You are on page 1of 44

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/340603111

Diuretic Resistance in Heart Failure

Article  in  Cardiology in Review · March 2020


DOI: 10.1097/CRD.0000000000000310

CITATIONS READS
0 69

7 authors, including:

M Rubayat Rahman Pablo Paz


Texas Tech University Health Sciences Center Texas Tech University Health Sciences Center
23 PUBLICATIONS   22 CITATIONS    18 PUBLICATIONS   0 CITATIONS   

SEE PROFILE SEE PROFILE

Mohamed Elmassry Barbara Mantilla


Texas Tech University Texas Tech University
22 PUBLICATIONS   0 CITATIONS    19 PUBLICATIONS   5 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Internet Journals, Mega journals, and Ethical publishing View project

Native valve endocarditis with candida glaberata View project

All content following this page was uploaded by Kenneth Nugent on 09 May 2020.

The user has requested enhancement of the downloaded file.


`

The Cardiology in Review Journal Publish Ahead of Print

DOI: 10.1097/CRD.0000000000000310

Diuretic Resistance in Heart Failure

Rubayat Rahman MD1, Pablo Paz MD1, Mohamed Elmassry MD1, Barbara Mantilla MD1,
Downloaded from https://journals.lww.com/cardiologyinreview by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3POoLCqWplEckHH+R8Lafs3Va2y11oTUC5bT9K0im5bQ= on 05/09/2020

Logan Dobbe MD2, Scott Shurmur MD1, Kenneth Nugent MD1

D
1
Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock,

Texas; 2Department of Graduate Medical Education, Madigan Army Medical Center, Tacoma,

TE
Washington

Running Head: Diuretic Resistance in Heart Failure

Corresponding author: Kenneth Nugent MD, 3601 4th Street, Lubbock, Texas 79430
EP
Kenneth.Nugent@ttuhsc.edu,

Conflicts of interest: none

Financial support: none


C
C
A

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Abstract

Decompensated heart failure accounts for approximately one million hospitalizations in

the United States annually, and this number is expected to increase significantly in the near

future. Diuretics provide the initial management in most patients with fluid overload. However,

D
the development of diuretic resistance remains a significant challenge in the treatment of heart

failure. Due to the lack of a standard definition, the prevalence of this phenomenon remains

difficult to determine, with some estimates suggesting that 25% to 30% of patients with heart

TE
failure have diuretic resistance. Certain characteristics, including low systolic blood pressures,

renal impairment, and atherosclerotic disease, help predict the development of diuretic

resistance. The underlying pathophysiology is likely multifactorial, with pharmacokinetic


EP
alterations, hormonal dysregulation, and the cardiorenal syndrome having significant roles. The

therapeutic approach to this common problem typically involves increases in the diuretic dose

and/or frequency, sequential nephron blockade, and mechanical fluid movement removal with

ultrafiltration or peritoneal dialysis. Paracentesis is potentially useful in patients with intra-


C

abdominal hypertension.

Key words: heart failure, diuretics, diuretic resistance, renal dysfunction


C
A

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Congestive heart failure (CHF) is a leading cause of hospitalization in the United States.1

Loop diuretics remain the cornerstone in the management of patients with acute decompensated

heart failure and are used in up to 90% of hospitalized patients, despite the lack of evidence of

improved survival in these patients.1,2 Consequently, diuretic resistance has become an important

concern since it is associated with deteriorating renal function and more frequent readmissions.3

D
Diuretic resistance seems to develop as a compensatory response to chronic diuretic treatment.

These responses include increases in plasma renin activity, stimulation of the sympathetic

TE
nervous system, and adaptive changes in nephron structure and function resulting from diuretic-

induced increases in the distal sodium load.4

DEFINITION OF DIURETIC RESISTANCE


EP
Despite its prevalence, the term diuretic resistance does not have a standard well-

accepted definition. Definitions include failure to eliminate extracellular fluid despite an

adequate trial of diuretics,5 or a decreased response to diuretics leading to persistent congestion,


6,7
or a fractional sodium excretion of <0.2% while on diuretics.8 Doering et al tried to define
C

diuretic resistance in patients with heart failure presenting to an emergency department. These

investigators compared three different urine parameters as markers of a poor diuretic response,
C

namely a fractional excretion of sodium of less than 0.2%, a spot urinary sodium less than 50

meq/L, and a urine sodium/potassium ratio less than 1.0. The patient populations identified by
A

each of the three parameters differed significantly and had very little overlap; however, the study

did find significantly higher rates of readmission in patients with spot urinary sodium less than

50 meq/L.9

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

PATIENT CHARACTERISTICS

Valente et al studied diuretic responses in 1,745 hospitalized patients with acute heart

failure and found that low systolic blood pressures, low serum potassium, high blood urea

nitrogen, diabetes, and atherosclerotic disease predicted a poor diuretic response.10 McClellan et

D
al reported that 38% of patients with CHF have impaired renal function, placing them at

increased risk of developing diuretic resistance.11 Prerenal azotemia is present to some degree in

almost all patients with severe CHF.6 This condition involves diminished renal blood flow which

TE
reduces tubular delivery of loop diuretics and increases competition from accumulating anions

for the organic anion transporter.12

EPIDEMIOLOGY
EP
An acute heart failure registry reported that there are an estimated 1 million

hospitalizations with a primary diagnosis of CHF annually, and this number is expected to

increase over the next 2 decades.1 Ninety percent of patients admitted with decompensated heart

failure receive intravenous diuretics and have an average hospitalization lasting for 4.3 days.
C

However, 50% of admitted patients lose less than five pounds, 20% actually gain weight during

hospitalization, and 42% of patients leave the hospital with persistent symptoms. Inadequate
C

diuresis contributes to readmissions.1

Due to the lack of a standard definition, it is difficult to determine the exact frequency of
A

diuretic resistance, with some estimates placing the number at 25% to 30% of patients.4 Most

hospitalizations for heart failure are due to fluid retention in patients refractory to oral diuretics.

Neuberg et al found that almost 35% of patients had diuretic resistance, defined as requirement

of furosemide > 80 mg daily or bumetanide > 2 mg daily for treatment.13

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

PATHOPHYSIOLOGY

Pharmacokinetic Factors

Brater et al demonstrated that the time for diuretics to reach a steady state following oral

administration is increased in patients with CHF compared to healthy controls.14 This is most

D
likely explained by mucosal edema in the gastrointestinal tract of patients with severe heart

failure.15 More than 95% of furosemide that enters the bloodstream binds to albumin, and it has

been suggested that exogenous albumin could increase diuretic delivery to the kidneys. However,

TE
a recent meta-analysis did not reveal any significant clinical benefit with the co-administration of

furosemide and albumin in hypoalbuminemic patients.16

Furosemide is secreted into the convoluted tubules by the organic anion transporter and
EP
then blocks the Na-K-2Cl transporter on the luminal surface of the ascending limb of the loop of

Henle.17 In patients with CHF, renal impairment leads to the accumulation of endogenous

organic anions which compete with loop diuretics for secretion by binding to the organic anion

transporter.18 This can reduce intratubular concentrations of diuretics, potentially contributing to


C

diuretic resistance.15

Diuretic resistance may also be caused by interactions with medications, such as


C

nonsteroidal anti-inflammatory agents (NSAIDs). In severe CHF, prostaglandins help maintain

renal perfusion and promote salt and water excretion.19 Nonsteroidal anti-inflammatory agents
A

interfere with this process by inhibiting prostaglandin synthesis, thus reducing renal perfusion

and diuresis. Several studies have reported smaller reductions in mortality with conventional

heart failure treatment in patients taking aspirin.20-22

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Hyponatremia and Post-Diuretic Salt Retention

Hyponatremia has been frequently associated with reduced diuretic efficacy due to

diminished distal tubular sodium delivery and secondary hyperaldosteronism. Low sodium levels

are a marker for advanced heart failure and are associated with decreased survival.23

D
Hyponatremia can be caused by diuretic treatment but is more often secondary to severe heart

failure’s stimulating thirst and the vasopressin axis that ultimately reduces excretion of free

water.24,25 This is particularly important in patients who are not adherent to fluid and salt intake

TE
restrictions. Wilcox et al reported that in healthy subjects eating a high-sodium diet there is a

marked increase in sodium absorption 6 to 24 hours after administration of furosemide. This

phenomenon is referred to as post-diuretic salt retention and can prevent the negative sodium
EP
balance expected with diuresis. In contrast, with a low-sodium diet, this compensatory increase

in sodium reabsorption did not occur, because sodium reabsorption had been near maximal at

baseline, thus furosemide given once daily led to negative sodium balance.26 Dietary

noncompliance should be suspected in patients on diuretics, when sodium excretion is greater


C

than 100 mmol in 24 hours without any associated weight loss.27

Renal Blood Flow


C

Increased proximal convoluted tubule sodium reabsorption as a consequence of diuretic-

induced volume depletion and decreased renal blood flow is a possible explanation of diuretic
A

resistance. Diuretic administration can reduce the glomerular filtration rate (GFR) and increase

proximal fluid reabsorption at least over short study periods.28 However, this theory has been

questioned by the work of Loon et al.29 Following the work of Earley and Martino,30 Loon

assumed that a combination of loop diuretics and thiazides would block the major distal sites of

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

sodium reabsorption, and any residual sodium reabsorption could then be attributed to the

proximal convoluted tubule. They treated patients on loop diuretics and thiazides with different

test combinations of intravenous furosemide, thiazides, and spironolactone and found no

significant difference in sodium excretion compared to placebo, indicating that proximal

D
reabsorption was not greatly affected by diuretic therapy in chronically treated patients.

