You are on page 1of 11

768488 ACC European Heart Journal: Acute Cardiovascular CareVerbrugge

Education in Acute Cardiovascular Care


European Heart Journal: Acute Cardiovascular Care

Editor’s Choice-Diuretic resistance 2018, Vol. 7(4) 379­–389


© The European Society of Cardiology 2018
Reprints and permissions:
in acute heart failure sagepub.co.uk/journalsPermissions.nav
https://doi.org/10.1177/2048872618768488
DOI: 10.1177/2048872618768488
journals.sagepub.com/home/acc

Frederik H Verbrugge

Abstract
Diuretic resistance is a powerful predictor of adverse outcome in acute heart failure (AHF), irrespectively of underlying
glomerular filtration rate. Metrics of diuretic efficacy such as natriuresis, urine output, weight loss, net fluid balance, or
fractional sodium excretion, differ in their risk for measurement error, convenience, and biological plausibility, which
should be taken into account when interpreting their results. Loop diuretic resistance in AHF has multiple causes
including altered drug pharmacokinetics, impaired renal perfusion and effective circulatory volume, neurohumoral
activation, post-diuretic sodium retention, the braking phenomenon and functional as well as structural adaptations in
the nephron. Ideally, these mechanisms should guide specific treatment decisions with the goal of achieving complete
decongestion. Therefore, volume overload needs to be identified correctly to avoid poor diuretic response due to
electrolyte depletion or dehydration. Next, renal perfusion should be optimised if possible and loop diuretics should
be prescribed above their threshold dose. Addition of thiazide-type diuretics should be considered when a progressive
decrease in loop diuretic efficacy is observed with prolonged use (i.e., the braking phenomenon). Furthermore, thiazide-
type diuretics are a useful addition in patients with low glomerular filtration rate. However, they limit free water
excretion and are relatively contraindicated in cases of hypotonic hyponatremia, where acetazolamide is the better
option. Finally, ultrafiltration should be considered in patients with refractory diuretic resistance as persistent volume
overload after decongestive treatment is associated with worse outcomes. Whether more upfront use of any of these
individually tailored decongestion strategies is superior to monotherapy with loop diuretics remains to be shown by
adequately powered randomised clinical trials.

Learning objectives
•• To understand the concept of diuretic resistance in acute heart failure with its different definitions
•• To be aware of the prognostic significance of diuretic resistance in acute heart failure
•• To understand the mechanisms of diuretic resistance in acute heart failure
•• To develop a tailored approach to handle diuretic resistance in acute heart failure

Key points
•• Diuretic resistance in acute heart failure is defined either descriptively as the persistence of congestion despite
adequate decongestive treatment, or quantitatively as low natriuresis / urine output / net fluid loss / weight loss /
fractional sodium excretion per diuretic dose administered.
•• Irrespectively of its definition, diuretic resistance in acute heart failure is consistently associated with poor out-
comes and this remains true when adjusting for underlying glomerular filtration rate.
•• Diuretic resistance tracks poorly with changes in glomerular filtration rate during decongestive treatment, but is
more frequent in case of (more severe) underlying chronic kidney disease.

Department of Cardiology, Jessa Ziekenhuis, Belgium Corresponding author:


Frederik H Verbrugge, Department of Cardiology, Jessa Ziekenhuis,
Stadsomvaart 11, Hasselt, 3500, Belgium.
Email: frederik.verbrugge@zol.be
380 European Heart Journal: Acute Cardiovascular Care 7(4)

•• Diuretic resistance has multiple causes in acute heart failure including altered drug pharmacokinetics, impaired renal
perfusion and effective circulatory volume, neurohumoral activation, post-diuretic sodium retention, the braking
phenomenon and functional as well as structural adaptations in the nephron.
•• True diuretic resistance should be discerned from poor diuretic response due to electrolyte depletion or dehydration
•• The treatment approach to diuretic resistance in acute heart failure is focused ideally on the underlying mechanis-
tic cause implied.

Keywords
Cardio-renal syndrome, diuretics, heart failure, sodium

Date received: ; accepted:

Introduction congestion is associated with worse outcomes after AHF.


