Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 9.
Published in final edited form as: Am J Cardiol. 2013 Oct 4;113(2):321–327. doi: 10.1016/j.amjcard.2013.08.047

Effects of Interleukin-1 Blockade with Anakinra on Aerobic Exercise Capacity in Patients with Heart Failure and Preserved Ejection Fraction (from the D-HART Pilot Study)

Benjamin Wallace Van Tassell *,#, Ross Arena *,#, Giuseppe Biondi-Zoccai **, Justin McNair Canada *, Claudia Oddi *, Nayef Antar Abouzaki *, Arehzo Jahangiri *, Raquel Appa Falcao *, Michael Christopher Kontos *, Keyur Bharat Shah *, Norbert Felix Voelkel *, Charles Anthony Dinarello ***, Antonio Abbate *
PMCID: PMC4899612  NIHMSID: NIHMS530697  PMID: 24262762

Abstract

Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome of exercise intolerance due to impaired myocardial relaxation and/or increased stiffness. Patient with HFpEF often show signs of chronic systemic inflammation and experimental studies show that interleukin-1 (IL-1), a key pro-inflammatory cytokine, impairs myocardial relaxation. The aim of the current study was to determine the effects of IL-1 blockade with anakinra on aerobic exercise capacity in patients with HFpEF and plasma C-reactive protein (CRP) >2 mg/l (reflecting increased IL-1 activity). Twelve patients were enrolled in a double-blind, randomized, placebo-controlled, cross-over trial and assigned 1:1 to receive one of the 2 treatments (anakinra 100 mg or placebo) for 14 days and then an additional 14 days of the alternate treatment (placebo or anakinra). Cardiopulmonary exercise testing (CPX) was performed at baseline, after the first 14 days and after the second 14 days. Placebo-corrected interval change in peak oxygen consumption (VO2) was chosen as primary endpoint. All 12 patients enrolled in the study and receiving treatment completed both phases, and experienced no major adverse events. Anakinra led to a statistically significant improvement in peak VO2 (+1.2 ml•kg−1•min−1, P=0.009), and a significant reduction in plasma CRP levels (−74%, P=0.006). The reduction in CRP levels was correlated with the improvement in peak VO2 (R=-0.60, P=0.002). Three patients (25%) had mild and self-limiting injection site reactions. In conclusion, IL-1 blockade with anakinra for 14 days significantly quenches the systemic inflammatory response and improves aerobic exercise capacity in patients with HFpEF and elevated plasma CRP levels.

Keywords: heart failure, inflammation, diastole, obesity

INTRODUCTION

Heart failure (HF) is a clinical syndrome of exercise intolerance secondary to impaired cardiac function. Approximately 50% of patients with HF have preserved left ventricular (LV) systolic function (HF with preserved ejection fraction – HFpEF) characterized by impaired LV diastolic filling due to incompletely characterized mechanisms.1-3 Observational studies have linked markers of systemic inflammation with impaired cardiac function and poor prognosis in patients with HFpEF4-5 and in patients with chronic inflammatory diseases.6-8 Interleukin-1 (IL-1) is an apical cytokine involved in local and systemic inflammatory processes.9 IL-1 induces changes in systolic and diastolic function in experimental animal studies.10 Patients with rheumatoid arthritis (an IL-1 related disease) show signs of impaired LV diastolic function and treatment with anakinra—an IL-1 blocker—restored normal LV diastolic function within hours of treatment.7-8 Anakinra is approved for the treatment of chronic systemic inflammatory diseases, and has recently been shown to reduce the incidence of HF following ST-segment elevation AMI and to improve aerobic exercise capacity in patients with HF and reduced ejection fraction (HFrEF).11-12 We now present the effects of anakinra on aerobic exercise capacity and ventilatory efficiency in a randomized, double-blind, placebo-controlled cross-over pilot trial in patients with HFpEF and systemic inflammation (plasma CRP levels >2 mg/l).

