAL- 365(9474):18771889
2. American Heart Association (2007) Heart disease and stroke
ANCE) Study is an ongoing double-blinded, randomized, statistics2007 update. American Heart Association, Dallas
multicenter study aimed to evaluate the effects of titrated 3. Stewart S, Jenkins A, Buchan S, McGuire A, Capewell S,
oral lixivaptan in patients with hyponatremia hospitalized McMurray JJ (2002) The current cost of heart failure to the
for WHF. The study will include approximately 650 national health service in the UK. Eur J Heart Fail 4:361371
4. Krumholz HM, Parent EM, Tu N, Vaccarino V, Wang Y, Radford
patients, who will receive lixivaptan or placebo with dose MJ et al (1997) Readmission after hospitalization for congestive
titration for 60 days. The primary objective of the study is heart failure among medicare beneficiaries. Arch Intern Med
to determine whether lixivaptan can effectively and safely 157:e99e104
increase [Na] in HF patients with hyponatremia. Secondary 5. Rosamond W, Flegal K, Furie K et al (2008) Heart disease and
stroke statistics2008 update: a report from the American Heart
objectives are to assess all-cause mortality, cardiovascular Association Statistics Committee and Stroke Statistics Subcom-
effects, HF hospitalization, and acute change in BW [55]. mittee. Circulation 117(4):e25e146
Studies have shown that V2 receptor antagonists can 6. Adrogue HJ, Madias NE (2000) Hyponatremia. N Engl J Med
increase the plasma AVP levels. The increased V1A activity 342:15811589
7. De Luca L, Klein L, Udelson JE, Orlandi C, Sardella G, Fedele F,
and the resulting vasoconstriction, release of coagulation Gheorghiade M (2005) Hyponatremia in patients with heart
factors, and platelet aggregation would be deleterious to failure. Am J Cardiol 96:19L23L
patients with HF. Therefore, V1A/V2 receptors antagonists 8. Gheorghiade M, Abraham WT, Albert NM, Gattis Stough W,
may provide better outcomes in HF patients [65]. Greenberg BH, OConnor CM, She L, Yancy CW, Young J,
Fonarow GC (2007) Relationship between admission serum
So far the role of AVP receptor antagonists in the therapy sodium concentration and clinical outcomes in patients hospi-
of HF remains undefined. Further studies are needed to talized for heart failure: an analysis from the OPTIMIZE-HF
better define their role in the setting hyponatremia in HF. registry. Eur Heart J 28:980988
9. Gheorghiade M, Rossi JS, Cotts W, Shin DD, Hellkamp AS, Pina
IL, Fonarow GC, DeMarco T, Pauly DF, Rogers J, DiSalvo TG,
Future perspectives Butler J, Hare JM, Francis GS, Stough WG, OConnor CM
(2007) Characterization and prognostic value of persistent
Hyponatremia is a common and important comorbidity in hyponatremia in patients with severe heart failure in the ESCAPE
HF patients and is a strong independent predictor of mor- trial. Arch Intern Med 167:19982005
10. Klein L, OConnor CM, Leimberger JD, Gattis-Stough W, Pina
tality in HF risk assessment models. It is primarily caused IL, Felker GM, Adams KF Jr, Califf RM, Gheorghiade M (2005)
by increased AVP levels in HF patients, which in turn may Lower serum sodium is associated with increased short-term
be the determinant of the poor outcome. Moreover, hypo- mortality in hospitalized patients with worsening heart failure:
natremia seems to be an important marker of HF progres- results from the Outcomes of a Prospective Trial of Intravenous
Milrinone for Exacerbations of Chronic Heart Failure (OPTIME-
sion, consequential to neurohormonal systems activation CHF) study. Circulation 111:24542460
and impaired hemodynamics. These characteristics are 11. Sica DA (2005) Hyponatremia and heart failurepathophysiol-
translated into worse short-term and long-term outcomes. ogy and implications. Congest Heart Fail 11:274277
Vasopressin receptor antagonists appear promising in the 12. Kearney MT, Fox KA, Lee AJ, Prescott RJ, Shah AM, Batin PD,