Greenwood et al actually found that infusions of loop diuretics reduced proximal fluid

reabsorption and attributed this effect to non-specific actions of loop diuretics, including

TE
inhibition of carbonic anhydrase.31

Hormonal Dysregulation

Volume depletion secondary to diuretics stimulates the renin-angiotensin-aldosterone


EP
system (RAAS), and this may contribute to the development of diuretic resistance. As the heart

fails, renal blood flow is initially maintained by autoregulation in the kidneys. Activation of the

RAAS and the sympathetic nervous system (SNS) preserves GFR. This is achieved by an

increase in efferent arteriolar resistance with subsequent augmentation of glomerular hydrostatic


C

pressure and filtration fraction.32 In advanced heart failure, when renal perfusion falls below the

threshold of renal autoregulation, GFR becomes dependent on afferent and efferent arteriolar
C

resistance controlled by norepinephrine and angiotensin II.33 Upregulation of RAAS becomes a

maladaptive process since both systemic and renal vasoconstriction further reduce renal blood
A

flow, increase tubular sodium reabsorption, and increase congestion. However, this may not be

a major factor in diuretic resistance at least in patients with CHF since RAAS activity is already

elevated at baseline. Moreover, the convoluted tubules are the major sites of reabsorption in the

nephron, making it unlikely that aldosterone-induced reabsorption in the collecting tubules can

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

overcome the natriuresis induced by loop diuretics or thiazides. Loon et al demonstrated that

addition of an aldosterone antagonist did not restore urinary sodium excretion in patients treated

with furosemide to that seen in patients on placebo.29

The SNS and RAAS increase sodium and water reabsorption in the proximal tubule,

D
which leads to passive reabsorption of urea and a decrease in urine flow rate. Furthermore, as

GFR declines, vasopressin upregulates urea transporters in the collecting duct leading to an

additional increase in urea reabsorption. These mechanisms can help explain why the level of

TE
blood urea nitrogen (BUN) correlates with the degree of neuro-hormonal activation and is

associated with worsening heart failure.34 Therefore, the BUN can function as an indicator of

neuro-hormonal activation, reflects changes in GFR, and has a prognostic value in patients with
EP
heart failure.35,36

Testani et al analyzed 2,456 patients with compensated heart failure in the BEST trial

(Beta-blocker evaluation of Survival Trial) to determine if BUN could be used as a marker of

adverse effects of loop diuretics.37 The study classified patients into subgroups based on a
C

baseline BUN level above or below the median (21 mg/dL) to study the relationship between

high dose loop diuretics (HDLD) and mortality. HDLD treatment (defined as total daily dose of
C

≥160 mg of furosemide equivalents) was associated with higher mortality rates in patients with a

BUN above 21 mg/dL (hazard ratio [HR] 1.30; 95% confidence interval [CI]: 1.07 to 1.5;
A

p=0.009) but improved survival in patients with a BUN below 21 mg/dL (HR: 0.71; 95% CI:

0.49 to 0.96, p =0.0042) after controlling for confounding variables.37 Patients taking HDLDs

had higher plasma norepinephrine levels with a baseline BUN above the median compared to

patients below the median. These findings suggest a link between the BUN level and renal

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

neuro-hormonal activation that can help identify patients who will have adverse effects with loop

diuretics.37

Nuñez et al also studied the association between mortality and HDLD based on BUN and

carbohydrate antigen 125 (CA 125) levels following hospitalization in acute heart failure.38 The

D
CA 125 is used as a marker of systemic congestion, and in patients with normal CA 125 levels,

HDLDs (defined by ≥120 mg/day in furosemide equivalents) were associated with higher

mortality rates in patients with BUNs above the median but not with BUNs below the median.

TE
These findings support the results reported by Testani et al37 in patients with stable chronic heart

failure. However, in patients with significant volume overload (i.e., CA 125 > 35 U/mL),

HDLDs improved survival in patients with high BUNs and decreased survival in patients with
EP
low BUNs.38 These results suggest that the degree of fluid overload also has an important role in

the relationship between BUN and adverse diuretic effects.

Structural Changes in the Renal Parenchyma

Diuretic treatment causes structural changes in the nephron itself. Continuous infusions
C

of furosemide for 6 days in rats stimulate the proliferation of the distal convoluted tubules with

expansion of mitochondrial volume and basolateral membrane area. This suggests that the
C

transcellular transport capacity of the distal convoluted tubules increases in response to

diuretics.39 Na and coworkers used immunoblotting to demonstrate proliferation of sodium


A

channels in the ascending loop of Henle, the distal convoluted tubule, and collecting tubules

following continuous treatment with either loop and thiazide diuretics.40 Pendrin, a Cl-/HCO3-

cotransporter in the distal convoluted tubule, is upregulated in response to diuretic therapy in

mice; urine output increases 60% when this transporter is blocked.41 The carbonic anhydrase

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

inhibitor acetazolamide downregulates pendrin and can potentiate the diuretic effect of other

diuretics.42 However, carbonic anhydrase inhibitors should be used cautiously, if at all, in

patients with heart failure since these drugs act on the proximal convoluted tubule and can cause

metabolic acidosis.18

D
Renal Vein Hypertension and the Cardiorenal Syndrome

Patients with cardiorenal syndrome (CRS) have worsening renal function and diuretic

resistance. The syndrome can be divided into categories based on the organ affected first (kidney

TE
or heart) and on the acuity and nature of the insult.43 The pathophysiology is multifactorial and

is explained by the interaction of altered hemodynamics, hormonal dysregulation, and oxidative

stress. Both reduced renal blood flow and increased renal vein pressure contribute to the
EP
development of CRS.44 Central venous pressure (CVP) is an independent predictor of renal

function in CHF.45,46 In combination with pump failure, elevated CVP directly impairs renal

blood flow and renal perfusion pressure. As renal blood flow drops below the threshold of renal

autoregulation, renal perfusion pressure becomes directly pressure dependent, and any further
C

increase in CVP leads to progressive fluid extravasation from the renal microcirculation.

Increased interstitial pressure causes renal parenchymal hypoxia and tubular dysfunction with
C

activation of the renin-angiotensin-aldosterone system.47 This high degree of neurohormonal

activation causes pre-glomerular vasoconstriction and reduces GFR which reduces the kidneys’
A

response to diuretics. The use of diuretics to reduce fluid overload could further compromise

renal blood flow and GFR.

Mullens et al studied 145 patients with severe heart failure managed in an intensive care

unit with right heart catheterization.48 They found that patients with worsening renal function

10

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

defined by an increase in Cr of 0.3 mg/dL had higher CVPs on admission (18 ± 7 mm Hg vs. 12

± 6 mm Hg, p < 0.001) and after intensive medical therapy (11 ± 8 mm Hg vs. 8 ± 5 mm Hg, p =

0.04) than patients who did not have a deterioration in renal function. These results suggest that

venous congestion defined by an increase in CVP has an important role in the development of

CRS.48 A retrospective review of 2,557 patients who underwent right heart catheterization

D
showed a strong association between CVP and eGFR (r = −0.108, p < 0.0001) in patients with

both reduced and preserved cardiac function. In particular, the magnitude of the relationship

TE
between CVP and eGFR was greater in patients with relatively normal cardiac indices. This

study showed that all-cause mortality increases with increasing CVP, particularly with pressures

above 6 mm Hg.45 Elevated CVP was an independent predictor of death (hazard ratio: 1.03 per
EP
mm Hg increase, 95% confidence interval: 1.01 to 1.05, p = 0.0032).45 The Evaluation Study of

Congestive Heart Failure and Pulmonary Artery Catheterization Effectiveness (ESCAPE) trial

with 433 patients admitted with acute decompensated heart failure demonstrated that baseline

serum creatinine correlated with baseline right atrial pressures and not the cardiac index.49
C

Furthermore, there was no improvement in renal function with improvement in cardiac systolic

function.33,50 These results suggest that elevated CVPs have an important role in the development
C

of CRS and suggest that treatment targeted to reduce venous congestion might improve renal

function, diuretic response, and survival.