Indeed, even when coming at the cost of worsening renal
Signs and symptoms of volume overload are the predomi- function, better decongestion is associated with improved
nant reason for hospital admission in acute heart failure hospital-free survival.11 A reasonable treatment goal in
(AHF).1 Loop diuretics are nearly ubiquitously prescribed AHF is thus to pursue complete decongestion while trying
for this indication and are the only applied therapy in a to avoid iatrogenic harm, in particular with respect to the
majority of cases.2 Response to loop diuretic therapy kidneys. As intravenous loop diuretics are prescribed in the
may be assessed in many ways, either qualitatively (i.e. dis- overwhelming majority of patients to achieve this goal, it
appearance of clinical congestion signs or normalisation of makes sense to search for and validate metrics of their
cardiac filling pressures) or quantitatively (i.e. natriuresis, efficacy.3
urine output, net fluid loss or weight loss). Emerging evi-
dence indicates that response to decongestive treatment in
general and loop diuretics in particular may be an inde- Loop diuretic efficacy: prognostic
pendent prognostic factor in AHF.3 The frequently encoun- marker or causal factor in AHF?
tered clinical problem of diuretic resistance in AHF
therefore merits attention, especially because an evidence- Working mechanism of loop diuretics
based approach is lacking and different treatment strategies Loop diuretics inhibit the sodium–potassium–chloride
have failed to demonstrate meaningful benefits in ran- cotransporter (NKCC2) at the luminal side of the renal
domised clinical trials.4–6 This review aims to discuss the tubules in the thick ascending limb of Henle’s loop, which
concept of diuretic resistance in AHF, with its different accounts for approximately 25% of the total renal sodium
definitions and underlying mechanisms, to provide a better reabsorption.12 The primary pharmacodynamic effect of
pathophysiological insight and suggest an integrated treat- loop diuretics is therefore induction of natriuresis, chloru-
ment approach. resis and kaliuresis. Yet, because they also impair the uri-
nary concentration capacity of the kidneys dependent on
the NKCC2, loop diuretics stimulate water loss as well as
The goal of decongestion in AHF
producing hypo to isotonic urine.13
Over the past few decades, immense progress has been
made in the pharmacological and device treatment of
chronic heart failure with reduced ejection fraction. Indeed,
Loop diuretic efficacy
there has been a dramatic reduction in all-cause mortality, A very accurate metric for loop diuretic efficacy would be
from approximately 40% to 20% within 2 years after pres- the sodium content of tubular fluid that exits Henle’s loop.
entation.7 Unfortunately, this progress stands in stark con- To adjust for pharmacokinetics, one might subsequently
trast to the lack of advancements made in AHF, in which correct for loop diuretic plasma levels. Of course, tubular
large randomised clinical trials have repeatedly failed to fluid sampling is not achievable in patients with AHF and
show a reduction in either mortality or hospital readmis- systemic assessment of loop diuretic plasma levels would
sions despite various different treatment strategies.4–6,8–10 be cumbersome. Therefore, alternative metrics have been
Because there is little high-quality evidence to guide man- proposed to assess loop diuretic efficacy in a more practical
agement decisions in AHF, treatment often remains empiri- way. Although each has its strengths and limitations (Table
cal and according to local practice. One very consistent 1), findings have been very consistent in demonstrating that
finding however – both from real-world observational data poor loop diuretic efficacy – hence diuretic resistance – is
and post hoc analysis of clinical trials – is that persistent associated with detrimental clinical outcomes (Table 2).
Verbrugge 381

Table 1.  Strengths and weaknesses of different loop diuretic efficacy metrics.

Metric Measurement error Convenience Plausibility


Natriuresis ++++/++a + +++
Urine output ++++/+a ++ ++
Net fluid loss – + ++
Weight loss ++ +++ +
Fractional sodium excretion ++++ + +++
Urinary sodium/furosemide concentration ++++ – ++++
aWith/without bladder catheter.

Table 2.  Loop diuretic efficacy metrics and outcome.

Metric References Outcome with diuretic resistance


Natriuresisa Verbrugge et al. (2015)24 Higher all-cause mortality
Urine outputa Aronson et al. (2015)29 Higher all-cause mortality
  Ter Maaten et al. (2015)28 Increased death or heart failure readmission
Net fluid lossa Testani et al. (2014)14 Higher all-cause mortality
  Aronson et al. (2015)29 Higher all-cause mortality
Weight lossa Valente et al. (2014)26 Higher all-cause mortality
  Voors et al. (2014)27 Increased death or readmission due to
heart/kidney failure
  Ter Maaten et al. (2015)28 Increased death or heart failure readmission
  Ter Maaten et al (2016)68 Increased early readmission
  Aoki et al. (2017)69 Increased death or heart failure readmission
Fractional sodium excretion Kumar et al. (2015)70 Higher all-cause mortality
Urinary sodium/furosemide concentration Singh et al. (2014)23 Increased death/heart transplantation or
heart failure readmission
  Doering et al. (2017)71 Lower readmissions
aPer 40 mg intravenous furosemide or equivalent dose.

Loop diuretic efficacy and prognosis in AHF answer to this question may also depend on the metric
being used to define loop diuretic efficacy.
The exact mechanistic underpinning of the robust associa-
tion between loop diuretic efficacy and prognosis in AHF is
less obvious than at first sight. A logical explanation might Strengths and weaknesses of loop
be that AHF patients who present with loop diuretic resist- diuretic efficacy metrics
ance have a lower chance of achieving appropriate decon-
gestion. Yet, similar congestion signs were reported in Measurement error
some cohorts at discharge in patients with versus without An important issue with all metrics of loop diuretic efficacy
diuretic resistance and still there was a difference in out- is measurement error. Urine output is prone to substantial
comes.14 Alternatively, loop diuretic efficacy might be collection error in the absence of a bladder catheter, and
interpreted as a renal stress test, indicating the reserve func- even a simple measurement of weight can be hard to repro-
tion of the kidneys to excrete sodium and water in analogy duce when patients are frail and unable to stand alone. Net
to the maximal aerobic capacity being reflective of the car- fluid balance is the most susceptible to measurement error
diac reserve during exercise. Finally, poor diuretic response as both fluid intake and output must be assessed simultane-
might also indicate that volume overload is not present and ously. Indeed, even in rigorously collected data from large
sodium levels are possibly depleted.15,16 In the latter sce- randomised clinical trials, the correlation between metrics
nario, diuretic therapy is unlikely to target the underlying such as weight loss and net fluid balance is very poor.17
pathophysiological culprit of AHF and may be harmful
instead. Although diuretic resistance is clearly a risk marker
Convenience/complexity
in AHF, it remains an open question whether interventions
that increase loop diuretic efficacy also lead to improved Some metrics of loop diuretic efficacy require more effort
outcomes in AHF. This would make it a causal factor and to measure (i.e. net fluid balance) or need laboratory analy-
hence an attractive end-point in future clinical trials. The ses and/or calculations (i.e. natriuresis, fractional sodium
382 European Heart Journal: Acute Cardiovascular Care 7(4)

excretion, urinary sodium/furosemide ratio). In addition,


although urinary indices may contain interesting informa-
tion, their interpretation is not straightforward during diu-
retic therapy and requires more study, specifically in the
context of AHF.15