Baig W, Lindsay S, Callahan TS, Shell WE, Eckberg DL, Zaman
management of patients with volume overload and hypo- AG, Williams S, Neilson JM, Nolan J (2002) Predicting death
natremia. However, so far only V2 receptor antagonism due to progressive heart failure in patients with mild-to-moderate
(tolvaptan) has been studied in testing long-term outcomes chronic heart failure. J Am Coll Cardiol 40:18011808

123
Heart Fail Rev (2012) 17:1726 25

13. Senni M, De Maria R, Gregori D, Gonzini L, Gorini M, 29. Hasking GJ, Esler MD, Jennings GL, Burton D, Johns JA, Korner
Cacciatore G, Gavazzi A, Pulignano G, Porcu M, Maggioni AP PI (1986) Norepinephrine spillover to plasma in patients with
(2005) Temporal trends in survival and hospitalizations in out- congestive heart failure: evidence of increased overall and car-
patients with chronic systolic heart failure in 1995 and 1999. diorenal sympathetic nervous activity. Circulation 73:615621
J Card Fail 11:270278 30. DiBona GF, Herman PJ, Sawin LL (1988) Neural control of renal
14. Oren RM (2005) Hyponatremia in congestive heart failure. Am J function in edema-forming states. Am J Physiol 254:R1017
Cardiol 95(Suppl):2B7B R1024
15. Adhere Registry: Insights from the Adhere Registry: data from 31. Schrier RW, Abraham WT (1999) Hormones and hemodynamics
over 100,000 patient cases. 2005 in heart failure. N Engl J Med 341:577585
16. Kumar S, Rubin S, Mather PJ, Whellan DJ (2007) Hyponatremia 32. Schuster VL, Kokko JP, Jacobson HR (1984) Angiotensin II
and vasopressin antagonism in congestive heart failure. Clin directly stimulates sodium transport in rabbit proximal convo-
Cardiol 30(11):546551 luted tubules. J Clin Invest 73:507515
17. Gheorghiade M, Gattis WA, OConnor CM et al (2004) For the 33. Goldsmith SR (2005) Current treatments and novel pharmaco-
Acute and Chronic Therapeutic Impact of a Vasopressin Antag- logic treatments for hyponatremia in congestive heart failure. Am
onist in Congestive Heart Failure (ACTIV in CHF) Investigators. J Cardiol 95(9 Suppl 1):B14B23
Effects of tolvaptan, a vasopressin antagonist, in patients hospi- 34. Anderson RJ, Chung HM, Kluge R, Schrier RW (1985) Hypo-
talized with worsening heart failure: a randomized controlled trial natremia: a prospective analysis of its epidemiology and the
(ACTIV). JAMA 291:19631971 pathogenetic role of vasopressin. Ann Intern Med 102:164168
18. Konstam MA, Gheorghiade M, Burnett JC Jr, Grinfeld L, 35. Lee CR, Watkins ML, Patterson JH et al (2003) Vasopressin: a
Maggioni AP, Swedberg K, Udelson JE, Zannad F, Cook T, new target for the treatment of heart failure. Am Heart J 146:918
Ouyang J (2007) Effects of oral tolvaptan in patients hospitalized 36. Kalra PR, Anker SD, Coats AJ (2001) Water and sodium regu-
for worsening heart failure: the EVEREST outcome trial. JAMA lation in chronic heart failure: the role of natriuretic peptides and
297:13191331 vasopressin. Cardiovasc Res 51:495509
19. Aaronson KD, Schwartz JS, Chen TM, Wong KL, Goin JE, 37. Xu YJ, Gopalakrishnan V (1991) Vasopressin increases cytosolic
Mancini DM (1997) Development and prospective validation of a free [Ca2?] in the neonatal rat cardiomyocyte. Evidence for V1
clinical index to predict survival in ambulatory patients referred subtype receptors. Circ Res 69:239245
for cardiac transplant evaluation. Circulation 95(12):26602667 38. Nakamura Y, Haneda T, Osaki J, Miyata S, Kikuchi K (2000)
20. Lee DS, Austin PC, Rouleau JL, Liu PP, Naimark D, Tu JV Hypertrophic growth of cultured neonatal rat heart cells mediated
(2003) Predicting mortality among patients hospitalized for heart by vasopressin V(1A) receptor. Eur J Pharmacol 391:3948
failure derivation and validation of a clinical model. JAMA 39. Goldsmith SR (2006) The role of vasopressin in congestive heart
290(19):25812587 failure. Cleve Clin J Med 73(Suppl 3):S19S23.