A

Abdominal Compartment Syndrome

Abdominal congestion occurs in a significant percentage of patients with severe heart

failure. Intra-abdominal pressure (IAP) is increased in 60% of patients admitted with advanced

CHF.44 Splanchnic capacitance veins contain 25% of the total blood volume and are able to

11

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

buffer changes in volume status to maintain an adequate preload.44 Systemic congestion and

increased arteriolar vasoconstriction cause blood to shift from the effective circulatory volume to

splanchnic capacitance veins.50 At the same time sympathetic stimulation causes

venoconstriction reducing splanchnic capacitance.44 Splanchnic venous and interstitial

D
congestion are present when both venous capacitance and lymph flow cannot compensate for

excess fluid causing increased IAP. Abdominal congestion in turn causes increased renal vein

pressure and reduced perfusion, leading to worsening renal function.44,51 Elevated IAP is

TE
transmitted trans-diaphragmatically and may cause elevated intrathoracic pressures independent

of venous return.52,53

Mullens et al studied forty patients with severe heart failure receiving intensive medical
EP
therapy and reported a strong association between elevated IAP and worsening renal impairment

(p=0.009).48 Furthermore, in patients with elevated IAPs at baseline, reducing IAP correlated

with improvement in renal function (r = 0.77, p < 0.001). This correlation was independent of

changes in hemodynamic measurements, such as right and left-sided filling pressures and cardiac
C

index. The authors suggested that a persistently elevated IAP is a possible explanation for the

worsening of renal function that is sometimes observed despite improved hemodynamics with
C

aggressive therapy of decompensated heart failure.54 In these patients, considered refractory to

aggressive therapy, mechanical fluid removal can improve renal function. A prospective study of
A

9 patients with advanced heart failure refractory to diuretics found that fluid removal by either

paracentesis or ultrafiltration reduced IAP (from 13 ± 4 mm Hg to 7 ± 2 mm Hg, p=0.001) and

improved renal function (serum creatinine from 3.4 ± 1.4 mg/dL to 2.4 ± 1.1 mg/dL, p=0.01),

likely by reducing abdominal congestion.55

12

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

The pathogenesis of diuretic resistance involves multiple potential factors and likely

differs in individual patients (Table 1).14,15,18,19,26,28,29,40,47,48

Management Strategies

Medication

D
The medical management of diuretic resistance requires a careful review of relevant

clinical and pharmacological factors and sequential, often additive, changes in drug therapy.56

Rule-Out Noncompliance

TE
The first step in managing diuretic resistance is to ascertain whether or not dietary salt

intake is being restricted. In the absence of sodium restriction (<100 mmol/day), post-diuretic

salt and water retention can completely abolish the negative balance produced by diuresis.
EP
Measurement of 24-hour urinary sodium excretion can help assess sodium intake. Poor

adherence to medical treatment should be suspected when the daily salt excretion is greater than

100 mmol without associated weight loss.27

NSAIDs are an important cause of diuretic resistance, as discussed above. These


C

medications have been associated with increased risk of hospital readmissions in patients with

pre-existing heart failure.57 Patients with persistent congestion despite adequate diuretic dosage
C

and adherence to salt and fluid restriction should be asked about the use of NSAIDs.

Changes in Dosage and Route of Administration


A

Increased diuretic dosage and frequency can help overcome diuretic resistance to some

degree. Gerlag et al showed that high-dose furosemide (250–4000 mg/day, oral or IV) improved

symptoms, weight loss, and renal function in severe CHF unresponsive to conventional diuretic

treatment.58 Increased frequency of administration (3 times daily or more) reduces diuretic-free

13

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

intervals and can overcome post-diuretic salt retention.27 Switching to the intravenous route

ensures maximum bioavailability and circumvents the possibility of poor gastrointestinal

absorption. In patients who fail high dose intravenous diuretic therapy, continuous infusions of

diuretics has been found to be a safe and effective option in several studies.59-61 An often

D
overlooked concern with high dose infusions of loop diuretics is the development of ototoxicity,

particularly in patients receiving other ototoxic agents (e.g., aminoglycosides).62

Diuretic Choice: Torsemide vs Furosemide

TE
Torsemide has more consistent bioavailability than furosemide regardless of the presence

of gastrointestinal mucosal edema.63 Studies have shown that torsemide can reduce cardiac

sympathetic tone and ameliorate myocardial fibrosis and ventricular remodeling.64,65 Miles et al
EP
did a meta-analysis of 8,127 patients with heart failure from 14 studies and found that torsemide

reduced readmission rates and improved New York Heart Association (NYHA) functional class

at 12 months. However, there was no significant difference in mortality between the two

diuretics.66 In separate analyses with 12,725 patients with CHF (4,580 from the Duke
C

Echocardiography Lab Database, 7,141 from the Acute Study of Clinical Effectiveness of

Nesiritide in Decompensated Heart Failure (ASCEND-HF) trial, 1004 from the Placebo-
C

controlled Randomized study of the selective A1 adenosine receptor antagonist rolofylline for

patients hospitalized with acute heart failure and volume Overload to assess Treatment Effect on
A

Congestion and renal funcTion (PROTECT) trial. Mentz et al found no difference in 30-day

mortality rates in patients treated with torsemide or furosemide after adjustment for baseline

disease severity and comorbidities.67-69

14

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Sequential Nephron Blockade

Combination therapy with loop and thiazide diuretics has been well-studied in CHF

patients. 70-72 Thiazides counteract the compensatory increase in transcellular sodium transport in

the distal convoluted tubule in response to loop diuretics. This approach, termed sequential

D
nephron blockade, can increase urine output, salt excretion, and weight loss and improve

symptoms in patients refractory to loop diuretics alone. There is no difference in efficacy among

the commonly used thiazides in combination with loop diuretics to achieve sequential nephron

TE
blockade.18

Vasopressin Receptor Blockade

Vasopressin levels are increased in patients with heart failure and can increase fluid
EP
retention.73 Selective antagonists of vasopressin at the V2 receptors located in the kidney, such as

tolvaptan, increase urine output and reduce body weight in patients with heart failure.74 The

Kanagawa Aquaresis Investigators Trial of Tolvaptan on Heart Failure Patients with Renal

Impairment (K-STAR) study demonstrated that in patients with heart failure and persistent
C

congestion and renal dysfunction refractory to standard therapy, the addition of tolvaptan was

superior to increased doses of furosemide and increased urine output without more renal
C

dysfunction.75 However, there were no significant differences in the decrease in body weight or

in the improvement in signs and symptoms of congestion.75 Udelson et al demonstrated that by


A

increasing water excretion tolvaptan reduced pulmonary capillary wedge pressures, right atrial

pressures, and pulmonary artery pressures, which should be associated with symptom

improvement.76 Wang et al published a systematic review and meta-analysis on the use of

tolvaptan in patients with acute heart failure.77 This study included 6 randomized control trials

15

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

with 746 patients. Tolvaptan did not improve all-cause mortality, did not decrease the incidence

of clinical events or worsening renal function, and did not decrease the length of hospital stays in

patients. In patients with diuretic resistance, tolvaptan does not seem to be significantly better

than adding a different class of diuretics; its use might be preferred in patients with diuretic

resistance associated with hyponatremia and/or kidney dysfunction.78

D
Aldosterone Antagonists

Spironolactone has a mild natriuretic effect and reduces potassium excretion by loop

TE
diuretics.79,80 Aldosterone antagonists decrease mortality and heart failure hospitalization in

patients with heart failure and reduced ejection fractions.56 Therefore, there is good evidence to

support the use of aldosterone antagonists when the GFR is stable and serum potassium levels
EP
are less than 5.5 mEq/l.56,79 In a trial of 100 patients with acute heart failure, adding

spironolactone was associated with rapid symptomatic relief and improved renal function.81 A

smaller study with 21 patients showed that the addition of spironolactone was associated with

marked diuresis and quicker symptom relief in patients refractory to loop diuretics.82 However,
C

in the Aldosterone Targeted Neurohormonal Combined with Natriuresis Therapy in Heart Failure

(ATHENA-HF) trial, adding high dose spironolactone to the usual care for patients with acute
C

heart failure did not improve outcomes.83 Larger trials are needed to study the potential benefit of

aldosterone antagonists in patients with diuretic resistance.