METHODS

The study design was registered in www.clinicaltrials.gov (NCT01542502). An exemption for an investigational new drug use was granted by the Food and Drug Administration. The study was approved by the local Institutional Review Board and all patients provided written consent. Inclusion criteria were age >18 years, New York Heart Association (NYHA) class II-III HF symptoms (without changes in class or treatment in the past 3 months), preserved LV ejection fraction (>50%) with LV end-diastolic volume index (<97 ml/m2), and evidence of abnormalities in LV relaxation, filling, distensibility or stiffness as defined by the European Heart Society consensus document.3 Patients were excluded for any of the following conditions: HF hospitalization within the prior 12 months; recent (<3 months) changes in medical therapy for HF; concomitant conditions or treatments affecting completion or interpretation of the cardiopulmonary exercise test (CPX)(i.e. physical inability to walk on a treadmill, significant myocardial ischemia, angina, uncontrolled arterial hypertension [at rest or during the baseline exercise test], atrial fibrillation, moderate to severe aortic or mitral valve disease, chronic pulmonary disease limiting exertion, or anemia [defined as hemoglobin <10 g/dl]; recent use of systemic immunosuppressive or anti-inflammatory drugs (not including non-steroidal anti-inflammatory drugs); chronic inflammatory or infectious diseases; stage IV-V kidney disease; neutropenia (<2,000/mm3); pregnancy; any malignancy or any condition limiting survival or ability to complete the study. After initial screening, patients were assessed for systemic inflammation, defined as plasma high-sensitivity C reactive protein (CRP) levels >2 mg/l, using an automated high-sensitivity latex-enhanced assay.13

All patients underwent CPX at baseline and upon completion of 14 days and 28 days treatment. The CPX was administered using a metabolic cart that is interfaced with a treadmill (Vmax Encore, Viasys, Yorba Linda, CA). A conservative ramping treadmill protocol was used as described previously.12 Expired gases were sampled using a mouthpiece-mounted sensor, and analyzed to continuously measure oxygen consumption (VO2), carbon dioxide production (VCO2) and minute ventilation (VE). Peak VO2, the VE/VCO2 slope, and the oxygen uptake efficiency slope (OUES) were determined as described, with the first measuring aerobic exercise capacity and the other ventilatory efficiency, the ability to expel CO2 and consume VO2 at an appropriately low VE.12,14 All patients also completed a HF symptom questionnaire (Duke Activity Status Index [DASI])12. The investigational pharmacist performed randomization using a dedicated randomization algorithm prepared on randomization.com by one of the investigators not involved in the conduct of the study (seed #26404, created on 11/29/2011). For each patient, the pharmacist prepared two indistinguishable sets of 14 syringes containing 100 mg of anakinra (Kineret™, Swedish Orphan Biovitrum, Stockholm, Sweden) in 0.67 mL and matching NaCl 0.9% placebo. Treatment consisted of two courses of 14 daily subcutaneous injections without any washout period. Each patient was randomized to receive either active treatment first followed by placebo, or vice versa. All CPXs were performed by a single operator. Data were electronically transferred and all calculations related to the CPX were performed in Dr. Arena's core laboratory. The primary endpoint was the placebo-corrected difference in the interval change in peak VO2 from baseline to the post-treatment follow-up. Secondary endpoints included placebo-corrected differences in interval changes in CRP plasma levels and ventilatory efficiency (VE/VCO2 slope and OUES). Sample size calculations relied upon a pilot study of anakinra in patients with HFrEF, in which an identical 14-day treatment with anakinra produced a median improvement in peak VO2 of +2.8 ml•kg−1•min−1, with a standard deviation of 1.2 ml•kg−1•min−1.12 We therefore calculated that a cross-over trial would require a sample size of 14 patients to provide a power of >80% (alpha 0.05) to detect a difference of 1.4 ml•kg−1•min−1 with a standard deviation of 1.2 ml•kg−1•min−1, and <20% loss to follow up. The values are reported as median and interquartile range to account for potential deviation from the Gaussian distribution. The differences in interval changes between groups are analyzed using a random-effect general linear model for repeated measures analyzing the effects of time and group allocation, comparing on-treatment time 1 and on- treatment time 2, and using baseline time 0 as a covariate,15 and are presented as changes in the median values for all variables. The Spearman correlation test was used to evaluate correlations between variables. Statistical significance was set at the 0.05 level and unadjusted p values are reported. Computations were performed with SPSS 21 (IBM, NY, USA).