Biological plausibility
An important limitation of urine output, weight loss and net
fluid balance is that these metrics do not distinguish
between dehydration and decongestion, the latter being the
removal of water and sodium. This distinction is relevant to
make as arginine vasopressin antagonists (i.e. vaptans) that Figure 1.  Loop diuretic pharmacokinetics.
induce pure water excretion without impact on natriuresis
relieve clinical congestion signs but have no impact on
long-term outcomes after AHF.18 It is important to recog-
nise that most AHF patients actually have normal to high
serum osmolality, reflecting total body water in proportion
to solute content.19,20 As extracellular volume is governed
by sodium and not water homeostasis, natriuresis and frac-
tional sodium excretion may better capture long-term
effects of loop diuretic therapy on volume status.12

Glomerular filtration rate versus loop


diuretic efficacy
Estimates of glomerular filtration rate (GFR) are widely
used to assess renal function. GFR reflects the clearance
function of the kidneys and therefore indicates well how
toxic waste products are removed from the blood. In con-
Figure 2.  Loop diuretic dose–response curve.
trast, sodium and volume homeostasis is predominantly
influenced by renal tubular function.12 During decongestive
treatment in AHF, only a modest correlation between wors- furosemide demonstrates a highly variable oral bioavailabil-
ening GFR and biomarkers of tubular injury has been ity (10-100%), especially when intake is combined with
found.21,22 This explains why loop diuretic efficacy tracks food.31 Food intake reduces the maximal plasma concentra-
poorly with incident worsening renal function during tion of all loop diuretics by approximately 50%, but pro-
decongestion and predicts outcomes in AHF irrespective of longs the time of their presence in the blood (Figure 1). As
underlying GFR.14,23–25 That said, the frequency of diuretic loop diuretic plasma levels need to exceed a critical thresh-
resistance is clearly much higher with lower baseline GFR, old to induce natriuresis, food intake increases the risk of
when more advanced underlying chronic kidney disease is diuretic resistance, but in the case of diuretic efficacy effects
present.14,26–29 In conclusion, while GFR is an excellent are prolonged. In addition, in AHF with marked volume
marker for chronic kidney disease and hence a good risk overload, especially when right heart failure is present, loop
marker for chronic heart failure when assessed in stable diuretic absorption may be impaired because of gut oedema
conditions, loop diuretic resistance performs better for and poor intestinal perfusion.32,33
prognostic stratification in AHF.14,30
Protein binding and tubular secretion of loop diuretics. Once
absorbed, loop diuretics are more than 90% bound to plasma
Causes of loop diuretic resistance proteins. Therefore, the primary entrance of loop diuretics
into the renal tubules is not through glomerular filtration,
Altered drug pharmacokinetics but rather through secretion by organic anion transporters
Oral bioavailability.  To exert their effects on the NKCC2 at and the multidrug resistance-associated protein 4 in the
the luminal side of the renal tubules, orally administered proximal tubules.34 Uraemic anions compete with loop
loop diuretics must first be absorbed from the gastrointesti- diuretic agents for proximal secretion, and the transport of
nal tract. Different pharmacological agents have shown sub- both is inhibited by metabolic acidosis.35,36 This explains the
stantial differences in oral bioavailability. While torasemide shifted dose-response curve of loop diuretics in patients
and bumetanide are almost completely absorbed (80-100%), with chronic kidney disease (Figure 2). In addition, low
Verbrugge 383

Figure 3.  Post-diuretic sodium retention and the braking phenomenon.

plasma protein levels may decrease the amount of protein- triggered by inhibited chloride transport via the NKCC2 in
bound loop diuretics offered to peritubular capillaries in the macula densa cells lining the renal tubular lumen at the end
proximal renal tubules. Hence lower effective tubular con- of Henle’s loop.12 Indeed, low chloride concentrations
centrations of loop diuretics are reached for any given inside macula densa cells depolarise their cell membrane,
plasma level. Finally, significant proteinuria, as a result of leading to paracrine signalling towards the nearby afferent
glomerular or tubular damage, increases protein binding of arteriole where renin is ready to be released. With the
loop diuretics in the renal tubular lumen, preventing their renin-angiotensin–aldosterone system already potently
pharmacological action.37 activated in heart failure, this further contributes to sodium
avidity in between moments of diuretic administration.
This is especially relevant for furosemide and bumetanide
Impaired renal perfusion and effective that have a relatively short half-life (1-3 hours) in compari-
circulatory volume son with torasemide (approximately 6 hours). Post-diuretic
In AHF, volume overload is often present outside the vascular sodium retention implies that when sodium intake is high,
compartment (i.e. interstitial oedema, ascites and pleural effu- it can match or even exceed the loss achieved with inter-
sion) or pooled in systemic capacitance veins (mostly in the mittent loop diuretic administration (Figure 3).42 Interest-
splanchnic circulation).33 However, the kidneys are only able ingly, although plasma renin activity increases acutely
to regulate the effective circulatory volume. In normal circum- with appropriate decongestion, such a rise does not seem
stances, the plasma refill rate is approximately 3–4 mL/kg per to translate into worse outcomes.43 In contrast, neurohu-
hour, which explains how clinical congestion signs may disap- moral activation accompanied by poor diuretic efficacy or
pear over time with diuretic therapy. Yet situations may occur persistent volume overload – hence diuretic resistance –
in which renal perfusion is severely compromised and fluid clearly does.24,43
cannot be recruited. Obviously, in case of low-output heart
failure or cardiogenic shock, diuretic agents are ineffective The braking phenomenon.  It is well known that with repeated
and renal perfusion must first be restored. Furthermore, exces- administration of the same dose of loop diuretics, natriuresis
sive abdominal congestion is associated with increased intra- decreases over time (Figure 3). This may indicate decreasing
abdominal pressure and poor renal perfusion that can be extracellular volume overload and removal of excess sodium
reversed by ultrafiltration or mechanical removal of ascites.38,39 with appropriate decongestion.15 However, neurohumoral
Finally, evidence is accumulating that chronic sodium over- activation of the sympathetic nerve and renin–angiotensin–
load induces structural changes in the glycosaminoglycan con- aldosterone system as well as structural and functional adap-
tent of the interstitial compartment that may promote oedema tations in the nephron may lead to an excessive braking
formation and impair the plasma refill rate.40,41 phenomenon despite persistent volume overload.

