Clinical and Prognostic Significance of sST2 in Heart Failure

Clinical and Prognostic Significance of sST2 in Heart Failure

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 74, NO. 17, 2019 ª 2019 PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN COLLEGE OF CARDIOLOGY FOU...

1MB Sizes 0 Downloads 74 Views

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY

VOL. 74, NO. 17, 2019

ª 2019 PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

THE PRESENT AND FUTURE JACC REVIEW TOPIC OF THE WEEK

Clinical and Prognostic Significance of sST2 in Heart Failure JACC Review Topic of the Week Alberto Aimo, MD,a James L. Januzzi, JR, MD,b Antoni Bayes-Genis, MD, PHD,c Giuseppe Vergaro, MD, PHD,a,d Paolo Sciarrone, MD,d Claudio Passino, MD,a,d Michele Emdin, MD, PHDa,d

ABSTRACT Soluble suppression of tumorigenesis-2 (sST2) is released in response to vascular congestion and inflammatory and profibrotic stimuli, and is a strong, independent predictor of mortality and heart failure (HF) hospitalization in patients with acute or chronic HF. sST2 meets 2 fundamental criteria for clinically useful biomarkers: accurate, repeated measurements are available at a reasonable cost, and the biomarker provides information not already available from a careful clinical assessment. In particular, the prognostic value of sST2 is additive to natriuretic peptides and (in the case of chronic HF) to high-sensitivity troponin T. Nevertheless, the need for a multibiomarker approach to risk stratification and the role of sST2 as a guide to therapy decision-making remain to be established. Four years after a consensus document on sST2, and following major advances in the comprehension of the clinical value of this biomarker, the authors felt it worthwhile to reappraise current knowledge on sST2 in HF. (J Am Coll Cardiol 2019;74:2193–203) © 2019 Published by Elsevier on behalf of the American College of Cardiology Foundation.

L

aboratory diagnostics plays a major role in the

biomarkers, a growing body of evidence points to

diagnosis, risk stratification, therapy decision-

the clinical relevance of soluble suppression of

making, and monitoring of patients with heart

tumorigenesis-2 (sST2), which was first classified as

failure (HF). As proposed by Dr. Braunwald, bio-

an indicator of myocyte stress (1), but is mainly pro-

markers of HF can be classified according to the

duced in extracardiac tissues (2–4) in response to in-

main pathophysiological determinant of circulating

flammatory and fibrotic stimuli (5). Four years after

concentrations: inflammation, oxidative stress, extra-

the publication of a Consensus Document dedicated

cellular matrix remodeling, neurohormones, myocyte

to sST2 (6), and following major advances in our

injury, and myocyte stress (1). The most established

comprehension of the clinical value of this biomarker,

biomarkers are natriuretic peptides and troponins,

we update current knowledge on sST2 in HF with a

reflecting hemodynamic overload and cardiomyocyte

particular focus on its main application, that is,

damage, respectively. Besides these cardiac-specific

outcome prediction.

From the aInstitute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy; bMassachusetts General Hospital and Baim Institute for Clinical Research, Boston, Massachusetts; cHospital Universitari Germans Trias i Pujol, Badalona (Barcelona), CIBERCV, Listen to this manuscript’s

Barcelona, Spain; and the dCardiology Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy. Dr. Januzzi is supported

audio summary by

in part by the Hutter Family Professorship; is a Trustee of the American College of Cardiology; has received grant support from

Editor-in-Chief

Novartis Pharmaceuticals, Roche Diagnostics, Abbott, Singulex, and Prevencio; has received consulting income from Abbott,

Dr. Valentin Fuster on

Janssen, Novartis, Pfizer, Merck, Roche Diagnostics, and Critical Diagnostics; and participates in Clinical Endpoint Committees/

JACC.org.

Data Safety Monitoring Boards for Abbott, AbbVie, Amgen, Boehringer Ingelheim, Janssen, and Takeda. Dr. Bayes-Genis has received grant support from Roche Diagnosis; has received lecture honoraria from Roche Diagnostics and Critical Diagnostics; and has received consulting income from Roche Diagnostics, Critical Diagnostics, and Novartis. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose. David A. Morrow, MD, served as Guest Associate Editor for this paper. Manuscript received June 27, 2019; revised manuscript received August 7, 2019, accepted August 31, 2019.

ISSN 0735-1097/$36.00

https://doi.org/10.1016/j.jacc.2019.08.1039

2194

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

sST2 BIOLOGY

ABBREVIATIONS

HIGHLIGHTS

AND ACRONYMS

In 1989, ST2 was first identified by 2 inde-

HF = heart failure

pendent laboratories working on growth-

hs-TnT = high-sensitivity

stimulated fibroblasts (7,8). The definition

troponin T

“suppression of tumorigenesis” derives from

IL = interleukin LV = left ventricle/ventricular NT-proBNP = N-terminal

the hypothesis that ST2 might blunt uncontrolled cell proliferation; the researcher Shinichi Tominaga had previously identified

fragment of pro-brain natriuretic peptide

another protein called ST1 (9). Since the first

sST2 = soluble suppression of

description of ST2, it was noted that it

tumorigenesis-2

 sST2 is released in response to vascular congestion, inflammatory, and profibrotic stimuli.  sST2 is a strong predictor of outcome in HF.  The prognostic value of sST2 in HF is additive to natriuretic peptides.

had structural similarity to interleukin (IL)-1

variation at the gene locus predominantly regulates

receptors (7); hence, its alternative definition: “IL-1

ST2 expression (15). While ST2L is constitutively

receptor like-1.” Nonetheless, the function of this

expressed primarily in hematopoietic cells (Th2 and

protein remained unclear for several years.