RESULTS

From January 2012 to May 2013, we screened 23 patients with stable HFpEF (diagnosed according to the ESC criteria)3 and enrolled 14 with CRP>2.0 mg/l, while excluding the remaining 9 patients with CRP≤2 mg/l (Figure 1): one patient signed informed consent but did not complete any of the study tests nor received treatment, one patient completed the baseline CPX but experienced angina, and therefore was excluded from the study prior to administration of any study treatment. Twelve patients received study medication and were included in the final analysis. Of these, 11 patients (92%) were women, 4 (33%) were self-defined Caucasians and 8 (67%) African-Americans. Median age was 62 years. All 12 patients had systemic arterial hypertension and 3 patients [25%] had echocardiographic evidence of left ventricular hypertrophy. All patients were obese (BMI≥30, median BMI 39 kg/m2 [range 30-50], with 4 [33%] having BMI>40). Median LVEF was 58% and median E/E’ was 11. Six patients (50%) has NYHA class II symptoms and the remaining 6 patients (50%) class III symptoms, with a median duration of symptoms of 20 months (range 6-36). None of the patients had moderate or severe impairment in the glomerular filtration rate (<60 ml/kg/1.73 m2). All patients were treated with furosemide, 10 patients (83%) were treated with either an angiotensin-converting enzyme inhibitor or an angiotensin receptor blocker, and 10 (83%) were treated with beta-adrenergic blockers, with all 12 patients (100%) being on either class of drugs and 8 (67%) on both. Two patients (17%) were receiving spironolactone and 11 patients (92%) were treated with a statin plus aspirin. Clinical characteristics of the 12 patients are reported in Table 1.

FIGURE 1. Study design.

FIGURE 1

Schematic diagram of the study design and the enrollment process.

Abbreviations: CPX= cardiopulmonary exercise testing; CRP= C reactive protein; HFpEF= Heart failure with preserved ejection fraction.

Table 1.

Baseline clinical characteristics and placebo-corrected changes in peak VO2

Age (yrs) Gender Ethnicity Hypertension Diabetes NYHA Class BNP hs-CRP LVEF LVEDVi LVMi E/E′ LVEDP Peak VO2 VE/VCO2 OUES Change in peak VO2
#1 40 F C Y N 2 43 4.7 63 33 72 24 n/a 16.9 24.6 2.08 +1.9
#2 52 F AA Y N 2 8 9.9 63 43 131 8 n/a 20.5 23.2 2.21 −1.1
#3 55 F AA Y Y 3 39 3.5 57 59 123 11 18 16.2 30.6 1.79 +1.7
#4 55 F AA Y N 2 19 6.6 58 33 91 11 n/a 18.7 22.3 2.85 −1.0
#5 57 F AA Y Y 2 2 17.0 57 41 79 11 17 14.1 25.2 2.12 +3.0
#6 60 F AA Y Y 3 25 11.5 60 62 125 15 n/a 13.8 24.9 1.92 −0.7
#7 62 F C Y Y 3 39 5.0 64 37 68 11 34 16.1 29.6 2.54 +2.1
#8 63 F AA Y N 2 11 15.2 74 34 89 21 20 14.4 22.2 1.8 +0.4
#9 64 F C Y Y 3 112 2.6 53 40 78 10 n/a 13.1 33 1.34 +1.7
#10 64 F AA Y Y 3 131 15.5 67 41 90 20 n/a 11.4 21.1 2.02 +3.5
#11 65 M C Y Y 2 23 3.1 59 60 68 11 n/a 17.3 31.1 1.73 +3.0
#12 70 F AA Y N 3 414 18.2 51 78 100 19 15 9.4 45.4 1.22 +1.8

Abbreviations: AA=African-American; BNP=Brain natriuretic peptide (pg/ml) ; C=Caucasian; E/E′= ratio of transmitral Doppler early filling velocity to tissue Doppler early diastolic mitral annular velocity; F=female; hs-CRP=high sensitivity C-reactive protein (mg/l); LVEDVi=left ventricular end-diastolic volumeindex (ml/m2); LVEF=left ventricular ejection fraction (%); LV Mass index=left ventricular mass index (g/m2); M=male; N=no; n/a=not available; NYHA Class=New York Heart Association functional classification system for heart failure; OUES= Oxygen-uptake efficiency slope; Peak VO2=maximal oxygen consumption during cardiopulmonary exercise testing (ml•kg−1•min−1); VE/VCO2=ventilatory equivalent ratio for carbon dioxide; Y=yes.