Neurohumoral activation, post-diuretic Structural and functional nephron


sodium retention and the braking adaptations causing diuretic resistance
phenomenon Distal nephron hypertrophy. Loop diuretics increase the
Post-diuretic sodium retention.  Loop diuretics directly stim- sodium and chloride load delivered to the distal nephron.
ulate renin release in the afferent arteriole, which is Over time, this exposure causes intrinsic renal adaptations
384 European Heart Journal: Acute Cardiovascular Care 7(4)

Figure 4.  Integrated management approach to treat volume overload and loop diuretic resistance in acute heart failure.

that impair natriuretic efficacy (i.e. distal tubular hypertro- is lower. The result is thicker blood with a higher oncotic
phy and increased local aldosterone secretion) and are pressure entering the peritubular capillaries through the
mediated by the thiazide-sensitive sodium-chloride sym- efferent arteriole. In turn, this stimulates isotonic reabsorp-
porter and sodium-independent chloride/iodide transporter tion in the proximal renal tubules and may lead to substan-
or pendrin.44,45 tially increased proximal tubular sodium reabsorption.12
The consequence is that less sodium (and chloride) is
Increased proximal tubular sodium reabsorption. For loop offered to the more distal nephron and Henle’s loop, impair-
diuretics to be effective, enough substrate must be available ing loop diuretic efficacy.12
to the NKCC2. In other words, enough sodium, potassium
and chloride should pass through the tubular lumen at the
Management approach to (loop) diuretic
level of the thick ascending limb of Henle’s loop. As
resistance in AHF
explained above, adequate renal perfusion is a prerequisite
to produce ultrafiltrate passing through the renal tubular As the mechanisms of loop diuretic resistance in AHF are
lumen and maintain a normal GFR. Importantly, renal auto- multifactorial, we propose an integrated management
regulation mechanisms aim to preserve GFR initially, even approach (Figure 4). It should be emphasised though,
when renal blood flow is already substantially depressed.12 because of the lack of high-quality evidence from ran-
This can be explained by a higher fraction of plasma being domised clinical trials, such recommendations rely heavily
filtered through the renal glomeruli when renal blood flow on pathophysiological insights and observational data.
Verbrugge 385

Is volume overload the target culprit? When considering more pronounced neurohumoral activation.9,50,51 The benefit
(loop) diuretic therapy in AHF, one should never forget the of the continuous administration of loop diuretics is that
primary aim of this therapy, which is creating a net nega- post-diuretic sodium retention does not occur, yet this can
tive sodium balance to reduce extracellular volume and also easily be overcome by repeating loop diuretic bolus
hence relief signs and symptoms of congestion. Yet, in administration at 6-8 hour intervals if the patient is still vol-
some presentations of AHF, volume overload may not be ume overloaded. In addition, it is easy to adjust the pace of
the target culprit. For example, cardiogenic shock due to decongestion with bolus therapy when the plasma refill rate
an acute coronary syndrome or fulminant myocarditis may is impaired. Finally, it is crucially important to adjust the
even require carefully administered fluids with the pur- dose of loop diuretics according to the GFR and plasma pro-
pose of haemodynamic stabilisation, despite the presence tein levels (Figure 2).
of significant pulmonary oedema. Furthermore, flash pul-
monary oedema due to a hypertensive crisis or severe Assess loop diuretic efficacy to recognise loop diuretic resistance
mitral valve regurgitation is caused by volume misdistri- early.  When administering loop diuretics, it is important to
bution rather than volume overload per se and might be assess the response to therapy and recognise diuretic resis-
addressed better with vasodilator therapy or valve inter- tance early to intervene. In most patients, urine output and
vention, respectively.46 In other phenotypes such as pri- weight loss suffice to evaluate loop diuretic efficacy. How-
mary right heart failure or constrictive pericarditis, some ever, in patients with or at high risk of cardiorenal syn-
degree of volume overload may be necessary to maintain drome, following natriuresis may offer a better perspective
adequate preload and is not the primary target of therapy. on decongestion success.15 The goal of diuretic treatment
Main indicators of volume overload are limb oedema, asci- should be to continue until clinical signs and symptoms of
tes, pleural effusion, or significant weight gain over a volume overload have completely disappeared and cardiac
short-term period. In contrast, jugular venous distension, filling pressures have normalised.
orthopnoea, bendopnoea and pulmonary oedema are less
specific as they reflect increased cardiac filling pressures, Continuously assess the effective circulatory volume. When
but not necessarily volume overload. clinical signs and symptoms of volume overload persist
despite rapidly decreasing loop diuretic efficacy, one should
Are the kidneys adequately perfused?  If volume overload is consider that the effective circulatory volume is decreased.
present and deemed to be the target culprit in AHF, there is This might be further supported by deteriorating GFR or
a clear indication for diuretic therapy. However, for diuretic very low cardiac filling pressures. A simple treatment
therapy to be successful, renal perfusion must be adequate. response might be just to slow down the pace of diuretic
Renal blood flow depends on mean arterial pressure, cen- administration and allow more time for plasma refilling.
tral venous pressure and intrarenal vascular resistance. Sometimes there might be fluid entrapment in third space
Arterial hypotension during decongestive therapy is compartments which can be tapped (e.g. ascites or pleural
strongly associated with worsening renal function and effusion). Alternatively, the plasma refill rate may be
should be avoided when possible.47 Furthermore, lowering severely compromised because of low plasma oncotic pres-
central venous pressure through administration of vasodila- sure, vascular leak, or alterations in the interstitial matrix.40,41
tor therapy may lower the need for diuretics in AHF.48 Spe- Compression stockings should be considered in such cases.
cifically in the case of low cardiac output with normal or Intravenous albumin administration just before diuretic
elevated arterial blood pressure (i.e. high systemic vascular therapy is effective to recruit peripheral oedema, especially
resistance), intravenous sodium nitroprusside improves when plasma levels are low, but is expensive and has not
organ perfusion and hence renal blood flow.49 Significant been shown to improve outcomes in AHF.
ascites in the setting of AHF is usually associated with
increased intra-abdominal pressure, which impairs renal Optimise chronic heart failure therapy in the case of reduced
perfusion and may be alleviated by paracentesis.39 Finally, ejection fraction. Although pharmacological therapies in
when arterial blood pressure is low due to cardiogenic AHF have not been demonstrated to reduce all-cause mor-
shock, inotropes and/or mechanical assist devices should tality or heart failure readmissions in the long term, a hos-
be considered. pital admission should always be considered as an
opportunity to improve chronic heart failure therapy.52 The
Consider loop diuretic pharmacokinetics. When a patient is neurohumoral system is both an important treatment target
admitted to the hospital with marked volume overload, intra- in heart failure with reduced ejection fraction and mecha-
venous administration of loop diuretics may be preferred nistically implied in the occurrence of (loop) diuretic
over oral intake to overcome concerns about gastrointestinal resistance. Indeed, it has been well established that renin-
reabsorption and oral bioavailability. There is no difference angiotensin-aldosterone system blockers decrease the
between bolus versus continuous administration with respect need for diuretics in congestive heart failure, especially in
to successful decongestion, but the latter is associated with the long term.53,54
386 European Heart Journal: Acute Cardiovascular Care 7(4)