mast cells), the expression of sST2 is largely induced

In 2002, Weinberg et al. (10) analyzed the expression of 7,000 genes in cardiomyocytes under-

in response to mechanical stretch and to inflammatory and pro-fibrotic stimuli (5).

going mechanical strain, and found that ST2 mark-

The major source of circulating sST2 in patients

edly increased as a response to this stimulus. In the

with HF has not been fully established, although

following years, IL-33 was identified as the ligand of

extracardiac production seems predominant: sST2

ST2 (11). IL-33 is mainly expressed by stromal cells

levels in HF are 10% higher in the peripheral venous

(fibroblasts, smooth muscle, epithelial, and endo-

blood, compared with the coronary sinus (2), and

thelial cells) (5), and signals to local immune cells the

there is no evident myocardial gradient of sST2 (16).

presence of tissue damage after exposure to patho-

Furthermore, in HF with preserved ejection fraction,

gens, injury-induced stress, or necrotic death (5).

sST2 concentrations are associated with systemic

Within the heart, IL-33 inhibits cardiomyocyte hy-

inflammation, right ventricular dysfunction, and

pertrophy and fibrosis in response to stimuli such as

systemic congestion, but not with indexes of left

angiotensin-II or phenylephrine, both in isolated

ventricular (LV) morphology or function (3,17). The

cardiomyocytes and in the murine model (12),

lungs have been proposed as an important site of sST2

possibly through the modulation of nuclear factor

production in HF (4). While the mechanisms of

kappa-light-chain-enhancer of activated B cells ac-

intracellular ST2L proteolysis have been clarified (18),

tivity (5,12). Furthermore, IL-33 reduces apoptosis by

to our knowledge the sites of degradation of sST2 and

induction of antiapoptotic factors and suppression of

the specific proteases involved have not been eluci-

caspase-3

dated so far.

activity

(13).

IL-33

exerts

these

car-

dioprotective effects by binding ST2-ligand (ST2L), which is a receptor expressed on plasma membrane

sST2 ASSAYS

of cardiomyocytes and fibroblasts, containing an extracellular domain of 3 linked immunoglobulin-

The assay methods for sST2 are thought to measure

like motifs, a transmembrane segment, and an

both free sST2 and sST2 complexed with IL-33,

intracellular Toll/IL-1 receptor cytoplasmic domain.

although it is not clear which epitopes are detected

sST2 is a circulating form of this receptor, which

by the antibodies against sST2 used for the different

lacks the transmembrane and cytoplasmic domains

methods (19). Among the 4 main ELISA assays (Pres-

and acts as a decoy receptor for IL-33 (5,12) (Central

age assay, Critical Diagnostics, San Diego, California;

Illustration).

MBL International, Woburn, Massachusetts; Ray

The gene named ST2 is located on human chro-

Biotech, Peachtree Corners, Georgia; R&D, Minneap-

mosome 2q12 and is part of the larger IL-1 gene clus-

olis, Minnesota), the Presage is the only one that has

ter. The ST2 gene has a proximal and a distal

received the Conformitè Européenne Mark and has

promoter; alternative promoter splicing and 3ʹ pro-

been cleared by the U.S. Food and Drug Administra-

cessing of the mRNA are believed to account for the

tion. Assay automation is expected in the near future,

production of either sST2 or ST2L (5,14). The mecha-

and should reduce dosage times considerably. As

nisms regulating the expression of sST2 and ST2L

established for the Presage assay, sST2 concentra-

have not been clarified (5), however common genetic

tions appear to be independent of fasting status,

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

C ENTR AL I LL U STRA T I O N Biology and Clinical Application of Suppression of Tumorigenesis-2

Aimo, A. et al. J Am Coll Cardiol. 2019;74(17):2193–203.

Soluble suppression of tumorigenesis-2 (sST2) is produced mostly by alveolar cells and vessel wall cells, and to a lower extent by cardiac fibroblasts and cardiomyocytes. Interleukin (IL)-33 is released by stromal cells in cardiac and extracardiac tissues; it binds membrane suppression of tumorigenesis-2 (ST2) receptors on cardiomyocytes and cardiac fibroblasts, blunting myocardial hypertrophy and fibrosis and inhibiting cardiomyocyte apoptosis. sST2 acts as a decoy receptor for IL-33, reducing these positive effects. sST2 measurement holds prognostic significance in acute or chronic heart failure (HF). NP ¼ natriuretic peptide.

2195

2196

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

T A B L E 1 Analytical Performance of Assays for Soluble ST2

Assay/Kit

Limit of Detection

Measurement Range

Intra-assay CV

Presage ST2 kit

1.3 ng/ml

Up to 200 ng/ml

<7%

<9%

Human ST2 ELISA kit

0.032 ng/ml

Up to 20 ng/ml

<6%

<6%

Human IL-1 R4/ST2 ELISA kit

0.002 ng/ml

Up to 1.2 ng/ml

<10%

<12%

ST2/IL-1 R4 DuoSet ELISA or Quantikine ELISA

0.005 ng/ml

Up to 2.0 ng/ml

<6%

<8%

Manufacturer

Critical Diagnostics (San Diego, California) MBL International (Woburn, Massachusetts) RayBiotech (Peachtree Corners, Georgia) R&D Systems (Minneapolis, Minnesota)

Interassay CV

Modified with permission from Mueller and Jaffe (19), 2015. CV ¼ coefficient of variation; ST2 ¼ suppression of tumorigenesis-2.

unaffected by freeze-thaw cycles, and with minimal

cardiac stretch, myocardial injury, and fibrosis; and

effects by hemolysis, lipemia, icterus, or rheumatoid

with abnormal echocardiographic parameters, but not

factor (19).