Six patients received anakinra first and 6 placebo, with the first dose given immediately after baseline CPX. The patients were then given a supply of 13 additional syringes to self-administer at home daily. All 12 patients completed the second CPX 14 days later, received the second batch of (cross-over) syringes, and completed the third CPX an additional 14 days later (see Figure 1). With the exception of the patient who experienced angina during baseline CPX (prior to any treatment), none of the treated patients experienced any serious adverse events or requested discontinuation of treatment. Three patients (25%) experienced mild injection site reactions with anakinra, whereas no injection site reactions were reported with placebo.

On-treatment peak VO2 was 16.3 ml•kg−1•min−1 [13.8-17.5] with anakinra vs. 15.1 ml•kg−1•min−1 [11.3-17.3] with placebo, leading to a placebo-corrected difference in median peak VO2 (primary endpoint) of 1.2 ml•kg−1•min−1 (+8%, P=0.009)(Figure 2). Placebo-corrected changes in peak VO2 for each patient are reported in Table 1. Anakinra treatment also led to a significant reduction in plasma CRP levels (placebo-corrected difference of −6.1 mg/l [-74%], P=0.006, Figure 2). No significant effect was seen on VE/VCO2 slope, a measure of ventilatory efficiency (Figure 2), however a sensitivity analysis showed a significant improvement in the VE/VCO2 slope in the subgroup of patients who were above median VE/VCO2 slope values (>25, N=5) at baseline (placebo-corrected difference 3.8 [12%] favoring anakinra, P=0.047). Treatment with anakinra improved the OUES, another measure of ventilatory efficiency (placebo-corrected difference 0.14 [7%], P=0.006, Figure 2). The interval changes in CRP levels were inversely correlated with the changes in peak VO2 (R=-0.60, P=0.002), with greater reduction in CRP being associated with greater increase in peak VO2 (Figure 3). Similarly, leukocyte count in the peripheral blood was significantly reduced with anakinra (placebo corrected difference −1,300/mm3, P=0.005, with a 23% reduction in neutrophil count [P=0.007] and a 20% reduction in monocyte count [P=0.039]). Despite small changes in the leukocyte count, it significantly correlated with changes in peak VO2, with greater reduction (with anakinra) predicting greater increases in peak VO2 (Figure 4). Conversely, no differences in brain-type natriuretic peptide (BNP) levels changes were observed, nor were the changes correlated with changes in peak VO2 (Supplemental Figure 1). There was a trend toward greater DASI scores (reflecting improved perceived functional capacity) favoring anakinra (median +5.4 [0/+12.6], P=0.068). Anakinra had no detectable effects on body weight, resting or maximal heart rate, resting or maximal systolic or diastolic blood pressure (data not shown). Although limited by the small numbers, none of the baseline clinical characteristics (such as gender, age, ethnicity, NYHA class, BNP levels, LVEF, E/E’ ratio, LVM or left atrial volume, diabetes, medications) explored at sensitivity analyses (using the variable as covariate) influenced the effects of treatment on peak VO2 (all P values >0.10 for interaction).

FIGURE 2. Effects of IL-1 blockade with anakinra on exercise parameters and C-reactive protein plasma levels.

FIGURE 2

The figure shows interval changes in the peak oxygen consumption (VO2 – top left panel), C reactive protein plasma levels (CRP – bottom left panel), ventilatory efficiency (VE/VCO2 – top right panel), and oxygen utilization efficiency slope (OUES – bottom right panel) after placebo and anakinra treatment phases.

FIGURE 3. C reactive protein levels.

FIGURE 3

Correlation between the changes in C-reactive protein (CRP) plasma levels with treatment and the effects on aerobic exercise capacity (peak oxygen consumption [VO2]).

FIGURE 4. Leukocyte count.

FIGURE 4

Changes in blood leukocyte count and changes in peak oxygen consumption (peak VO2) after treatment.