Thiazide-type diuretics.  Thiazide-type diuretics are the most efficacy and are among the most promising drugs under
frequently considered therapy in the case of loop diuretic investigation in chronic heart failure.63
resistance in AHF. They are effective mainly when the
cause of diuretic resistance is distal tubular hypertrophy Ultrafiltration.  As loop diuretic therapy results in the pro-
because of prolonged exposure to loop diuretics.44 Alter- duction of hypotonic urine, while ultrafiltration removes
natively, adding a thiazide-type diuretic up front is useful isotonic plasma and hence more sodium for the same
in the case of a low GFR to boost the fractional sodium amount of water, it has been hypothesised that the latter
excretion and ensure adequate natriuresis.55 A drawback of might be a superior decongestion strategy.64 Indeed, in the
thiazide-type diuretics is that they limit free water excre- Ultrafiltration versus Intravenous Diuretics for Patients
tion or the urinary dilution capacity of the kidneys and Hospitalized for Acute Decompensated Heart Failure
therefore they should be avoided in hypotonic hyponatre- (UNLOAD) study, which included 200 patients with AHF
mia.56 Most clinical evidence to support the use of thia- and clear signs of volume overload, ultrafiltration outper-
zide-type together with loop diuretics in AHF comes from formed loop diuretics, producing greater weight and fluid
small observational studies. Those indicate a probable loss.65 Moreover, the 90-day readmission rate was also sig-
class effect and a 75-90% response rate in patients with nificantly lower in the ultrafiltration compared to loop
loop diuretic resistence.57 In addition, some intriguing data diuretic group, presumably due to more effective deconges-
from the stepwise pharmacological care arm in the Cardio- tion.65 These findings are somewhat in contradiction to the
renal Rescue Study in Acute Decompensated Heart Failure results of the much larger CARRESS-HF study(n=2033),
(CARRESS-HF) suggest that the combination of a diuretic which found no better decongestion – and hence no better
efficacy-guided approach with step-up of thiazide-based clinical outcome – with ultrafiltration compared to pharma-
therapy provides effective diuresis without compromising cological care with strong emphasis on combinational treat-
GFR, even when directly compared to an aggressive ultra- ment.4 Moreover, catheter-related adverse events and
filtration approach.58 bleeding complications were more frequent in the ultrafil-
tration group. In addition, GFR improved significantly
Acetazolamide.  Acetazolamide is an old and largely forgot- more with pharmacological care after 60 days.4 In light of
ten diuretic, which is still in use for the treatment of moun- these results, we would recommend optimising pharmaco-
tain disease and obstructive sleep apnoea syndrome. As a logical therapy first, with ultrafiltration reserved as bail-
carbonic anhydrase inhibitor, it blocks sodium bicarbonate out in cases of refractory congestion. Nevertheless, it
reabsorption in the proximal tubules.12 Consequently, it should be noted that such patients generally have a very
improves sodium delivery to the NKCC2 hence boosting poor prognosis.66 One important point of criticism to CAR-
loop diuretic efficacy. One observational study in patients RESS-HF might be, however, that the pharmacological
with AHF and marked volume overload found that the care arm allowed decongestive treatment to be titrated very
addition of acetazolamide improved loop diuretic efficacy carefully on an individual base, according to diuretic effi-
with approximately 100 mmol sodium excreted per 40 mg cacy. In contrast, the ultrafiltration rate was pre-set at a
of furosemide-equivalent dose.24 Furthermore, acetazol- constant rate of 200 mL per hour, which was only changed
amide also improves thiazide-type diuretic efficacy, as it for technical reasons or clinical requirements, as assessed
potently downregulates pendrin expression in the distal by the treating physician. This has potentially put patients
nephron.59 Pendrin, also known as the sodium-independent in the ultrafiltration arm at greater risk of intravascular vol-
chloride/iodide transporter, compensates for sodium and ume depletion and hypotensive episodes, which might have
chloride loss in the distal convoluted tubules and might be been associated with more harmful neurohumoral activa-
an unrecognised source of diuretic resistance.45,60 While the tion. The Aquapheresis Versus Intravenous Diuretics and
diuretic and natriuretic capacity of acetazolamide is poor Hospitalization for Heart Failure (AVOID–HF) trial could
on its own, it might well be a very efficient booster of have filled this important gap in the evidence.67 Regretta-
diuretic efficacy in combination diuretic therapy.59,61 This bly, the sponsor of this trial unilaterally decided to termi-
concept is further supported by one small randomised trial nate the study after only 27% of the intended study
including 24 patients with volume overload refractory to population was recruited, despite the absence of any futility
loop diuretic therapy.62 All these patients demonstrated a or safety concerns, rendering the trial severely underpow-
greatly reduced fractional sodium excretion, which was ered. For this reason, results should be interpreted very cau-
easily overcome by the addition of acetazolamide. At the tiously, but the estimated days to a first heart failure event
moment, there are insufficient data to recommend add-on were in favour of the adjustable ultrafiltration group versus
or even up-front therapy with acetazolamide in addition to the adjustable loop diuretics group (62 versus 34 days; P
loop diuretics. Intriguingly, however, sodium-glucose value 0.106). Therefore, individually titrated ultrafiltration
transporter 2 inhibitors that also block sodium transport in therapy may still have a role to play in the treatment of
the proximal renal tubules greatly increase loop diuretic cardiorenal syndrome, yet more evidence is needed.
Verbrugge 387