with renal function (25). Higher sST2 concentrations

The analytical characteristics of sST2 assay are

were associated with an elevated risk of incident HF

discussed in dedicated publications (19,20), and

and cardiovascular death, although the prognostic

summarized in Table 1. Notably, sST2 has a lower

significance

reference change value than B-type natriuretic pep-

NT-proBNP or both NT-proBNP and high-sensitivity

tide (BNP) or N-terminal pro–B-type natriuretic pep-

troponin T (hs-TnT) to the model (25). Among 3,428

tide (NT-proBNP) both in healthy subjects (21), and in

participants in the Framingham Heart Study, sST2

patients with chronic HF (22); therefore, variations in

levels predicted death, incident HF, and major car-

sST2 across serial measurements could reflect more

diovascular events independently from standard

closely changes in clinical status than variations in

cardiovascular risk factors, as well as from other

natriuretic peptides.

biomarkers of cardiovascular stress (growth-derived

sST2 TO DIAGNOSE HF OR PREDICT ITS DEVELOPMENT Reference values of plasma sST2 were defined by using the Presage assay on 3 cohorts of apparently healthy adult subjects: 1 European and 2 American (men: 4 to 31, 9 to 50, and 11 to 45 ng/ml; women: 2 to

resulted

attenuated

when

adding

factor-15, hs-TnI, BNP, hs-C-reactive protein) (26). In the Dallas Heart Study (n ¼ 3,294), sST2 was predictive

of

all-cause

and

cardiovascular

mortality

regardless of the presence of HF at baseline, although the predictive value for incident HF was not explored (27).

sST2 FOR RISK STRATIFICATION IN ACUTE HF

21, 7 to 33, and 9 to 35 ng/ml, respectively) (19). Additionally, data from the Framingham Heart Study

The prognostic significance of sST2 was first reported

provided important information regarding distribu-

in 2003. In a subanalysis of the PRAISE-2 (Prospective

tion and clinical correlates in a generally healthy,

Randomized

ambulatory population (23). Unlike cardiac-specific

study, conducted on 161 patients with HF and New

biomarkers such as natriuretic peptides, there is

York Heart Association functional class III or IV,

overlap among sST2 levels in healthy subjects, HF

baseline sST2 levels were correlated with natriuretic

patients, or people affected by inflammatory disor-

peptides, and their variation within 2 weeks was

ders (pneumonia, chronic obstructive pulmonary

predictive of all-cause mortality and need for heart

disease) (20). sST2 measurement is then not helpful

transplantation regardless of natriuretic peptide

to discriminate between cardiac and noncardiac dys-

levels (28). A strong prognostic value of sST2, inde-

pnea (24).

pendent from NT-proBNP, emerged in a cohort of 593

Amlodipine

Survival

Evaluation-2)

sST2 levels are lower in healthy women than men,

patients presenting at the emergency department

contrary to HF, where no significant sex-related dif-

with acute dyspnea (confirmed in the subset of 209

ferences in circulating concentrations and prognostic

patients with final HF diagnosis) (24), and in a pop-

significance of sST2 have been reported. In elderly,

ulation of 346 patients with acute HF (29). In the

community-dwelling individuals (n ¼ 3,915), circu-

latter study, mortality increased progressively with

lating sST2 was associated with older age, African-

sST2 deciles, exceeding 50% in the tenth decile; when

American race, and traditional cardiovascular risk

both ST2 and natriuretic peptides were elevated, the

factors; with other biomarkers of inflammation,

highest rates of death were observed in cumulative

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

2197

sST2 in Heart Failure

T A B L E 2 Proposed Cut-Offs of sST2 in Patients With Acute or Chronic Heart Failure and in Apparently Healthy Individuals

Study Type

N

Setting

Assay

Proposed sST2 Cut-Off (ng/ml)

Endpoints

Method for Cut-Off Selection

Original study

3,915

Elderly individuals

Presage

35

Incident HF, CV death

Cut-off previously proposed in CHF

Rehman et al. (29), 2008

Original study

346

AHF

Presage

49

All-cause death (1-yr)

Youden index

Pascual-Figal et al. (31), 2011

Original study

107

AHF

Presage

65

All-cause death

Youden index

Pascual-Figal et al. (30), 2019

Original study

316

AHF

Presage

35

All-cause death (1-yr)

Cut-off previously proposed in CHF

Original study

1,141

CHF

Presage

35

All-cause death or heart transplant

Youden index

Individual patient 4,268 data meta-analysis

CHF

Presage

28

All-cause and CV death, HF hospitalization

Youden index

First Author (Ref. #), Year

Parikh et al. (25), 2016

Ky et al. (37), 2011 Emdin et al. (40), 2018

The 35-ng/ml cut-off has been adopted by the U.S. Food and Drug Administration as a prognostic cut-off (21). AHF ¼ acute heart failure; CHF ¼ chronic heart failure; HF ¼ heart failure; sST2 ¼ soluble suppression of tumorigenesis-2.

hazard analysis (p < 0.001). In the presence of a low

increase of baseline log2-sST2: 1.30; 95% confidence

ST2 level, natriuretic peptides did not predict mor-

interval: 1.08 to 1.56; p ¼ 0.005). When repeated

tality (29). The risk threshold commonly used in

measurements were taken into account, the hazard

chronic HF (i.e., 35 ng/ml) has been evaluated in a

ratio per 1-SD increase of sST2 during follow-up was

recent study, displaying a modest prognostic perfor-

1.85 (95% confidence interval: 1.02 to 3.33; p ¼ 0.044),

mance (30); higher risk thresholds have been pro-

after adjustment for clinical factors and repeated

posed, most notably 49 (29) and 65 ng/ml (31,32)

measurements of NT-proBNP. Furthermore, ST2

(Table 2).