DISCUSSION

The pathophysiologic mechanisms leading to exercise intolerance in HFpEF are incompletely understood.1-3 The results of this pilot study, showing an improvement in aerobic exercise capacity and ventilatory efficiency in patients with HFpEF treated with an IL-1 blocker, suggest that enhanced IL-1 activity may represent an additional mechanism contributing to the pathophysiology of HFpEF. Increased myocardial stiffness and impaired relaxation lead to increased filling pressure and symptoms of pulmonary and systemic congestions in patients with HFpEF. Myocardial stiffness is related to structural changes such as cardiomyocyte hypertrophy and interstitial fibrosis, as well as to molecular changes in the isoform and phosphorylation state of titin, the key protein anchoring the sarcomere to the cytoskeleton providing increased stiffness at the cardiomyocyte level.2-3 Cardiomyocyte relaxation is, on the other hand, an active process that is modulated by myocardial and systemic factors.2-3,10,16 None of these factors alone explain the symptoms or hemodynamic changes seen in patients with HFpEF.2-3 Symptoms, functional limitations, and prognosis in patients with HFpEF are less likely to improve with standard HF treatments which have been shown to be effective in HFrEF, suggesting that the current paradigm of neuro-adrenergic and renin-angiotensin-aldosterone blockade fails to address one ore more key mechanisms involved in HFpEF. Experimental studies in cardiomyocyte cultures and animals show that IL-1 modulates myocardial contraction and relaxation.10,16 Ikonimidis and coll.7 described that patients with rheumatoid arthritis, without known cardiovascular disease, had evidence of abnormal myocardial relaxation which improved significantly within 3 hours of a single dose of anakinra, showing that IL-1 activity acutely modulates myocardial relaxation. More recently, the same group confirmed a significant improvement in diastolic function, myocardial strain, and vascular compliance following anakinra treatment also in patients with rheumatoid arthritis and heart disease.8

The current D-HART pilot study shows for the first time that patients with stable HFpEF and evidence of systemic inflammation (CRP>2 mg/l) have markedly reduced aerobic exercise capacity, and that treatment with anakinra to block IL-1 activity for 14 days leads to a statistically significant reduction in systemic inflammation and a significant improvement in exercise capacity. The reduction in CRP levels, a surrogate marker for IL-1 activity, correlated with the improvement in peak VO2. These results confirm the role of IL-1 in regulating cardiovascular function and provide the first evidence that an IL-1-targeted therapeutic strategy may be valuable in HFpEF.

Peak VO2 was chosen as the primary endpoint for the current study given its long established history as a robust prognostic marker in HF, and is considered the gold standard for quantifying functional capacity.17-18 The improvement in peak VO2 seen with anakinra in the D-HART study (placebo corrected difference in median peak VO2 +1.2 ml•kg−1•min−1) appears to be smaller than what has been observed in patients with HF and reduced EF,12 however there are differences in baseline impairment between HFpEF and HFrEF. Although smaller than expected, the improvement in peak VO2 is very likely to be clinically relevant, as even smaller improvements were associated with improved outcomes in the HF-ACTION study.19 The recent RELAX trial had set the very same target on the assumption it would be clinically relevant.20 Anakinra also improved the OUES and the VE/VCO2 slope (in those patients with above median values at baseline). Both an elevated VE/VCO2 slope and diminished OUES reflect ventilatory inefficiency, indicative of increased ventilation-perfusion mismatch, exaggerated mechano- and ergoreceptors, and impaired oxidative capacity of skeletal muscle, and portend a poor prognosis in HF.21-22

The choice of 14 days of treatment with anakinra may be viewed as arbitrary. The same regimen has been used in two pilot studies in patients with ST-segment elevation myocardial infarction in which a protective effect against new onset HF was seen,11,23 and in one pilot study in patients with chronic HFrEF (<40%) in which a significant improvement in peak VO2 was seen.12 It is unknown whether changes in peak VO2 would have occurred even with a single dose or a shorter duration of treatment, or whether anakinra would have provided greater benefit if given for a longer period. The favorable changes seen within a short time frame suggest that the benefits of anakinra are not due to changes in myocardial structure but rather due to changes in the function/compliance of the heart, vasculature and/or the skeletal muscles.7-8 IL-1 inhibits contractility, lusitropy, and beta-adrenergic responsiveness within few hours of administration.10,16 We cannot exclude, however, that a longer treatment with anakinra may provide more favorable changes due to a combined effect on function and structure (i.e hypertrophy or fibrosis). Moreover, inflammation appears also to regulate oxygen transport and utilization in skeletal muscle.24