21. Kearney MT, Nolan J, Lee AJ, Brooksby PW, Prescott R, Shah 40. Xu DL, Martin PY, Ohara M et al (1997) Upregulation of aqu-
AM, Zaman AG, Eckberg DL, Lindsay HS, Batin PD, Andrews aporin-2 water channel expression in chronic heart failure rat.
R, Fox KA (2003) A prognostic index to predict long-term J Clin Invest 99:15001505
mortality in patients with mild to moderate chronic heart failure 41. Nakamura K, Aoyagi T, Hiroyama M, Kusakawa S, Mizutani R,
stabilised on angiotensin converting enzyme inhibitors. Eur J Sanbe A, Yamauchi J, Kamohara M, Momose K, Tanoue A
Heart Fail 5(4):489497 (2009) Both (V1A) and (V1B) vasopressin receptors deficiency
22. Levy WC, Mozaffarian D, Linker DT, Sutradhar SC, Anker SD, result in impaired glucose tolerance. Eur J Pharmacol
Cropp AB, Anand I, Maggioni A, Burton P, Sullivan MD, Pitt B, 613(13):182188
Poole-Wilson PA, Mann DL, Packer M (2006) The Seattle heart 42. Goldsmith SR (2002) Congestive heart failure: potential role of
failure model: prediction of survival in heart failure. Circulation arginine vasopressin antagonists in the therapy of heart failure.
113:14241433 Congest Heart Fail 8:251256
23. Felker GM, Leimberger JD, Califf RM, Cuffe MS, Massie BM, 43. Francis GS, Benedict C, Johnstone DE et al (1990) Comparison
Adams KF Jr, Gheorghiade M, OConnor CM (2004) Risk of neuroendocrine activation in patients with left ventricular
stratification after hospitalization for decompensated heart fail- dysfunction with and without congestive heart failure. A substudy
ure. J Card Fail 10(6):460466 of the Studies of Left Ventricular Dysfunction (SOLVD). Cir-
24. Vazquez R, Bayes-Genis A, Cygankiewicz I, Pascual-Figal D, culation 82:17241729
Grigorian-Shamagian L, Pavon R, Gonzalez-Juanatey JR, Cubero 44. Rouleau JL, Packer M, Moye L et al (1994) Prognostic value of
JM, Pastor L, Ordonez-Llanos J, Cinca J, Bayes de Luna A on neurohormonal activation in patients with an acute myocardial
behalf of the MUSIC Investigators (2009) The MUSIC risk score: infarction: effect of captopril. J Am Coll Cardiol 24:583591
a simple method for predicting mortality in ambulatory patients 45. Szatalowicz VL, Arnold PE, Chaimovitz C et al (1981) Radio-
with chronic heart failure. Eur Heart J 30:10881096 immunoassay of plasma arginine vasopressin in hyponatremic
25. OConnor CM, Hasselblad V, Mehta RH, Tasissa G, Califf RM, patients with congestive heart failure. N Engl J Med 305:263266
Fiuzat M, Rogers JG, Leier CV, Stevenson LW (2010) Triage 46. Yamane Y (1968) Plasma ADH level in patients with chronic
after hospitalization with advanced heart failure: the ESCAPE congestive heart failure. Jpn Circ J 32:745759
(Evaluation Study of Congestive Heart Failure and Pulmonary 47. Goldsmith SR, Francis GS, Cowley AW, Levine TB, Cohn JN
Artery Catheterization Effectiveness) risk model and discharge (1983) Increased plasma arginine vasopressin levels in patients
score. J Am Coll Cardiol 55(9):872878 with congestive heart failure. J Am Coll Cardiol 1:13851390
26. Oh MS (2002) Pathogenesis and diagnosis of hyponatremia. 48. Verbalis JG, Goldsmith SR, Greenberg A, Schrier RW, Sterns RH
Nephron 92(Suppl 1):28 (2007) Hyponatremia treatment guidelines 2007: expert panel
27. Neilsen S, Kwon TH, Christensen BM, Promeneur D, Frkiaer J, recommendations. Am J Med 120(11 Suppl 1):S121
Marples D (1999) Physiology and pathophysiology of renal 49. Palm C, Gross P (1999) 2-vasopressin receptor antagonists-
aquaporins. J Am Soc Nephrol 10:647663 mechanism of effect and clinical implications in hyponatraemia.