levels started to increase several weeks before the

Studies evaluating the prognostic significance of

occurrence of the primary endpoint (35) (Figure 1).

sST2 in acute HF have consistently reported that this

In a recent subanalysis of the PIONEER-HF (Com-

biomarker helps refine risk stratification, particularly

parison of Sacubitril/valsartan vs. Enalapril on Effect

for fatal endpoints. A meta-analysis by our group

on

showed that sST2 levels, measured both on admission

[NT-proBNP] in Patients Stabilized from an Acute HF

and at discharge, predict all-cause and cardiovascular

Episode) trial, baseline sST2 concentrations yielded

mortality. Moreover, unlike measurement at the time

prognostic significance for the composite outcome of

of admission, sST2 plasma concentration at discharge

cardiovascular

yielded prognostic significance for hospital read-

Notably, patients in the sacubitril/valsartan arm dis-

mission for HF (33).

played a greater reduction in circulating sST2 than

N-terminal

pro-brain

death

or

natriuretic

HF

peptide

rehospitalization.

The usefulness of serial sST2 measurement in

enalapril after as early as 1 week, as well as a better

acute HF has been specifically evaluated by several

outcome. sST2 levels decreased less than hs-TnT or

studies. In a population of 150 patients undergoing

NT-proBNP (6.4% vs. 15.8% and 29.4%, respectively),

daily blood sampling for sST2, percent change in ST2

possibly reflecting a lower impact of decongestion on

was strongly predictive of 90-day mortality: patients

sST2 levels than hs-TnT or NT-proBNP (36).

whose ST2 values decreased by 15.5% or more during the study period had a 7% chance of death, whereas

sST2 FOR RISK STRATIFICATION IN

patients whose ST2 levels failed to decrease by 15.5%

CHRONIC HF

in this time interval had a 33% chance of death. The prognostic value of sST2 changes was again inde-

In 2011, Ky et al. (37) performed a meta-analysis of

pendent from variations in NT-proBNP (34). In a

sST2 for prognosticating outcomes in chronic HF in 3

multicenter study from the Netherlands, 496 patients

cohorts. These results were further validated in the

with acute HF underwent repeated blood sampling

HF-ACTION (Heart Failure: A Controlled Trial Inves-

(on admission; at days 2 and 4; at discharge; after 2

tigating Outcomes of Exercise Training) cohort (38),

and 4 weeks; and at 3, 6, 9, and 12 months). After

leading to regulatory approval in the United States. In

adjustment for clinical factors and NT-proBNP, base-

2016, a much larger meta-analysis, considering

line sST2 was associated with an increased risk of the

pooled data from 7 studies and 6,372 patients, re-

primary endpoint (composite of all-cause mortality

ported that unadjusted sST2 values predict all-cause

and HF rehospitalization: hazard ratio per 1-SD

and cardiovascular mortality in chronic HF (39). The

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

F I G U R E 1 Trends of sST2 and Outcome After a Hospitalization for HF

100

80 sST2 (ng/mL)

2198

60

40

20

140

120

100

80 60 Time to Event (Days)

Patients with Event

40

20

0

Patients without Event

EVENT

Soluble suppression of tumorigenesis-2 (sST2) trends and individual sST2 values during follow-up after a heart failure (HF) hospitalization are reported. Patients experiencing the “event” (all-cause death or new HF hospitalization) (blue) and those who did not (red) are distinguished. Reproduced with permission from van Vark et al. (35).

sST2 >35-ng/ml cut-off was associated with a signifi-

While galectin-3 (another biomarker of inflam-

cantly higher risk of mortality at 1 year, also when

mation and fibrosis) has been reported to be more

adjusting for several covariates (39). An analysis of

predictive than sST2 in acute HF (43), several pa-

4,268 individual patient data from 11 cohorts found

pers suggest that sST2 holds superior prognostic

that the risk of all-cause death, cardiovascular death,

meaning than galectin-3 in chronic HF (44). sST2

and HF hospitalization increases exponentially with

was also equally predictive of outcome in either

sST2 levels, with 28 ng/ml identified as the best cut-

reduced or preserved ejection fraction (40,45), and

off at receiver-operating characteristic curves for the

independently predicted reverse remodeling (i.e.,

3 endpoints (40). When considering median values of

the improvement of LV geometry and function in

sST2, NT-proBNP, and hs-TnT (27 ng/ml, 1,360 ng/l,

response to guideline-recommended HF therapy),

and 18 ng/l, respectively), patients with only sST2 $

thus favorably affecting long-term outcome (46,47).

median had an increased risk of all-cause death, car-

Lupón et al. (46,47) have developed and validated

diovascular death, and HF hospitalization by 100%,

the ST2-R2 score, which includes sST2 <48 ng/ml

50%, and 10%, respectively, compared with those

(3 points), nonischemic etiology (5 points), absence

with no biomarker $ median; those with the 3

of left bundle branch block (4 points), history of

biomarkers $ median had a risk increased by 850%,

HF <1 year (2 points), LV ejection fraction <24%

640%, and 590%, respectively; and those with sST2

(1

and another biomarker $ median had an intermediate

(2 points). The likelihood of reverse remodeling

risk profile, with a greater risk of all-cause death

(assessed over 1 year) increased in parallel with the

associated with the combination of sST2 and hs-

score points, while the risk of mortality decreased

TnT $ median (Figure 2). The prognostic value of

(46,47).