There are several limitations to the D-HART pilot study, primarily the small number of patients and short duration of treatment and observation. All but one patient in this pilot study were women, the majority of patients were self-defined African-Americans, all were obese and all were affected by systemic arterial hypertension. While these characteristics are common to many HFpEF study populations, the generalizability of the results to other patients may be limited. The lack of BNP levels elevation in many subjects despite impaired diastolic function and moderate-to-severe limitations at CPX fits well with the paradigm of a lower BNP levels in HFpEF, a relative deficit in BNP elevation especially in obese patients with HFpEF, and a lack of correlation of changes in pro-BNP and outcomes in HFpEF. 25-27 The choice to use CRP as a screening tool and exclude patients with CRP≤2 mg/l may also appear to be arbitrary, and we cannot exclude that such patients would have equally benefited from anakinra treatment. Nevertheless, for this pilot study, we chose CRP>2mg/l as it appears to be a validated cut-off to identify a subgroup of patients at greater risk and a target for treatment.28-29 Moreover, it is impossible to know whether a longer duration of treatment would have provided more favorable effects or unforeseen intolerance to the treatment. Future studies are needed in this area.

Supplementary Material

01

Acknowledgments

Funding: This study was funded by an American Heart Association Scientist Development Grant (10SDG 3030051) Dr. Abbate, and by the internal funds of the VCU Pauley Heart Center and Victoria Johnson Research Laboratories. Dr. Van Tassell was supported by an Institutional National Institute of Health K12 award (KL2RR031989). Dr. Dinarello was supported by a National Institute of Health grant (AI-15614).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of interest: Dr. Abbate has received research funds or served on advisory boards for Swedish Orphan Biovitrum, Gilead, Janssen, Novartis and XOMA. Dr. Van Tassell has received research funds or served on advisory boards for Gilead and Novartis. Dr. Biondi-Zoccai has consulted or lectured for AstraZeneca, Bristol Myers Squibb, Eli Lilly, and Sanofi Aventis. Dr. Kontos has consulted for Astellas, GE, Sanofi-Aventis, and Well Point. Dr. Voelkel has received research funds from Actelion. Dr. Dinarello has received consulting fees from Swedish Orphan Biovitrum.