A

Dopamine

Low dose dopamine (<3 µg/kg/minute) works selectively on renal dopaminergic

receptors causing vasodilation of the renal vasculature and increased renal blood flow. The

Dopamine in Acute Decompensated Heart Failure II (DAD-HF II) trial and the Renal

16

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Optimization Strategies Evaluation (ROSE) trial showed that adding low-dose dopamine to low-

dose furosemide infusions was equally effective as high dose furosemide infusions.84,85 In

general, dopamine has no role in patients with acute heart failure and normal blood pressures

according to the available data.56,86,87 While dopamine has no proven benefit in diuretic

resistance, it may be used as a last resort in these patients given its theoretical benefits.56

D
Nesiritide

Nesiritide is a recombinant B-type natriuretic peptide that has favorable effects on

TE
hemodynamics and dyspnea in small clinical trials and is Food and Drug Administration

approved for symptomatic relief. However, larger clinical trials, such as the ROSE trial and the

ASCEND HF trial, reported no clear benefit of nesiritide in the management of diuretic


EP
resistance.85,88 The ASCEND-HF trial randomized 7,141 heart failure patients to receive

nesiritide or placebo with standard treatment with loop diuretics. This study reported mild

improvement of dyspnea in the nesiritide group but no significant change in urine output when

compared to placebo.89,90
C

The Low Dose Dopamine or Low Dose Nesiritide in Acute Heart Failure with Renal

Dysfunction (ROSE acute heart failure) trial compared both regimens to placebo but showed no
C

increase in urine output or improvement in renal function when added to diuretic therapy in

patients with acute heart failure and renal dysfunction.91 However, it is worth mentioning that
A

heart failure trials with nesiritide and dopamine were not focused on patients with diuretic

resistance.56

Hypertonic Saline

17

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Intravenous hypertonic saline can augment loop diuretic efficacy in refractory patients.

Hypertonic saline osmotically “pulls” free water from interstitial fluid into the renal vasculature

increasing renal blood flow, as well as improving sodium delivery to the loop of Henle, thus

restoring some of the loop diuretics’ effect.92

D
Several studies showed that adding hypertonic saline to loop diuretic therapy improves

diuresis, improves renal function, and shortens hospitalizations.93,94 The Sodium Management in

Acute and Chronic Heart Failure trial compared the short- and long-term effects of intravenous

TE
furosemide plus hypertonic saline with intravenous furosemide alone in 1,771 decompensated

NYHA functional class III patients. The addition of hypertonic saline infusions improved

diuresis and reduced hospital stays compared to furosemide alone. Moreover, after a follow-up
EP
of 57 months, the furosemide plus hypertonic saline patients had fewer hospital readmissions

(18.5% vs 34.2%; P < 0.0001) and a lower mortality rate (12.9% vs 23.8%; P < 0.0001)

compared with the group that received furosemide alone.95

Management Strategies: Medical Devices


C

Ultrafiltration

The 2013 American College of Cardiology Foundation/American Heart Association


C

(ACCF/AHA) Task Force Guidelines recommend ultrafiltration as a possible treatment option

for patients with refractory congestion who are not responding to medical therapy.96 Standard
A

ultrafiltration is a cumbersome process, requiring placement of a dual lumen central venous line,

and has several complications, including infection, hemorrhage, electrolyte disorders, anemia,

and thrombocytopenia.97 With technological advances, newer ultrafiltration devices have become

available that are smaller and more portable and require only peripheral venous access.98

18

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

The data which support the efficacy of ultrafiltration in comparison to standard diuresis

are inconsistent. Two randomized controlled trials with 240 patients in total, the UNLOAD

(Ultrafiltration versus Intravenous Diuretics for Patients Hospitalized for Acute Decompensated

Congestive Heart Failure) trial and the RAPID-CHF (Relief for Acutely Fluid-Overloaded

D
Patients with Decompensated Congestive Heart Failure) trial, reported greater weight loss and

fluid loss with ultrafiltration than with intravenous diuretics.99,100 Neither trial specifically tested

the efficacy of ultrafiltration in patients resistant to diuretics. The ULTRAfiltration vs. DIureticS

TE
on clinical, biohumoral and haemodynamic variables in patients with deCOmpensated heart

failure (ULTRADISCO) study recruited 30 patients with decompensated heart failure and found

significant improvement in symptoms, plasma aldosterone levels, and hemodynamics with


EP
ultrafiltration compared to diuretic therapy.101 The Cardiorenal Rescue Study in Acute

Decompensated Heart Failure (CARRESS-HF) trial studied 188 patients with severe heart failure

and cardiorenal syndrome and found ultrafiltration inferior to step diuretic therapy based on two

end points: changes in serum creatinine and body weight at 96 hours.97 In addition, the
C

ultrafiltration arm had a higher risk of serious adverse events (72 % vs. 57 %). This trial was

unblinded and limited the ultrafiltration rate to 200 mL/hr; other studies allowed ultrafiltration
C

rates of up to 500 mL/hr. Teo et al retrospectively reviewed 44 patients with decompensated

heart failure and diuretic resistance and found greater fluid loss (3,815 mL vs. 1,355 mL, p =
A

0.0003) and greater weight loss (5.0 kg vs. 1.0 kg, p < 0.0001) at 48 hours with ultrafiltration

compared to standard care.102 The ultrafiltration group also had shorter hospitalizations (5.0 days

vs. 9.5 days, p = 0.0010) and fewer emergency department visits during a 90 day follow-up

period after discharge.

19

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Peritoneal Dialysis

Extracorporeal ultrafiltration can reduce fluid overload in hospitalized patients with acute

decompensated heart failure; however, its complexity and expense make it a poor option for

most patients with chronic heart failure during outpatient management. Peritoneal dialysis for the

treatment of resistant heart failure was first described in 1949 by Schneierson et al.103 Multiple

D
cohort studies since then have demonstrated the effectiveness of peritoneal dialysis in advanced

heart failure and cardiorenal syndrome. A 10-year retrospective study involving 21 patients

TE
found significantly reduced rate of all-cause hospitalizations in peritoneal dialysis patients

compared to baseline.104 Koch et al followed 118 patients with NYHA class III or IV heart

failure and chronic kidney disease and reported peritoneal dialysis was well tolerated with
EP
significant reductions in body weight after 6 months (78.7 to 74.7 kg).105 Another prospective

single center study followed seventeen patients with CHF undergoing peritoneal dialysis and

found a dramatic reduction in hospitalization rates (62 ± 16 to 11 ± 5 days/patient/year, P =

0.003) together with significant improvement in NYHA functional status and pulmonary artery
C

systolic pressure (44 ± 12 versus 27 ± 9 mmHg, P = 0.007) without changes in LVEF.106

Courivaud reported the outcomes in 126 patients treated with peritoneal dialysis for chronic
C

refractory heart failure. These patients had a mean age of 72 ± 11 and a mean estimated GFR of

33.5 ±15.1 mL/min/1.73 m². The mean time on peritoneal dialysis was 16 ±16 months.107
A

Patients with low ejection fractions had increases in ejection fraction during the first year of

peritoneal dialysis. The number of hospital days for acute decompensated heart failure was

reduced after the initiation of peritoneal dialysis. The number of days preceding dialysis was 3.3

± 2.6 days per patient-month compared to 0.3 ± 0.5 days after dialysis was started. The overall

20

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

1-year mortality was 42%. Peritoneal dialysis appears to decrease the rate of hospitalization for

decompensated heart failure and may allow beneficial adjustments in chronic medications used

in these patients.

Lu et al reported a systematic review of 21 studies on patients with refractory congestive

heart failure who underwent peritoneal dialysis for fluid management.108 This review included

D
673 patients with a mean age of 67.4 years and a mean follow-up time of 33.2 months. There

was a significant reduction in the number of hospital days from 6.3 to 1.2 days/year following

TE
induction of peritoneal dialysis. The left ventricular ejection fraction increased by approximately

4.1%, and the NYHA classification improved from 3.53 to 2.17 post dialysis. There was no

significant change in renal function in patients who did not have extremely low estimated GFRs
EP
at the onset of peritoneal dialysis. The mean incidence of peritonitis was 14.5 %/year; the

number of noninfectious complications was relatively low.