Conclusions 11. Metra M, Davison B, Bettari L, et al. Is worsening renal


function an ominous prognostic sign in patients with acute
Loop diuretic resistance is a frequently encountered problem heart failure? The role of congestion and its interaction with
in AHF and portends poor prognosis. It is therefore impor- renal function. Circ Heart Fail 2012; 5: 54–62.
tant for the clinician to assess loop diuretic efficacy in AHF 12. Verbrugge FH, Dupont M, Steels P, et al. The kidney in con-
patients, when providing decongestive treatment. Different gestive heart failure: ‘are natriuresis, sodium, and diuretics
metrics for loop diuretic resistance have all been consistently really the good, the bad and the ugly?’. Eur J Heart Fail
associated with adverse clinical outcomes. Yet it remains an 2014; 16: 133–142.
open question whether therapies that boost diuretic efficacy 13. Mullens W, Verbrugge FH, Nijst P, et al. Renal sodium avid-
ity in heart failure: from pathophysiology to treatment strate-
or prevent diuretic resistance improve prognosis in AHF. The
gies. Eur Heart J 2017; 38: 1872–1882.
mechanisms of loop diuretic resistance in AHF are multifac- 14. Testani JM, Brisco MA, Turner JM, et al. Loop diuretic effi-
torial and require an integrated approach. ciency: a metric of diuretic responsiveness with prognostic
importance in acute decompensated heart failure. Circ Heart
Conflict of interest Fail 2014; 7: 261–270.
The author declares that there is no conflict of interest. 15. Verbrugge FH, Nijst P, Dupont M, et al. Urinary composition
during decongestive treatment in heart failure with reduced
ejection fraction. Circ Heart Fail 2014; 7: 766–772.
Funding 16. Verbrugge FH, Grodin JL, Mullens W, et al. Transient

This research received no specific grant from any funding agency hyponatremia during hospitalization for acute heart failure.
in the public, commercial, or not-for-profit sectors. Am J Med 2016; 129: 620–627.
17. Testani JM, Brisco MA, Kociol RD, et al. Substantial dis-
crepancy between fluid and weight loss during acute decom-
References pensated heart failure treatment. Am J Med 2015; 128:
1. Adams KF Jr, Fonarow GC, Emerman CL, et al. 776–783 e4.
Characteristics and outcomes of patients hospitalized for 18. Konstam MA, Gheorghiade M, Burnett JC Jr, et al. Effects
heart failure in the United States: rationale, design, and of oral tolvaptan in patients hospitalized for worsening heart
preliminary observations from the first 100,000 cases in failure: the EVEREST Outcome Trial. JAMA 2007; 297:
the Acute Decompensated Heart Failure National Registry 1319–1331.
(ADHERE). Am Heart J 2005; 149: 209–216. 19. Vaduganathan M, Goldsmith SR, Senni M, et al. Contrasting
2. Fonarow GC and Corday E. Overview of acutely decom- acute and chronic effects of tolvaptan on serum osmolality in
pensated congestive heart failure (ADHF): a report from the the EVEREST trial. Eur J Heart Fail 2016; 18: 185–191.
ADHERE registry. Heart Fail Rev 2004; 9: 179–185. 20. Verbrugge FH. Decongestion: more than meets the eye! Eur
3. Verbrugge FH, Mullens W and Tang WH. Management of J Heart Fail 2016; 18: 192–194.
cardio-renal syndrome and diuretic resistance. Curr Treat 21. Dupont M, Shrestha K, Singh D, et al. Lack of significant renal
Options Cardiovasc Med 2016; 18: 11. tubular injury despite acute kidney injury in acute decompen-
4. art BA, Goldsmith SR, Lee KL, et al. Ultrafiltration in sated heart failure. Eur J Heart Fail 2012; 14: 597–604.
decompensated heart failure with cardiorenal syndrome. N 22. Verbrugge FH, Dupont M, Shao Z, et al. Novel urinary bio-
Engl J Med 2012; 367: 2296–2304. markers in detecting acute kidney injury, persistent renal
5. Chen HH, Anstrom KJ, Givertz MM, et al. Low-dose dopa- impairment, and all-cause mortality following decongestive
mine or low-dose nesiritide in acute heart failure with renal therapy in acute decompensated heart failure. J Card Fail
dysfunction: the ROSE acute heart failure randomized trial. 2013; 19: 621–628.
JAMA 2013; 310: 2533–2543. 23. Singh D, Shrestha K, Testani JM, et al. Insufficient natriuretic
6. Packer M, O’Connor C, McMurray JJV, et al. Effect of ula- response to continuous intravenous furosemide is associated
ritide on cardiovascular mortality in acute heart failure. N with poor long-term outcomes in acute decompensated heart
Engl J Med 2017; 376: 1956–1964. failure. J Card Fail 2014; 20: 392–399.
7. Sacks CA, Jarcho JA and Curfman GD. Paradigm shifts in 24. Verbrugge FH, Dupont M, Bertrand PB, et al. Determinants
heart-failure therapy – a timeline. N Engl J Med 2014; 371: and impact of the natriuretic response to diuretic therapy in
989–991. heart failure with reduced ejection fraction and volume over-
8. Massie BM, O’Connor CM, Metra M, et al.; for the load. Acta Cardiol 2015; 70: 265–273.
PROTECT Investigators and Committees. Rolofylline, an 25. Ter Maaten JM, Rao VS, Hanberg JS, et al. Renal tubular
adenosine A1-receptor antagonist, in acute heart failure. N resistance is the primary driver for loop diuretic resistance in
Engl J Med 2010; 363: 1419–1428. acute heart failure. Eur J Heart Fail 2017; 19: 1014–1022.
9. Felker GM, Lee KL, Bull DA, et al. Diuretic strategies in 26. Valente MA, Voors AA, Damman K, et al. Diuretic response
patients with acute decompensated heart failure. N Engl J in acute heart failure: clinical characteristics and prognostic
Med 2011; 364: 797–805. significance. Eur Heart J 2014; 35: 1284–1293.
10. O’Connor CM, Starling RC, Hernandez AF, et al. Effect of 27. Voors AA, Davison BA, Teerlink JR, et al.; for the RELAX–
nesiritide in patients with acute decompensated heart failure. AHF Investigators. Diuretic response in patients with acute
N Engl J Med 2011; 365: 32–43. decompensated heart failure: characteristics and clinical
388 European Heart Journal: Acute Cardiovascular Care 7(4)