point),

and

treatment

with

beta-blockers

sST2 was independent from NT-proBNP, hs-TnT, and

The Barcelona Bio-HF score includes sST2 and 10

other variables with prognostic significance (40). In

other variables, and predicts the risk of death at 1, 2,

the same study, age, sex, body mass index, and renal

and 3 years with good performance (Online Figure 1)

function did not correlate with sST2 concentrations

(48). This score, which provides a further demon-

(40), as confirmed in other studies (41,42).

stration of the strong prognostic value of sST2 in

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

F I G U R E 2 Additive Value of sST2 to NT-proBNP and hs-TnT for Outcome Prediction in Chronic HF

> sST2 597 241 163

964

392 423

264 > NT

> TnT

< NT, < sST2, < TnT 1,224 Whole Population 4,268 RR 10 9 8 7 6 5 4 3 2 1 0 CV Hospitalization < NT < TnT < sST2

> NT < TnT < sST2

< NT > TnT < sST2

< NT < TnT > sST2

CV Death > NT > TnT < sST2

All-Cause Death > NT < TnT > sST2

< NT > TnT > sST2

> NT > TnT > sST2

Relative risk (RR) of 3 endpoints (all-cause death, cardiovascular [CV] death, and HF hospitalization) were computed. Patients were classified according to median biomarker levels: N-terminal fragment of pro-brain natriuretic peptide (NT) 1,360 ng/l; high-sensitivity troponin T (TnT) 18 ng/l; sST2 27 ng/ml. Reproduced with permission from Emdin et al. (40). Abbreviations as in Figure 1.

chronic HF, particularly for the prediction of fatal

Elevated sST2 was shown to predict sudden cardiac

outcomes, could possibly be expanded by including

death in patients with chronic HF, with additive

the use of sacubitril valsartan and mineralocorticoid

prognostic significance to NT-proBNP (49). sST2 has

receptor antagonists.

also been proposed as a predictor of risk of death or

2199

2200

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

T A B L E 3 Guideline Recommendations on the Clinical Use of sST2 in Either CHF or AHF

Society

Year (Ref. #)

Setting

Recommendation

Class

LOE

2013 (57)

CHF

Measurement of other clinically available tests such as biomarkers of myocardial injury or fibrosis may be considered for additive risk stratification.

IIb

B

Measurement of biomarkers of myocardial injury or fibrosis may be considered for additive risk stratification.

IIb

Measurement of other clinically available tests such as biomarkers of myocardial injury or fibrosis may be considered for additive risk stratification.

IIb

No specific recommendation.



ACC/AHA

Gaggin et al. (56) reported that rising sST2 over 1 year’s time identified patients at higher risk for progressive LV dysfunction (odds ratio: 1.22; p ¼ 0.01). These results were recapitulated by Lupón et al.

2017 (58)

2013 (57)

CHF

AHF

(46,47)

with

the

ST2-R2

score

to

predict

LV

remodeling.

GUIDELINE RECOMMENDATIONS B-NR

The 2013 American College of Cardiology Foundation/ American Heart Association HF guidelines recomA

mended to assess biomarkers of inflammation and fibrosis to refine prognostic stratification, albeit with a low class of recommendation (IIb; Level of Evidence: B for chronic HF, Level of Evidence: A for

ESC 2016 (59)





acute HF) (57), and not citing any specific biomarker. The 2017 update of these guidelines includes a similar

ACC/AHA ¼ American College of Cardiology/American Heart Association; ESC ¼ European Society of Cardiology; LOE ¼ Level of Evidence; NR ¼ nonrandomized; other abbreviations as in Table 2.

recommendation for chronic HF, whereas acute HF is not mentioned (58). On the other hand, the 2016 European Society of Cardiology Guidelines state that the evidence of sST2 and other emerging biomarkers is

ventricular arrhythmias in patients eligible for car-

still not so compelling to recommend their use in

diac resynchronization therapy and defibrillation.

clinical practice (59) (Table 3).

Among 684 patients from the MADIT (Multicenter Automated Defibrillator Implantation Trial), elevated baseline sST2 was associated with an increased risk of death, death or HF, and death or ventricular arrhythmias even when adjusting for BNP. Patients presenting lower baseline sST2 (<35 ng/ml) had greater risk reduction with cardiac resynchronization therapy and defibrillation (p ¼ 0.006) (50). In patients with stage D HF, elevated concentrations of sST2 were associated with higher risk for failure of HF treatment and impending mortality/transplant/ventricular assist device placement (51). sST2 tends to decrease after LV assist device implantation (52), and that sST2 levels after heart transplantation are associated with a higher risk of cellular rejection and longterm risk of mortality (53).

sST2 IN HF: OPEN QUESTIONS AND FUTURE PERSPECTIVES Similar to natriuretic peptides (60) and troponins (61,62), and different from almost all other biomarkers listed by Dr. Braunwald back in 2008 (1), sST2 meets 2 fundamental criteria for clinically useful biomarkers: 1) accurate, repeated measurements are available at a reasonable cost, with assay automatization in the next future likely providing further impulse to its diffusion; and 2) it provides additive information to a careful clinical assessment, especially for risk stratification (Table 4). However, our knowledge of sST2 and of its assay methodology is far from being complete, starting from the mechanisms regulating sST2 production in healthy subjects and

sST2, CARDIAC STRUCTURE, AND FUNCTION

diseased patients, and the binding sites of sST2 assays. Even more important, at least from a clinical

Circulating levels of sST2 are not as closely associated

perspective, we are still striving to understand when

with parameters of cardiac structure and function, in

and how to measure sST2 levels, and how to translate

agreement with the notion of a predominant extrac-

this information into better patient care.