Patients with refractory fluid overload frequently require ultrafiltration to reduce fluid

accumulation. This typically requires hospitalization. The use of peritoneal dialysis at home can
C

provide the same benefit with less cost and less inconvenience. These patients typically undergo

nocturnal peritoneal dialysis 2-3 times per week. However, peritoneal dialysis has definite
C

limitations which include line migration and obstruction from infectious complications, such as

peritonitis and tunnel site infection, long-term metabolic derangements, and peritoneal sclerosis.
A

The ultrafiltration goal can be programmed specifically, but the ultrafiltration capacity of

peritoneal dialysis patients varies widely and requires close follow-up and frequent adjustments

over time.109 There is a need for large randomized controlled trials to assess the effectiveness and

21

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

utility of peritoneal dialysis compared to both conventional medical therapy and extracorporeal

ultrafiltration.

Peritoneal Catheters

Aisenberg reported the use of a peritoneal catheter for the removal of massive cardiac

ascites in one patient.110 This patient had a French pigtail catheter inserted in the peritoneal

D
cavity and had a daily drainage of 5 L ascites for 14 days. This patient then was discharged and

remained stable for at least 4 months. Bevan et al reported the use of a permanent peritoneal

TE
catheter for palliation of diuretic resistant cardiac ascites in one patient.111 This patient had

frequent hospitalizations for fluid accumulation and had required paracentesis once. This patient

performed home drainage of between 2 to 4 L of ascitic fluid per week. His peripheral edema
EP
resolved, his GFR increased, his pro BNP decreased, and he had no heart failure related

hospitalizations.

The accumulation of large volumes of ascitic fluid in some patients with chronic heart

failure likely has important consequences. The fluid volume can decrease mobility and quality
C

of life. In addition, the fluid likely increases intra-abdominal pressure and may compromise both

gastrointestinal function and renal function. Consequently, removal of this fluid using
C

paracentesis may have short term benefits. However frequent paracenteses present practical

difficulties. The use of long-term peritoneal catheters can allow this to take place at home at
A

lower costs. Many healthcare organizations use pleural catheters to manage malignant pleural

effusions. These organizations would have the experience and the staff needed to manage

outpatient peritoneal catheters.

Cardiac Resynchronization

22

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Some patients with advanced heart failure have both decreased left ventricular function

and cardiac dyssynchrony. Cleland and colleagues enrolled 813 patients in NYHA class III or

IV heart failure who had reduced left ventricular function and cardiac dyssynchrony into a

randomized control trial of medical therapy alone or medical therapy with cardiac

resynchronization.112 Patients in the cardiac resynchronization arm had better outcomes with a

D
decreased death from any cause and decreased unplanned hospitalization for cardiac events.

These patients also had reduced intraventricular mechanical delay, reduced end-systolic volume,

TE
and a reduced area of mitral regurgitant jet; they had increased left ventricular ejection fractions

(difference between means-6.9% at 18 months) and improved quality of life (NYHA class 2.1 vs.

27 at 90 days). This study demonstrates that patients who have significant cardiac dyssynchrony
EP
can benefit from cardiac resynchronization when added onto standard medical therapy.

Medical devices can improve outcomes in patients with diuretic resistance and refractory

fluid overload (Table 2).55,99-101,108,110-112

Management Strategies in Patients with Hypotension and or Hyponatremia


C

Management of Patients with Hypotension

Treatment for patients with acute heart failure and hypotension differs in patients with
C

heart failure with reduced ejection fractions (HFrEF) and in patients with heart failure with

preserved ejection fractions (HFpEF).


A

For patients with HFrEF associated with hypotension or clinical signs of shock and

evidence of an adequate preload, the addition of inotropic drugs (milrinone, dobutamine or

dopamine) is recommended.113 If worsening hypotension or new-onset tachyarrhythmia occur,

the dose of inotropes should be reduced or discontinued. If shock persists after adequate inotrope

23

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

use and optimization of filling pressures, vasopressors (norepinephrine and high-dose dopamine)

might increase perfusion to vital organs. But these drugs will increase left ventricular afterload.

In patients with significant decreases in left ventricular function and severe hemodynamic

compromise despite adequate pharmacologic therapy, mechanical cardiac support

D
(extracorporeal ventricular assist device, intra-aortic balloon pump or extracorporeal circulatory

membrane oxygenator) can be used.96,113

In patients with HFpEF, inotropes are contraindicated since dynamic left ventricular

TE
outflow tract obstruction, if present, can increase with the use of inotropes. These patients are

treated with beta blockers, and if there is no evidence of pulmonary congestion, cautious fluid

resuscitation. Vasopressors (phenylephrine and norepinephrine) can be added if hypotension


EP
persists after adequate hydration.114

Management of Patients with Hyponatremia

The presence of hyponatremia in patients with heart failure usually predicts a poor

prognosis; there is no evidence that correction of hyponatremia improves clinical outcomes.115


C

Thus, correction of hyponatremia is reserved for patients with a sodium level < 120 mEq/L,

presence of symptoms associated with hyponatremia, or asymptomatic hyponatremia that could


C

predispose patient to falls. The main therapy for hyponatremia in patients with heart failure is

water restriction.115 Angiotensin-converting enzyme inhibitors (ACE), angiotensin II receptor


A

blockers (ARB), and loop diuretics, typically used in patients with heart failure, can also increase

sodium levels. ACE inhibitors decrease water reabsorption by antagonizing the effect of

antidiuretic hormone (ADH) in the collecting tubules.116 The increased cardiac output with the

24

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

use of ACE inhibitors or ARB decreases the release of ADH.117 Loop diuretics reduce water

reabsorption by decreasing the concentration gradient in the renal medulla.118

Vasopressin receptor antagonists (tolvaptan and conivaptan) are approved for the

treatment of hyponatremia in patients with heart failure. These drugs improve sodium levels by

producing selective water diuresis.119 The Efficacy of Vasopressin Antagonism in Heart Failure

D
Outcome Study with Tolvaptan [EVEREST] study and the Study of Ascending Levels of

Tolvaptan in Hyponatremia 1 and 2 [SALT-1 and SALT-2] have demonstrated the efficacy of

TE
tolvaptan to increase sodium level in patients with heart failure. In the EVEREST outcome trial,

tolvaptan significantly increased sodium levels in the first week of treatment (5.5 vs. 1.9 mEq/L

with placebo).120 The SALT-1 and SALT-2 trials included patients with hyponatremia and either
EP
heart failure, syndrome of inappropriate antidiuretic hormone, or cirrhosis; tolvaptan increased

the sodium levels by 7.0 vs. 2.5 mEq/L when compared to placebo.121 The use of tolvaptan is

potentially limited by hepatic toxicity with increases liver enzymes by more than 2.5-fold in

some patients.122 Based on the previous finding, the Food and Drug Administration recommends
C

to not use tolvaptan for more than 30 days and to not use tolvaptan in patients with liver disease.

CONCLUSION
C

Despite its limitations, stepped pharmacologic therapy remains the mainstay of treatment

for diuretic resistance. This best approach begins with excluding medication nonadherence,
A

increasing the dose of diuretic or switching to continuous infusion, and eventually using

combination diuretic therapy. Measuring intra-abdominal pressure may help risk stratify patients,

and mechanical fluid removal with paracentesis, peritoneal dialysis, or ultrafiltration can

improve diuresis in patients with intra-abdominal hypertension. Ultrafiltration should be

25

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

considered in patients who fail treatment with high intensity sequential nephron blockade.

Resynchronization can improve outcomes in patients with reduced left ventricular function and

cardiac dyssynchrony. There remains a paucity of clinical trial evidence to help guide therapy in

these patients, and in particular, clinicians need simple laboratory tests to identify patients who

D
are unlikely to respond to diuretics. High BUN levels in combination with clinical assessment of

volume status can help identify patients at risk for adverse effects associated with diuretic

management.

TE
EP
C
C
A

26

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

REFERENCES

1. Adams KF, Jr, Fonarow GC, Emerman CL, et al. Characteristics and outcomes of

patients hospitalized for heart failure in the United States: rationale, design, and preliminary

observations from the first 100,000 cases in the Acute Decompensated Heart Failure National

D
Registry (ADHERE). Am Heart J 2005;149:209-16.

2. Sato N, Kajimoto K, Asai K, et al. Acute decompensated heart failure syndromes

(ATTEND) registry. A prospective observational multicenter cohort study: rationale, design, and

TE
preliminary data. Am Heart J 2010;159:949-55 e1.

3. Ronco C, Cicoira M, McCullough PA. Cardiorenal syndrome type 1: pathophysiological

crosstalk leading to combined heart and kidney dysfunction in the setting of acutely
EP
decompensated heart failure. J Am Coll Cardiol 2012;60:1031-42.