outcome – an analysis from RELAX–AHF. Eur J Heart Fail 45. Amlal H and Soleimani M. Pendrin as a novel target for diu-
2014; 16: 1230–1240. retic therapy. Cell Physiol Biochem 2011; 28: 521–526.
28. er Maaten JM, Dunning AM, Valente MA, et al. Diuretic 46. Verbrugge FH, Grieten L and Mullens W. New insights into
response in acute heart failure – an analysis from ASCEND– combinational drug therapy to manage congestion in heart
HF. Am Heart J 2015; 170: 313–321. failure. Curr Heart Fail Rep 2014; 11: 1–9.
29. Aronson D and Burger AJ. Diuretic response: clinical and 47. Dupont M, Mullens W, Finucan M, et al. Determinants of
hemodynamic predictors and relation to clinical outcome. J dynamic changes in serum creatinine in acute decompen-
Card Fail 2016; 22: 193–200. sated heart failure: the importance of blood pressure reduc-
30. Damman K, Valente MA, Voors AA, et al. Renal impairment, tion during treatment. Eur J Heart Fail 2013; 15: 433–440.
worsening renal function, and outcome in patients with heart 48. Cotter G, Metzkor E, Kaluski E, et al. Randomised trial of
failure: an updated meta-analysis. Eur Heart J 2014; 35: high-dose isosorbide dinitrate plus low-dose furosemide ver-
455–469. sus high-dose furosemide plus low-dose isosorbide dinitrate
31. McCrindle JL, Li Kam Wa TC, et al. Effect of food on the in severe pulmonary oedema. Lancet 1998; 351: 389–393.
absorption of frusemide and bumetanide in man. Br J Clin 49. Mullens W, Abrahams Z, Francis GS, et al. Sodium nitro-
Pharmacol 1996; 42: 743–746. prusside for advanced low-output heart failure. J Am Coll
32. Vasko MR, Cartwright DB, Knochel JP, et al. Furosemide Cardiol 2008; 52: 200–207.
absorption altered in decompensated congestive heart fail- 50. Mentz RJ, Stevens SR, DeVore AD, et al. Decongestion strat-
ure. Ann Intern Med 1985; 102: 314–318. egies and renin-angiotensin-aldosterone system activation in
33. Verbrugge FH, Dupont M, Steels P, et al. Abdominal contri- acute heart failure. JACC Heart Fail 2015; 3: 97–107.
butions to cardiorenal dysfunction in congestive heart fail- 51. Verbrugge FH, Tang WH and Mullens W. Renin–angiotensin–
ure. J Am Coll Cardiol 2013; 62: 485–495. aldosterone system activation during decongestion in acute
34. Hasegawa M, Kusuhara H, Adachi M, et al. Multidrug resist- heart failure: friend or foe? JACC Heart Fail 2015; 3: 108–111.
ance-associated protein 4 is involved in the urinary excretion 52. Verbrugge FH, Duchenne J, Bertrand PB, et al. Uptitration of
of hydrochlorothiazide and furosemide. J Am Soc Nephrol renin–angiotensin system blocker and beta-blocker therapy
2007; 18: 37–45. in patients hospitalized for heart failure with reduced versus
35. Loon NR and Wilcox CS. Mild metabolic alkalosis impairs preserved left ventricular ejection fractions. Am J Cardiol
the natriuretic response to bumetanide in normal human sub- 2013; 112: 1913–1920.
jects. Clin Sci (Lond) 1998; 94: 287–292. 53. van Vliet AA, Donker AJ, Nauta JJ, et al. Spironolactone in
36. Uwai Y, Saito H, Hashimoto Y, et al. Interaction and trans- congestive heart failure refractory to high-dose loop diuretic
port of thiazide diuretics, loop diuretics, and acetazolamide and low-dose angiotensin-converting enzyme inhibitor. Am J
via rat renal organic anion transporter rOAT1. J Pharmacol Cardiol 1993; 71: 21A–28A.
Exp Ther 2000; 295: 261–265. 54. Ponikowski P, Voors AA, Anker SD, et al.; and Authors/
37. Kirchner KA, Voelker JR and Brater DC. Intratubular albu- Task Force Members. 2016 ESC Guidelines for the diag-
min blunts the response to furosemide – a mechanism for nosis and treatment of acute and chronic heart failure: the
diuretic resistance in the nephrotic syndrome. J Pharmacol Task Force for the diagnosis and treatment of acute and
Exp Ther 1990; 252: 1097–1101. chronic heart failure of the European Society of Cardiology
38. Mullens W, Abrahams Z, Skouri HN, et al. Elevated intra- (ESC) developed with the special contribution of the Heart
abdominal pressure in acute decompensated heart failure: a Failure Association (HFA) of the ESC. Eur Heart J 2016;
potential contributor to worsening renal function? J Am Coll 37: 2129–2200.
Cardiol 2008; 51: 300–306. 55. Knauf H and Mutschler E. Functional state of the nephron
39. Mullens W, Abrahams Z, Francis GS, et al. Prompt reduction and diuretic dose-response – rationale for low-dose combina-
in intra-abdominal pressure following large-volume mechan- tion therapy. Cardiology 1994; 84 (Suppl. 2): 18–26.
ical fluid removal improves renal insufficiency in refractory 56. Verbrugge FH, Steels P, Grieten L, et al. Hyponatremia in
decompensated heart failure. J Card Fail 2008; 14: 508–514. acute decompensated heart failure: depletion versus dilution.
40. Nijst P, Cops J, Martens P, et al. Endovascular shedding mark- J Am Coll Cardiol 2015; 65: 480–492.
ers in patients with heart failure with reduced ejection frac- 57. Jentzer JC, DeWald TA and Hernandez AF. Combination of
tion. Microcirculation 2018; 25: doi: 10.1111/micc.12432 loop diuretics with thiazide-type diuretics in heart failure. J
41. Nijst P, Verbrugge FH, Grieten L, et al. The pathophysio- Am Coll Cardiol 2010; 56: 1527–1534.
logical role of interstitial sodium in heart failure. J Am Coll 58. Grodin JL, Stevens SR, de Las Fuentes L, et al.