28. Penit J, Faure M, Jard S (1983) Vasopressin and angiotensin II Nephrol Dial Transplant 14:25592562
receptors in rat aortic smooth muscle cells in culture. Am J 50. Palm C, Reimann D, Gross P (2001) The role of V2 vasopressin
Physiol 244:E72E82 antagonists in hyponatremia. Cardiovasc Res 51:403408

123
26 Heart Fail Rev (2012) 17:1726

51. Gheorghiade M, Gattis WA, Barbagelata A et al (2003) Rationale 60. Annane D, Decaux G, Smith N, for the Conivaptan Study Group
and study design for a multicenter, randomized, double-blind, (2009) Efficacy and safety of Oral Conivaptan, a Vasopressin-
placebo controlled study of the effects of tolvaptan on the acute Receptor Antagonist, evaluated in a randomized, controlled trial
and chronic outcomes of patients hospitalized with worsening in patients with Euvolemic or Hypervolemic Hyponatremia. Am J
congestive heart failure. Am Heart J 145(Suppl 2):S51S54 Med Sci 337(1):2836
52. Burrell LM, Risvanis J, Johnston CI et al (2000) Vasopressin 61. Ghali JK, Koren MJ, Taylor JR, Brooks-Asplund E, Fan K, Long
receptor antagonism-a therapeutic option in heart failure and WA et al (2006) Efficacy and safety of oral conivaptan: a V1A/
hypertension. Exp Physiol 85:259S265S V2 vasopressin receptor antagonist, assessed in a randomized,
53. Thibonnier M, Coles P, Thibonnier A, Shoham M (2001) The placebo-controlled trial in patients with euvolemic or hypervo-
basic and clinical pharmacology of nonpeptide vasopressin lemic hyponatremia. J Clin Endocrinol Metab 91:21452152
receptor antagonists. Annu Rev Pharmacol Toxicol 41:175202 62. Gheorghiade M, Niazi I, Ouyang J et al (2003) Vasopressin V2-
54. Goldsmith SR (1999) Vasopressin: a therapeutic target in con- receptor blockade with tolvaptan in patients with chronic heart
gestive heart failure? J Card Fail 5:347356 failure. Circulation 107:26902696
55. Quittnat F, Gross P (2006) Vaptans and the treatment of water 63. Gheorghiade M, Konstam MA, Burnett JC Jr et al (2007) Efficacy
retaining disorders. Semin Nephrol 26:234243 of Vasopressin Antagonism in Heart Failure Outcome Study with
56. Package insert (2008) Vaprisol (conivaptan). Astellas Pharma Tolvaptan (EVEREST) Investigators. Short-term clinical effects
US, Deerfield of tolvaptan, an oral vasopressin antagonist, in patients hospi-
57. Package insert (2009) Samsca (tolvaptan). Otsuka America talized for heart failure: the EVEREST clinical status trials.
Pharmaceutical, Inc., Rockville JAMA 297:13321343
58. CardioKine Inc. THE BALANCE study: treatment of hypona- 64. Abraham WT, Shamshirsaz AA, McFann K, Oren RM, Schrier
tremia based on Lixivaptan in NYHA class III/IV cardiac patient RW (2006) Aquaretic effect of lixivaptan, an oral, non-peptide,
evaluation. http://www.clinicaltrials.gov selective V2 receptor vasopressin antagonist, in New York Heart
59. Zeltser D, Rosansky S, van Rensburg H, Verbalis JG, Smith N, Association Functional Class II and III chronic heart failure
Conivaptan Study Group (2007) Assessment of the efficacy and patient. J Am Coll Cardiol 47(8):16151621
safety of intravenous conivaptan in euvolemic and hypervolemic 65. Goldsmith SR, Gheorghiade M (2005) Vasopressin antagonism in
hyponatremia. Am J Nephrol 27:447457 heart failure. J Am Coll Cardiol 46:17851791

123

You might also like