4. Ellison DH. Diuretic therapy and resistance in congestive heart failure. Cardiology

2001;96:132-43.

5. Reed BN, Devabhakthuni S. Diuretic resistance in acute decompensated heart failure: a


C

challenging clinical conundrum. Crit Care Nurs Q 2017;40:363-73.

6. Kramer BK, Schweda F, Riegger GA. Diuretic treatment and diuretic resistance in heart
C

failure. Am J Med 1999;106:90-6.

7. ter Maaten JM, Valente MA, Damman K, et al. Diuretic response in acute heart failure-
A

pathophysiology, evaluation, and therapy. Nat Rev Cardiol 2015;12:184-92.

8. Knauf H, Mutschler E. Sequential nephron blockade breaks resistance to diuretics in

edematous states. J Cardiovasc Pharmacol 1997;29:367-72.

27

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

9. Doering A, Jenkins CA, Storrow AB, et al. Markers of diuretic resistance in emergency

department patients with acute heart failure. Intern J Emerg Med 2017;10:17.

10. Valente MA, Voors AA, Damman K, et al. Diuretic response in acute heart failure:

clinical characteristics and prognostic significance. Eur Heart J 2014;35:1284-93.

D
11. McClellan WM, Flanders WD, Langston RD, et al. Anemia and renal insufficiency are

independent risk factors for death among patients with congestive heart failure admitted to

community hospitals: a population-based study. J Am Soc Neph 2002;13:1928-36.

TE
12. Risler T, Kramer B, Muller GA. The efficacy of diuretics in acute and chronic renal

failure. Focus on torasemide. Drugs 1991;41 Suppl 3:69-79.

13. Neuberg GW, Miller AB, O'Connor CM, et al. Diuretic resistance predicts mortality in
EP
patients with advanced heart failure. Am Heart J 2002;144:31-8.

14. Brater DC, Seiwell R, Anderson S, et al. Absorption and disposition of furosemide in

congestive heart failure. Kidney Intern 1982;22:171-6.

15. Brater DC. Resistance to loop diuretics. Why it happens and what to do about it. Drugs
C

1985;30:427-43.

16. Kitsios GD, Mascari P, Ettunsi R, et al. Co-administration of furosemide with albumin
C

for overcoming diuretic resistance in patients with hypoalbuminemia: a meta-analysis. J Crit

Care 2014;29:253-9.
A

17. Aronson D. The complexity of diuretic resistance. Eur J Heart Fail 2017;19:1023-6.

18. Shah N, Madanieh R, Alkan M, et al. A perspective on diuretic resistance in chronic

congestive heart failure. Therapeutic Advances Cardiov Dis 2017;11:271-8.

28

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

19. Koopmans PP, Thien T, Gribnau FW. Influence of non-steroidal anti-inflammatory drugs

on diuretic treatment of mild to moderate essential hypertension. British Med J (Clinical research

ed) 1984;289:1492-4.

20. Pfeffer MA, Braunwald E, Moye LA, et al. Effect of captopril on mortality and morbidity

D
in patients with left ventricular dysfunction after myocardial infarction. Results of the survival

and ventricular enlargement trial. The SAVE Investigators. N Engl J Med 1992;327:669-77.

21. Cleland JG, Erhardt L, Murray G, et al. Effect of ramipril on morbidity and mode of

TE
death among survivors of acute myocardial infarction with clinical evidence of heart failure. A

report from the AIRE Study Investigators. Eur Heart J 1997;18:41-51.

22. Hall D. Controversies in heart failure. Are beneficial effects of angiotensin-converting


EP
enzyme inhibitors attenuated by aspirin in patients with heart failure? Cardiol Clin 2001;19:597-

603.

23. Lee WH, Packer M. Prognostic importance of serum sodium concentration and its

modification by converting-enzyme inhibition in patients with severe chronic heart failure.


C

Circulation 1986;73:257-67.

24. Friedman E, Shadel M, Halkin H, et al. Thiazide-induced hyponatremia. Reproducibility


C

by single dose rechallenge and an analysis of pathogenesis. Ann Intern Med 1989;110:24-30.

25. Riegger GA, Liebau G, Kochsiek K. Antidiuretic hormone in congestive heart failure.
A

Am J Med 1982;72:49-52.

26. Wilcox CS, Mitch WE, Kelly RA, et al. Response of the kidney to furosemide. I. Effects

of salt intake and renal compensation. J Lab Clin Med 1983;102:450-8.

29

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

27. De Bruyne LK. Mechanisms and management of diuretic resistance in congestive heart

failure. Postgraduate Med J 2003;79:268-71.

28. Edwards BR, Baer PG, Sutton RA, et al. Micropuncture study of diuretic effects on

sodium and calcium reabsorption in the dog nephron. J Clin Invest 1973;52:2418-27.

D
29. Loon NR, Wilcox CS, Unwin RJ. Mechanism of impaired natriuretic response to

furosemide during prolonged therapy. Kidney International 1989;36:682-9.

30. Earley LE, Martino JA. Influence of sodium balance on the ability of diuretics to inhibit

TE
tubular reabsorption. A study of factors that influence renal tubular sodium reabsorption in man.

Circulation 1970;42:323-34.

31. Greenwood SL, White SJ, Green R. The effect of loop diuretics on fluid reabsorption
EP
from the rat proximal convoluted tubule. Expeerimental Physiology 1990;75:375-82.

32. Ronco C, Bellasi A, Di Lullo L. Cardiorenal syndrome: an overview. Adv Chronic

Kidney Dis 2018;25:382-90.

33. Nijst P, Mullens W. The acute cardiorenal syndrome: burden and mechanisms of disease.
C

Curr Heart Fail Rep 2014;11:453-62.

34. Lindenfeld J, Schrier RW. Blood urea nitrogen. J Am Coll Cardiol 2011;58:383-5.
C

35. Filippatos G, Rossi J, Lloyd-Jones DM, et al. Prognostic value of blood urea nitrogen in

patients hospitalized with worsening heart failure: insights from the acute and chronic
A

therapeutic impact of a vasopressin antagonist in chronic heart failure (ACTIV in CHF) study. J

Card Failure 2007;13:360-4.

36. Cauthen CA, Lipinski MJ, Abbate A, et al. Relation of blood urea nitrogen to long-term

mortality in patients with heart failure. Am J Cardiol 2008;101:1643-7.

30

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

37. Testani JM, Cappola TP, Brensinger CM, et al. Interaction between loop diuretic-

associated mortality and blood urea nitrogen concentration in chronic heart failure. J Am Coll

Cardiol 2011;58:375-82.

38. Núñez J, Núñez E, Miñana G, et al. Differential mortality association of loop diuretic

D
dosage according to blood urea nitrogen and carbohydrate antigen 125 following a

hospitalization for acute heart failure. Euro J Heart Failure 2012;14:974-84.

39. Kaissling B, Bachmann S, Kriz W. Structural adaptation of the distal convoluted tubule

TE
to prolonged furosemide treatment. Am J Physiology 1985;248:F374-81.

40. Na KY, Oh YK, Han JS, et al. Upregulation of Na+ transporter abundances in response to

chronic thiazide or loop diuretic treatment in rats. Am J Physiology Renal physiology 2003;284:
EP
F133-43.

41. Cil O, Haggie PM, Phuan PW, et al. Small-molecule inhibitors of pendrin potentiate the

diuretic action of furosemide. J Am Soc Neph : JASN 2016;27:3706-14.

42. Zahedi K, Barone S, Xu J, et al. Potentiation of the effect of thiazide derivatives by


C

carbonic anhydrase inhibitors: molecular mechanisms and potential clinical implications. PLoS

One 2013;8:e79327.
C

43. Ronco C, Haapio M, House AA, et al. Cardiorenal syndrome. J Am Coll Cardiol

2008;52:1527-39.
A

44. Verbrugge FH, Dupont M, Steels P, et al. Abdominal contributions to cardiorenal

dysfunction in congestive heart failure. J Am Coll Cardiol 2013;62:485-95.

31

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

45. Damman K, van Deursen VM, Navis G, et al. Increased central venous pressure is

associated with impaired renal function and mortality in a broad spectrum of patients with

cardiovascular disease. J Am Coll Cardiol 2009;53:582-8.

46. Damman K, Navis G, Smilde TD, et al. Decreased cardiac output, venous congestion and

D
the association with renal impairment in patients with cardiac dysfunction. Eur J Heart Fail

2007;9:872-8.

47. Guazzi M, Gatto P, Giusti G, et al. Pathophysiology of cardiorenal syndrome in

TE
decompensated heart failure: role of lung-right heart-kidney interaction. Intern J Cardiol

2013;169:379-84.