ardiac production of this biomarker. In chronic HF,

In acute HF, it seems reasonable to suggest that

higher sST2 has been associated with LV diastolic

sST2 be measured at least on admission and at the

dysfunction, increased

systolic

planned discharge. Patients whose sST2 levels do not

pressure, and hypokinesis (54). In arrhythmogenic

decrease might be considered to be at higher risk,

right ventricular cardiomyopathy, the function of

which may suggest a prolongation of the hospital

both ventricles decreased with increasing plasma

stay, a more rapid up-titration of HF drugs (after he-

sST2, suggesting more extensive fibrofatty replace-

modynamic stabilization), as well as more frequent

ment. Accordingly, sST2 was higher in patients with

visits after discharge or the use of monitoring systems

ventricular arrhythmias than in those without, even

to detect pulmonary congestion. In the chronic

after adjusting for right ventricular function (55).

setting, point sST2 values predict outcome and

right

ventricular

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

T A B L E 4 Clinical Value of NPs, Cardiac hs Troponins, and sST2 in HF

NPs hs-troponins sST2

Multimarker strategy (AHF: ST2 þ NPs) (CHF: sST2 þ NPs þ hs-troponins)

Diagnosis: HF

Risk Stratification: AHF

Risk Stratification: CHF

Guide to Treatment

þþþ (57–59)

þþþ (57–59)

þþþ (57–59)

þ/

n/a (key to ACS diagnosis)

þþ (62)

þþ (61)

n/a

 (24)

þþ þ  Admission and discharge sST2:  Strong, independent predictor univariate predictors of all-cause of outcome (all-cause death, CV and CV death after discharge (33). death, HF hospitalization) (40).  Predictor of reverse remodeling  Discharge sST2: univariate predictor (46,47). of HF hospitalization (33).  % change in ST2 during  Predictor of SCD (49) and benefit from CRT-D (50). hospitalization: predictor of 90-day mortality (34).  sST2 increase over time: predictive of 1-year all-cause death or HF hospitalization (35).

n/a

n/a

þþ þ  Prognostic value of admission  Additive prognostic value of sST2 independent from NT-proBNP sST2 to NPs and hs-troponins (24,29). (40,48).  Percent change in ST2 predictive of 90-day mortality regardless of variations in NT-proBNP (34).  Increase in sST2 over time of 1-year all-cause death or HF hospitalization independent from repeated NTproBNP measurements (35).

n/a

Numbers in parentheses are reference citations. þþþ ¼ strong evidence, guideline-recommended; þþ ¼ clear-cut evidence from individual patient data meta-analyses; þ ¼ evidence from multiple original studies and/or pooled data meta-analyses; þ/ ¼ controversial issue;  ¼ negative evidence from the literature; n/a: not available data; ACS ¼ acute coronary syndrome; CRT-D ¼ cardiac resynchronization therapy and defibrillation; hs ¼ high-sensitivity; NP ¼ natriuretic peptide; NT-proBNP ¼ N-terminal fragment of pro–B-type natriuretic peptide; SCD ¼ sudden cardiac death; other abbreviations as in Table 2.

reverse remodeling in patients with chronic HF on

research is to establish if a sST2-guided management

guideline-recommended treatment. sST2 measure-

of acute or chronic HF patients could have a positive

ment is then a valuable tool for risk stratification,

effect on patient symptoms and disease evolution.

either alone or together with natriuretic peptides and troponins, but the possibility to guide HF treatment

ADDRESS FOR CORRESPONDENCE:

on this basis (e.g., up-titrating or switching drugs, or

Emdin, Scuola Superiore Sant’Anna and Fonda-

deciding whether to refer a patient with nonischemic

zione Toscana Gabriele Monasterio, Via G. Moruzzi

dilated cardiomyopathy and mild systolic dysfunc-

1—56124 Pisa, Italy. E-mail: [email protected] OR

tion for defibrillator implantation) remains to be

[email protected]. Twitter: @MicheleEmdin,

investigated. Overall, the main challenge of future

@JJheart_doc.

Dr.

Michele

REFERENCES 1. Braunwald E. Biomarkers in heart failure. N Engl J Med 2008;358:2148–59. 2. Truong QA, Januzzi JL, Szymonifka J, et al. Coronary sinus biomarker sampling compared to peripheral venous blood for predicting outcomes in patients with severe heart failure undergoing cardiac resynchronization therapy: the BIOCRT study. Heart Rhythm 2014;11:2167–75. 3. AbouEzzeddine OF, McKie PM, Dunlay SM, et al. Suppression of tumorigenicity 2 in heart failure with preserved ejection fraction. J Am Heart Assoc 2017;6:e004382. 4. Pascual-Figal DA, Pérez-Martínez MT, AsensioLopez MC, et al. Pulmonary production of soluble ST2 in heart failure. Circ Heart Fail 2018;11: e005488.

5. Pascual-Figal DA, Januzzi JL. The biology of ST2: the International ST2 Consensus Panel. Am J Cardiol 2015;115:3B–7B. 6. Bayes-Genis A, Januzzi JL. The International ST2 Consensus Panel: introduction. Am J Cardiol 2015;115:1B–2B. 7. Tominaga S. A putative protein of a growth specific cDNA from BALB/c-3T3 cells is highly similar to the extracellular portion of mouse interleukin 1 receptor. FEBS Lett 1989;258:301–4. 8. Werenskiold AK, Hoffmann S, Klemenz R. Induction of a mitogen responsive gene after expression of the Ha-ras oncogene in NIH 3T3 fibroblasts. Mol Cell Biol 1989;9:5207–14. 9. Tominaga S. Murine mRNA for the beta-subunit of integrin is increased in BALB/c-3T3 cells

entering the G1 phase from the G0 state. FEBS Lett 1988;238:315–9. 10. Weinberg EO, Shimpo M, De Keulenaer GW, et al. Expression and regulation of ST2, an interleukin-1 receptor family member, in cardiomyocytes and myocardial infarction. Circulation 2002;106:2961–6. 11. Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005;23: 479–90. 12. Sanada S, Hakuno D, Higgins LJ, et al. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 2007;117:1538–49.