Cardiol 2015; 65: 378–388. Intensification of medication therapy for cardiorenal syn-
42. Aliti GB, Rabelo ER, Clausell N, et al. Aggressive fluid and drome in acute decompensated heart failure. J Card Fail
sodium restriction in acute decompensated heart failure: 2016; 22: 26–32.
a randomized clinical trial. JAMA Intern Med 2013; 173: 59. Zahedi K, Barone S, Xu J, et al. Potentiation of the effect
1058–1064. of thiazide derivatives by carbonic anhydrase inhibitors:
43. Nijst P, Verbrugge FH, Martens P, et al. Plasma renin activity molecular mechanisms and potential clinical implications.
in patients with heart failure and reduced ejection fraction PLoS One 2013; 8: e79327.
on optimal medical therapy. J Renin Angiotensin Aldosterone 60. Soleimani M, Barone S, Xu J, et al. Double knockout of pen-
Syst 2017; 18: 1470320317729919. drin and Na-Cl cotransporter (NCC) causes severe salt wast-
44. Kim GH. Long-term adaptation of renal ion transporters to ing, volume depletion, and renal failure. Proc Natl Acad Sci
chronic diuretic treatment. Am J Nephrol 2004; 24: 595–605. USA 2012; 109: 13368–13373.
Verbrugge 389

61. Hanley T and Platts MM. Acetazolamide (diamox) in the 67. Costanzo MR, Negoianu D, Fonarow GC, et al. Rationale
treatment of congestive heart failure. Lancet 1956; 270: and design of the Aquapheresis Versus Intravenous Diuretics
357–359. and Hospitalization for Heart Failure (AVOID–HF) trial. Am
62. Knauf H and Mutschler E. Sequential nephron block-
Heart J 2015; 170: 471–482.
ade breaks resistance to diuretics in edematous states. J 68. Ter Maaten JM, Valente MA, Damman K, et al. Combining
Cardiovasc Pharmacol 1997; 29: 367–372. diuretic response and hemoconcentration to predict rehospi-
63. Verbrugge FH, Martens P and Mullens W. SGLT-2 inhibitors talization after admission for acute heart failure. Circ Heart
in heart failure: implications for the kidneys. Curr Heart Fail Fail 2016; 9: pii: e002845.
Rep 2017; 14: 331–337. 69. Aoki S, Okumura T, Sawamura A, et al. Usefulness of the
64. Ali SS, Olinger CC, Sobotka PA, et al. Loop diuretics can combination of in-hospital poor diuretic response and sys-
cause clinical natriuretic failure: a prescription for volume temic congestion to predict future cardiac events in patients
expansion. Congest Heart Fail 2009; 15: 1–4. with acute decompensated heart failure. Am J Cardiol 2017;
65. Costanzo MR, Guglin ME, Saltzberg MT, et al. Ultrafiltration 119: 2010–2016.
versus intravenous diuretics for patients hospitalized for 70. Kumar D and Bagarhatta R. Fractional excretion of

acute decompensated heart failure. J Am Coll Cardiol 2007; sodium and its association with prognosis of decompen-
49: 675–683. sated heart failure patients. J Clin Diagn Res 2015; 9:
66. Patarroyo M, Wehbe E, Hanna M, et al. Cardiorenal out- OC01–OC03.
comes after slow continuous ultrafiltration therapy in refrac- 71. Doering A, Jenkins CA, Storrow AB, et al. Markers of diu-
tory patients with advanced decompensated heart failure. J retic resistance in emergency department patients with acute
Am Coll Cardiol 2012; 60: 1906–1912. heart failure. Int J Emerg Med 2017; 10: 17.

You might also like