48. Mullens W, Abrahams Z, Francis GS, et al. Importance of venous congestion for
EP
worsening of renal function in advanced decompensated heart failure. J Am Coll Cardiol

2009;53:589-96.

49. Binanay C, Califf RM, Hasselblad V, et al. Evaluation study of congestive heart failure

and pulmonary artery catheterization effectiveness: the ESCAPE trial. JAMA 2005;294:1625-33.
C

50. Agrawal A, Naranjo M, Kanjanahattakij N, et al. Cardiorenal syndrome in heart failure

with preserved ejection fraction-an under-recognized clinical entity. Heart Fail Rev 2019;24:421-
C

37.

51. Nguyen VQ, Gadiraju TV, Patel H, et al. Intra-abdominal hypertension: an important
A

consideration for diuretic resistance in acute decompensated heart failure. Clin Cardiol

2016;39:37-40.

52. Barnes GE, Laine GA, Giam PY, et al. Cardiovascular responses to elevation of intra-

abdominal hydrostatic pressure. Am J Physiology 1985;248:R208-13.

32

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

53. Malbrain ML, Deeren D, De Potter TJ. Intra-abdominal hypertension in the critically ill:

it is time to pay attention. Current Opinion Crit Care 2005;11:156-71.

54. Mullens W, Abrahams Z, Skouri HN, et al. Elevated intra-abdominal pressure in acute

decompensated heart failure: a potential contributor to worsening renal function? J Am Coll

D
Cardiol 2008;51:300-6.

55. Mullens W, Abrahams Z, Francis GS, et al. Prompt reduction in intra-abdominal pressure

following large-volume mechanical fluid removal improves renal insufficiency in refractory

TE
decompensated heart failure. J Card Fail 2008;14:508-14.

56. Bowman BN, Nawarskas JJ, Anderson JR. Treating diuretic resistance. Cardiol Rev

2016;24:256-60.
EP
57. Feenstra J, Heerdink ER, Grobbee DE, et al. Association of nonsteroidal anti-

inflammatory drugs with first occurrence of heart failure and with relapsing heart failure: the

Rotterdam Study. Arch Intern Med 2002;162:265-70.

58. Gerlag PG, van Meijel JJ. High-dose furosemide in the treatment of refractory congestive
C

heart failure. Arch Intern Med 1988;148:286-91.

59. Lahav M, Regev A, Ra'anani P, et al. Intermittent administration of furosemide vs


C

continuous infusion preceded by a loading dose for congestive heart failure. Chest

1992;102:725-31.
A

60. van Meyel JJ, Smits P, Dormans T, et al. Continuous infusion of furosemide in the

treatment of patients with congestive heart failure and diuretic resistance. J Intern Med

1994;235:329-34.

33

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

61. Dormans TP, van Meyel JJ, Gerlag PG, et al. Diuretic efficacy of high dose furosemide

in severe heart failure: bolus injection versus continuous infusion. J Am Coll Cardiol

1996;28:376-82.

62. Rybak LP. Ototoxicity of loop diuretics. Otolaryn Clinics N Am 1993;26:829-44.

D
63. Felker GM, Mentz RJ. Diuretics and ultrafiltration in acute decompensated heart failure.

J Am Coll Cardiol 2012;59:2145-53.

64. Kasama S, Toyama T, Hatori T, et al. Effects of torasemide on cardiac sympathetic nerve

TE
activity and left ventricular remodelling in patients with congestive heart failure. Heart (British

Cardiac Society) 2006;92:1434-40.

65. Lopez B, Querejeta R, Gonzalez A, et al. Effects of loop diuretics on myocardial fibrosis
EP
and collagen type I turnover in chronic heart failure. J Am Coll Cardiol 2004;43:2028-35.

66. Miles JA, Hanumanthu BK, Patel K, et al. Torsemide versus furosemide and

intermediate-term outcomes in patients with heart failure: an updated meta-analysis. J

Cardiovascular Med (Hagerstown, Md) 2019;20:379-88.


C

67. Mentz RJ, Velazquez EJ, Metra M, et al. Comparative effectiveness of torsemide versus

furosemide in heart failure patients: insights from the PROTECT trial. Future Cardiol
C

2015;11:585-95.

68. Mentz RJ, Buggey J, Fiuzat M, et al. Torsemide versus furosemide in heart failure
A

patients: insights from Duke University Hospital. J Cardiovasc Pharmacol 2015;65:438-43.

69. Mentz RJ, Hasselblad V, DeVore AD, et al. Torsemide versus furosemide in patients

with acute heart failure (from the ASCEND-HF Trial). Am J Cardiol 2016;117:404-11.

34

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

70. Kiyingi A, Field MJ, Pawsey CC, et al. Metolazone in treatment of severe refractory

congestive cardiac failure. Lancet 1990;335:29-31.

71. Dormans TP, Gerlag PG. Combination of high-dose furosemide and hydrochlorothiazide

in the treatment of refractory congestive heart failure. Eur Heart J 1996;17:1867-74.

D
72. Channer KS, McLean KA, Lawson-Matthew P, et al. Combination diuretic treatment in

severe heart failure: a randomised controlled trial. Brit Heart J 1994;71:146-50.

73. Szatalowicz VL, Arnold PE, Chaimovitz C, et al. Radioimmunoassay of plasma arginine

TE
vasopressin in hyponatremic patients with congestive heart failure. N Engl J Med 1981;305:263-

6.

74. Gheorghiade M, Niazi I, Ouyang J, et al. Vasopressin V 2 -receptor blockade with


EP
tolvaptan in patients with chronic heart failure. Circulation 2003;107:2690-6.

75. Inomata T, Ikeda Y, Kida K, et al. Effects of additive tolvaptan vs. increased furosemide

on heart failure with diuretic resistance and renal impairment- results from the K-STAR Study.

Circ J 2017;82:159-67.
C

76. Udelson JE, Orlandi C, Ouyang J, et al. Acute hemodynamic effects of tolvaptan, a

vasopressin V2 receptor blocker, in patients with symptomatic heart failure and systolic
C

dysfunction: The ECLIPSE International, multicenter, randomized, placebo-controlled trial. J

Card Failure 2007;13:793-4.


A

77. Wang C, Xiong B, Cai L. Effects of Tolvaptan in patients with acute heart failure: a

systematic review and meta-analysis. BMC Cardiovasc Disord 2017;17:164.

78. Cox ZL, Hung R, Lenihan DJ, et al. Diuretic strategies for loop diuretic resistance in

acute heart failure. JACC: Heart Failure 2019.

35

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

79. Ellison DH. Diuretic therapy and resistance in congestive heart failure. Cardiol

2001;96:132-43.

80. Verbrugge FH, Mullens W, Tang WHW. Management of cardio-renal syndrome and

diuretic resistance. Curr Treat Opt Cardiovasc Med 2016;18:11

D
81. van Vliet A, Donker AJM, Nauta JJP, et al. Spironolactone in congestive heart failure

refractory to high-dose loop diuretic and low-dose angiotensin-converting enzyme inhibitor. Am

J Card 1993;71:A21-A8.

TE
82. Santos J, Planas R, Pardo A, et al. Spironolactone alone or in combination with

furosemide in the treatment of moderate ascites in nonazotemic cirrhosis. A randomized

comparative study of efficacy and safety. J Hepatology 2003;39:187-92.


EP
83. Butler J, Anstrom KJ, Felker GM, et al. Efficacy and safety of spironolactone in acute

heart failure. JAMA Cardiology 2017;2:950.

84. Giamouzis G, Butler J, Starling RC, et al. Impact of dopamine infusion on renal function

in hospitalized heart failure patients: results of the Dopamine in Acute Decompensated Heart
C

Failure (DAD-HF) Trial. J Card Failure 2010;16:922-30.

85. Wan S-H, Stevens SR, Borlaug BA, et al. Differential response to low-dose dopamine or
C

low-dose nesiritide in acute heart failure with reduced or preserved ejection fraction. Circulation:

Heart Failure 2016;9.


A

86. Vazir A, Cowie MR. Decongestion: Diuretics and other therapies for hospitalized heart

failure. Indian Heart J 2016;68:S61-S8.

87. Elkayam U, Ng TMH, Hatamizadeh P, et al. Renal vasodilatory action of dopamine in

patients with heart failure. Circulation 2008;117:200-5.

36

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

88. ter Maaten JM, Dunning AM, Valente MAE, et al. Diuretic response in acute heart

failure—an analysis from ASCEND-HF. Am Heart J 2015;170:313-21.e4.