2201

2202

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

sST2 in Heart Failure

13. Seki K, Sanada S, Kudinova AY, et al. Interleukin-33 prevents apoptosis and improves survival after experimental myocardial infarction through ST2 signaling. Circ Heart Fail 2009;2: 684–91. 14. Bergers G, Reikerstorfer A, Braselmann S, Graninger P, Busslinger M. Alternative promoter usage of the Fos-responsive gene Fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J 1994;13:1176–88. 15. Ho JE, Chen WY, Chen MH, et al. Common genetic variation at the IL1RL1 locus regulates IL33/ST2 signaling. J Clin Invest 2013;123:4208–18.

28. Weinberg EO, Shimpo M, Hurwitz S, et al. Identification of serum soluble ST2 receptor as a novel heart failure biomarker. Circulation 2003; 107:721–6. 29. Rehman SU, Mueller T, Januzzi JL Jr. Characteristics of the novel interleukin family biomarker ST2 in patients with acute heart failure. J Am Coll Cardiol 2008;52:1458–65. 30. Pascual-Figal DA, Bayes-Genis A, AsensioLopez MC, et al. The interleukin-1 axis and risk of death in patients with acutely decompensated heart failure. J Am Coll Cardiol 2019;73:1016–25.

man hypertrophy and failure is related to diastolic load. J Am Coll Cardiol 2008;52:2166–217.

31. Pascual-Figal DA, Manzano-Fernández S, Boronat M, et al. Soluble ST2, high-sensitivity troponin T- and N-terminal pro-B-type natriuretic peptide: complementary role for risk stratification in acutely decompensated heart failure. Eur J Heart Fail 2011;13:718–25.

17. Pacho C, Domingo M, Núñez R, et al. Predictive biomarkers for death and rehospitalization in comorbid frail elderly heart failure patients. BMC Geriatr 2018;18:109.

32. Aimo A, Maisel AS, Castiglione V, et al. sST2 for outcome prediction in acute heart failure: which is the best cut-off? J Am Coll Cardiol 2019; 74:478–9.

18. Zhao J, Wei J, Mialki RK, et al. F-box protein FBXL19-mediated ubiquitination and degradation of the receptor for IL-33 limits pulmonary inflammation. Nat Immunol 2012;13:651–8.

33. Aimo A, Vergaro G, Ripoli A, et al. Meta-analysis of soluble suppression of tumorigenicity-2 and prognosis in acute heart failure. J Am Coll Cardiol HF 2017;5:287–96.

19. Mueller T, Jaffe AS. Soluble ST2-analytical considerations. Am J Cardiol 2015;115:8B–21B.

34. Boisot S, Beede J, Isakson S, et al. Serial sampling of ST2 predicts 90-day mortality following destabilized heart failure. J Card Fail 2008;14:732–8.

16. Bartunek J, Delrue L, Van Durme F, et al. Nonmyocardial production of ST2 protein in hu-

20. Mueller T, Dieplinger B. The Presage() ST2 Assay: analytical considerations and clinical applications for a high-sensitivity assay for measurement of soluble ST2. Expert Rev Mol Diagn 2013;13:13–30.

35. van Vark LC, Lesman-Leegte I, Baart SJ, et al. Prognostic value of serial ST2 measurements in patients with acute heart failure. J Am Coll Cardiol 2017;70:2378–88.

21. Wu AH, Wians F, Jaffe A. Biological variation of galectin-3 and soluble ST2 for chronic heart failure: implication on interpretation of test results. Am Heart J 2013;165:995–9. 22. Piper S, deCourcey J, Sherwood R, et al. Biologic variability of soluble ST2 in patients with stable chronic heart failure and implications for monitoring. Am J Cardiol 2016;118:95–8. 23. Coglianese EE, Larson MG, Vasan RS, et al. Distribution and clinical correlates of the interleukin receptor family member soluble ST2 in the Framingham Heart Study. Clin Chem 2012;58: 1673–81. 24. Januzzi JL Jr., Peacock WF, Maisel AS, et al. Measurement of the interleukin family member ST2 in patients with acute dyspnea: results from the PRIDE (Pro-Brain Natriuretic Peptide Investigation of Dyspnea in the Emergency Department) study. J Am Coll Cardiol 2007;50:607–13. 25. Parikh RH, Seliger SL, Christenson R, et al. Soluble ST2 for prediction of heart failure and cardiovascular death in an elderly, communitydwelling population. J Am Heart Assoc 2016;5: e003188. 26. Wang TJ, Wollert KC, Larson MG, et al. Prognostic utility of novel biomarkers of cardiovascular stress: the Framingham Heart Study. Circulation 2012;126:1596–604. 27. Chen LQ, de Lemos JA, Das SR, et al. Soluble ST2 is associated with all-cause and cardiovascular mortality in a population-based cohort: the Dallas Heart Study. Clin Chem 2013;59:536–46.