89. O'Connor CM, Starling RC, Hernandez AF, et al. Effect of nesiritide in patients with

acute decompensated heart failure. N Engl J Med 2011;365:32-43.

D
90. Gottlieb SS, Voors A, Stebbins A, et al. Predictors of urine output and the effects of

nesiritide in acute decompensated heart failure: results from ASCEND-HF. J Am Coll Card

2013;61:E563.

TE
91. Chen HH, Anstrom KJ, Givertz MM, et al. Low-dose dopamine or low-dose nesiritide in

acute heart failure with renal dysfunction: the ROSE acute heart failure randomized trial. JAMA

2013;310:2533-43.
EP
92. De Vecchis R, Esposito C, Ariano C, et al. Hypertonic saline plus i.v. furosemide

improve renal safety profile and clinical outcomes in acute decompensated heart failure: A meta-

analysis of the literature. Herz 2015;40:423-35.

93. Licata G, Pasquale PD, Parrinello G, et al. Effects of high-dose furosemide and small-
C

volume hypertonic saline solution infusion in comparison with a high dose of furosemide as

bolus in refractory congestive heart failure: Long-term effects. Am Heart J 2003;145:459-66.


C

94. Paterna S, Di Pasquale P, Parrinello G, et al. Effects of high-dose furosemide and small-

volume hypertonic saline solution infusion in comparison with a high dose of furosemide as a
A

bolus, in refractory congestive heart failure. Euro J Heart Failure 2000;2:305-13.

95. Paterna S, Fasullo S, Cannizzaro S, et al. Short-term effects of hypertonic saline solution

in acute heart failure and long-term effects of a moderate sodium restriction in patients with

37

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

compensated heart failure with New York Heart Association Class III (Class C) (SMAC-HF

Study). Am J Med Sci 2011;342:27-37.

96. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management

of heart failure: a report of the American College of Cardiology Foundation/American Heart

D
Association Task Force on Practice Guidelines. J Am Coll Card 2013;62:e147-239.

97. Bart BA, Goldsmith SR, Lee KL, et al. Ultrafiltration in decompensated heart failure with

cardiorenal syndrome. N Engl J Med 2012;367:2296-304.

TE
98. Jaski BE, Ha J, Denys BG, et al. Peripherally inserted veno-venous ultrafiltration for

rapid treatment of volume overloaded patients. J Card Failure 2003;9:227-31.

99. Bart BA, Boyle A, Bank AJ, et al. Randomized controlled trial of ultrafiltration versus
EP
usual care for hospitalized patients with heart failure: preliminary report of the Rapid Trial. J

Card Failure 2004;10:S23.

100. Costanzo MR, Guglin ME, Saltzberg MT, et al. Ultrafiltration versus intravenous

diuretics for patients hospitalized for acute decompensated heart failure. J Am Coll Card
C

2007;49:675-83.

101. Giglioli C, Landi D, Cecchi E, et al. Effects of ULTRAfiltration vs. DIureticS on clinical,
C

biohumoral and haemodynamic variables in patients with deCOmpensated heart failure: the

ULTRADISCO study. Euro J Heart Failure 2011;13:337-46.


A

102. Teo LY, Lim CP, Neo CL, et al. Ultrafiltration in patients with decompensated heart

failure and diuretic resistance: an Asian centre’s experience. Singapore Med J2016;57:378-83.

103. Schneierson SJ. Continuous peritoneal irrigation in the treatment of intractable edema of

cardiac origin. Am J Med Sci 1949;218:76-9.

38

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

104. Cnossen TT, Kooman JP, Konings CJ, et al. Peritoneal dialysis in patients with primary

cardiac failure complicated by renal failure. Blood Purification 2010;30:146-52.

105. Koch M, Haastert B, Kohnle M, et al. Peritoneal dialysis relieves clinical symptoms and

is well tolerated in patients with refractory heart failure and chronic kidney disease. Euro J Heart

D
Failure 2012;14:530-9.

106. Sanchez JE, Ortega T, Rodriguez C, et al. Efficacy of peritoneal ultrafiltration in the

treatment of refractory congestive heart failure. Nephrology Dialysis Transplantation

TE
2009;25:605-10.

107. Courivaud C, Kazory A, Crépin T, et al. Peritoneal dialysis reduces the number of

hospitalization days in heart failure patients refractory to diuretics. Perit Dial Int 2014;34:100-8.
EP
108. Lu R, Muciño-Bermejo MJ, Ribeiro LC, et al. Peritoneal dialysis in patients with

refractory congestive heart failure: a systematic review. Cardiorenal Med 2015;5:145-56.

109. François K, Ronco C, Bargman JM. Peritoneal dialysis for chronic congestive heart

failure. Blood Purification 2015;40:45-52.


C

110. Aisenberg GM. Peritoneal catheter for massive cardiac ascites. BMJ Case Rep

2013;2013.
C

111. Bevan J, Penn LA, Mitchell AR. Permanent catheter drainage system for palliation of

diuretic-resistant cardiac ascites. ESC Heart Fail 2016;3:60-2.


A

112. Cleland JG, Daubert JC, Erdmann E, et al. The effect of cardiac resynchronization on

morbidity and mortality in heart failure. N Engl J Med 2005;352:1539-49.

113. McMurray JJ, Adamopoulos S, Anker SD, et al. ESC Guidelines for the diagnosis and

treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and

39

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology.

Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J

2012;33:1787-847.

114. Chockalingam A, Dorairajan S, Bhalla M, et al. Unexplained hypotension: The spectrum

D
of dynamic left ventricular outflow tract obstruction in critical care settings. Critical Care Med

2009;37:729-34.

115. Leier CV, Dei Cas L, Metra M. Clinical relevance and management of the major

TE
electrolyte abnormalities in congestive heart failure: hyponatremia, hypokalemia, and

hypomagnesemia. Am Heart J 1994;128:564-74.

116. Rouse D, Dalmeida W, Williamson FC, et al. Captopril inhibits the hydroosmotic effect
EP
of ADH in the cortical collecting tubule. Kidney International 1987;32:845-50.

117. Riegger GAJ, Kochsiek K. Vasopressin, renin and norepinephrine levels before and after

captopril administration in patients with congestive heart failure due to idiopathic dilated

cardiomyopathy. Am J Card1986;58:300-3.
C

118. Verbrugge FH, Steels P, Grieten L, et al. Hyponatremia in acute decompensated heart

failure. J Am Coll Card 2015;65:480-92.


C

119. Greenberg A, Verbalis JG. Vasopressin receptor antagonists. Kidney International

2006;69:2124-30.
A

120. Konstam MA. Effects of oral tolvaptan in patients hospitalized for worsening heart

failure. JAMA 2007;297:1319.

121. Schrier RW, Gross P, Gheorghiade M, et al. Tolvaptan, a selective oral vasopressin v2-

receptor antagonist, for hyponatremia. N Engl J Med 2006;355:2099-112.

40

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

122. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal

dominant polycystic kidney disease. N Engl J Med 2012;367:2407-18.

D
TE
EP
C
C
A

41

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Table 1 Pathogenesis of diuretic resistance

Alterations in Consequences
CHF
Pharmacokinetic Delayed drug Reduced Drug
factors absorption14,15 tubular secretion18 interactions, eg,

D
with NSAIDS19
Increased Post diuretic

TE
dietary salt Na absorption26
Reduced renal Reduced
blood flow GFR28
Activation of Renal Increased Na
RAAS/SNS29 vasoconstriction reabsorption
EP
Structural Increased Na
changes in the reabsorption
nephron40
Renal vein Reduced Increased Tubular
hypertension47 renal blood flow interstitial edema compression
C

Increased intra- Increased Decreased Decreased


abdominal pressure48 renal vein pressure perfusion pressure GFR
C

Na- sodium; GFR- glomerular filtration rate; RAAS- renin- angiotensin- aldosterone
system; SNS- sympathetic nervous system
A

42

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
`

Table 2 Device therapy in heart failure

Technique Use
Paracentesis55 Periodic use in patients with ascites and intra-
abdominal hypertension

D
Indwelling Either short-term or long-term use in patients with
peritoneal catheters110,111 ascites and intra-abdominal hypertension

TE
Peritoneal dialysis108 Long-term use in patients with fluid overload and
diuretic resistance
Ultrafiltration99,100,101 Hospital based use in patients with symptomatic fluid
overload and diuretic resistance
Resynchronization112 Long-term use in patients with reduced left ventricular
EP
ejection fraction and cardiac dyssynchrony
C
C
A

43

Copyright © 2020 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
View publication stats

You might also like