36. Morrow DA, Velazquez EJ, DeVore AD, et al. Cardiovascular biomarkers in patients with acute decompensated heart failure randomized to sacubitril-valsartan or enalapril in the PIONEERHF trial. Eur Heart J 2019 May 15 [E-pub ahead of print]. 37. Ky B, French B, McCloskey K, et al. Highsensitivity ST2 for prediction of adverse outcomes in chronic heart failure. Circ Heart Fail 2011;4: 180–7. 38. Felker GM, Fiuzat M, Thompson V, et al. Soluble ST2 in ambulatory patients with heart failure: Association with functional capacity and longterm outcomes. Circ Heart Fail 2013;6:1172–9. 39. Aimo A, Vergaro G, Passino C, et al. Prognostic value of soluble suppression of tumorigenicity-2 in chronic heart failure: a meta-analysis. J Am Coll Cardiol HF 2017;5:280–6. 40. Emdin M, Aimo A, Vergaro G, et al. sST2 predicts outcome in chronic heart failure beyond NT-proBNP and high-sensitivity troponin T. J Am Coll Cardiol 2018;72:2309–20. 41. Wojtczak-Soska K, Pietrucha T, Sakowicz A, et al. Soluble ST2 protein in chronic heart failure is independent of traditional factors. Arch Med Sci 2013;9:21–6. 42. Bayes-Genis A, Zamora E, de Antonio M, et al. Soluble ST2 serum concentration and renal function in heart failure. J Card Fail 2013;19:768–75. 43. Mueller T, Gegenhuber A, Leitner I, et al. Diagnostic and prognostic accuracy of galectin-3

and soluble ST2 for acute heart failure. Clin Chim Acta 2016;463:158–64. 44. Bayes-Genis A, de Antonio M, Vila J, et al. Head-to-head comparison of 2 myocardial fibrosis biomarkers for long-term heart failure risk stratification: ST2 versus galectin-3. J Am Coll Cardiol 2014;63:158–66. 45. Sanders-van Wijk S, van Empel V, Davarzani N, et al. Circulating biomarkers of distinct pathophysiological pathways in heart failure with preserved vs. reduced left ventricular ejection fraction. Eur J Heart Fail 2015;17:1006–14. 46. Lupón J, Gaggin HK, de Antonio M, et al. Biomarker-assist score for reverse remodeling prediction in heart failure: The ST2-R2 score. Int J Cardiol 2015;184:337–43. 47. Lupón J, Sanders-van Wijk S, Januzzi JL, et al. Prediction of survival and magnitude of reverse remodeling using the ST2-R2 score in heart failure: a multicenter study. Int J Cardiol 2016;204:242–7. 48. Lupón J, de Antonio M, Vila J, Peñafiel J, et al. Development of a novel heart failure risk tool: the Barcelona bio-heart failure risk calculator (BCN bio-HF calculator). PLoS One 2014;9:e85466. 49. Pascual-Figal DA, Ordonez-Llanos J, Tornel PL, et al. Soluble ST2 for predicting sudden cardiac death in patients with chronic heart failure and left ventricular systolic dysfunction. J Am Coll Cardiol 2009;54:2174–9. 50. Skali H, Gerwien R, Meyer TE, et al. Soluble ST2 and risk of arrhythmias, heart failure, or death in patients with mildly symptomatic heart failure: results from MADIT-CRT. J Cardiovasc Transl Res 2016;9:421–8. 51. Zilinski JL, Shah RV, Gaggin HK, et al. Measurement of multiple biomarkers in advanced stage heart failure patients treated with pulmonary artery catheter guided therapy. Crit Care 2012;16:R135. 52. Ahmad T, Wang T, O’Brien EC, et al. Effects of left ventricular assist device support on biomarkers of cardiovascular stress, fibrosis, fluid homeostasis, inflammation, and renal injury. J Am Coll Cardiol HF 2015;3:30–9. 53. Januzzi JL, Horne BD, Moore SA, et al. Interleukin receptor family member ST2 concentrations in patients following heart transplantation. Biomarkers 2013;18:250–6. 54. deFilippi C, Daniels LB, Bayes-Genis A. Structural heart disease and ST2: cross-sectional and longitudinal associations with echocardiography. Am J Cardiol 2015;115:59B–63B. 55. Broch K, Leren IS, Saberniak J, et al. Soluble ST2 is associated with disease severity in arrhythmogenic right ventricular cardiomyopathy. Biomarkers 2017;22:367–71. 56. Gaggin HK, Szymonifka J, Bhardwaj A, et al. Head-to-head comparison of serial soluble ST2, growth differentiation factor-15, and highlysensitive troponin T measurements in patients with chronic heart failure. J Am Coll Cardiol HF 2014;2:65–72. 57. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: executive summary: a report of the American College of Cardiology Foundation/

Aimo et al.

JACC VOL. 74, NO. 17, 2019 OCTOBER 29, 2019:2193–203

American Heart Association Task Force on practice guidelines. J Am Coll Cardiol 2013;62:e147–239. 58. Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/AHA/HFSA focused update of the 2013 ACCF/ AHA Guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. J Am Coll Cardiol 2017;70: 776–803. 59. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and

sST2 in Heart Failure

chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail 2016;18: 891–975. 60. Ibrahim NE, Januzzi JL Jr. The future of biomarker-guided therapy for heart failure after the Guiding Evidence-Based Therapy Using Biomarker Intensified Treatment in Heart Failure (GUIDE-IT) Study. Curr Heart Fail Rep 2018;15: 37–43. 61. Aimo A, Januzzi JL Jr., Vergaro G, et al. Prognostic value of high-sensitivity troponin T in chronic heart failure: an individual patient

data meta-analysis. 286–97.

Circulation

2018;137:

62. Felker GM, Mentz RJ, Teerlink JR, et al. Serial high sensitivity cardiac troponin T measurement in acute heart failure: insights from the RELAX-AHF study. Eur J Heart Fail 2015;17:1262–70.

KEY WORDS biomarker, heart failure, outcome, sST2

A PP END IX For a supplemental figure, please see the online version of this paper.

2203