References

  • 1.Udelson JE. Heart failure with preserved ejection fraction. Circulation. 2011;124:e540–543. doi: 10.1161/CIRCULATIONAHA.111.071696. [DOI] [PubMed] [Google Scholar]
  • 2.Borlaug BA, Paulus WJ. Heart failure with preserved ejection fraction: pathophysiology, diagnosis, and treatment. Eur Heart J. 2011;32:670–679. doi: 10.1093/eurheartj/ehq426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Paulus WJ, Tschope C, Sanderson JE, Rusconi C, Flachskampf FA, Rademakers FE, Marino P, Smiseth OA, De Keulenaer G, Leite-Moreira AF, Borbely A, Edes I, Handoko ML, Heymans S, Pezzali N, Pieske B, Dickstein K, Fraser AG, Brutsaert DL. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology. Eur Heart J. 2007;28:2539–2550. doi: 10.1093/eurheartj/ehm037. [DOI] [PubMed] [Google Scholar]
  • 4.Michowitz Y, Arbel Y, Wexler D, Sheps D, Rogowski O, Shapira I, Berliner S, Keren G, George J, Roth A. Predictive value of high sensitivity CRP in patients with diastolic heart failure. Int J Cardiol. 2008;125:347–351. doi: 10.1016/j.ijcard.2007.02.037. [DOI] [PubMed] [Google Scholar]
  • 5.Shah SJ, Marcus GM, Gerber IL, McKeown BH, Vessey JC, Jordan MV, Huddleston M, Foster E, Chatterjee K, Michaels AD. High-sensitivity C-reactive protein and parameters of left ventricular dysfunction. J Card Fail. 2006;12:61–65. doi: 10.1016/j.cardfail.2005.08.003. [DOI] [PubMed] [Google Scholar]
  • 6.Liang KP, Myasoedova E, Crowson CS, Davis JM, Roger VL, Karon BL, Borgeson DD, Therneau TM, Rodeheffer RJ, Gabriel SE. Increased prevalence of diastolic dysfunction in rheumatoid arthritis. Ann Rheum Dis. 2010;69:1665–1670. doi: 10.1136/ard.2009.124362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ikonomidis I, Lekakis JP, Nikolaou M, Paraskevaidis I, Andreadou I, Kaplanoglou T, Katsimbri P, Skarantavos G, Soucacos PN, Kremastinos DT. Inhibition of interleukin-1 by anakinra improves vascular and left ventricular function in patients with rheumatoid arthritis. Circulation. 2008;117:2662–2669. doi: 10.1161/CIRCULATIONAHA.107.731877. [DOI] [PubMed] [Google Scholar]
  • 8.Ikonomidis I, Txortzis S, Andreadou I, Dasou P, Katseli C, Kastimbru P, Paraskevaidis I, Parissis J, Lekakis J, Anastasious-Nana M. Inhibition of interleukin-1 activity by anakinra improves left ventricular myocardial deformation and torsion in patients with CAD and coexstistent rheumatoid artrhitis: a randomized trial. Eur Heart J. 2012;33(Suppl 1):19/338. [Google Scholar]
  • 9.Dinarello CA. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117:3720–3732. doi: 10.1182/blood-2010-07-273417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Van Tassell BW, Toldo S, Mezzaroma E, Abbate A. Targeting Interleukin-1 in heart disease. Circulation. 2013 doi: 10.1161/CIRCULATIONAHA.113.003199. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Abbate A, Van Tassell BW, Biondi-Zoccai G, Kontos MC, Grizzard JD, Spillman DW, Oddi C, Roberts CS, Melchior RD, Mueller GH, Abouzaki NA, Rengel LR, Varma A, Gambill ML, Falcao RA, Voelkel NF, Dinarello CA, Vetrovec GW. Effects of Interleukin-1 Blockade With Anakinra on Adverse Cardiac Remodeling and Heart Failure After Acute Myocardial Infarction [from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) Pilot Study]. Am J Cardiol. 2013;111:1394–1400. doi: 10.1016/j.amjcard.2013.01.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Van Tassell BW, Arena RA, Toldo S, Mezzaroma E, Azam T, Seropian IM, Shah K, Canada J, Voelkel NF, Dinarello CA, Abbate A. Enhanced interleukin-1 activity contributes to exercise intolerance in patients with systolic heart failure. PLoS One. 2012;7:e33438. doi: 10.1371/journal.pone.0033438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rifai N, Tracy RP, Ridker PM. Clinical efficacy of an automated high-sensitivity C-reactive protein assay. Clin Chem. 1999;45:2136–2141. [PubMed] [Google Scholar]
  • 14.Arena R, Myers J, Guazzi M. The clinical and research applications of aerobic capacity and ventilatory efficiency in heart failure: an evidence-based review. Heart Fail Rev. 2008;13:245–269. doi: 10.1007/s10741-007-9067-5. [DOI] [PubMed] [Google Scholar]
  • 15.Kenward MG, Jones B. The analysis of data 2x2 cross-over trials with baseline measurements. Stat Med. 1987;6:911–926. doi: 10.1002/sim.4780060806. [DOI] [PubMed] [Google Scholar]
  • 16.Van Tassell BW, Seropian IM, Toldo S, Mezzaroma E, Abbate A. Interleukin-1β induces a reversible cardiomyopathy in the mouse. Inflamm Res. 2013;62:637–640. doi: 10.1007/s00011-013-0625-0. [DOI] [PubMed] [Google Scholar]
  • 17.Cahalin LP, Chase P, Arena R, Myers J, Bensimhon D, Peberdy MA, Ashley E, West E, Forman DE, Pinkstaff S, Lavie CJ, Guazzi M. A meta-analysis of the prognostic significance of cardiopulmonary exercise testing in patients with heart failure. Heart Fail Rev. 2013;18:79–94. doi: 10.1007/s10741-012-9332-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Arena R, Myers J, Williams MA, Gulati M, Kligfield P, Balady GJ, Collins E, Fletcher G, American Heart Association Committee on Exercise, Rehabilitation, and Prevention of the Council on Clinical Cardiology. American Heart Association Council on Cardiovascular Nursing Assessment of functional capacity in clinical and research settings: a scientific statement from the American Heart Association Committee on Exercise, Rehabilitation, and Prevention of the Council on Clinical Cardiology and the Council on Cardiovascular Nursing. Circulation. 2007;116:329–343. doi: 10.1161/CIRCULATIONAHA.106.184461. [DOI] [PubMed] [Google Scholar]
  • 19.Swank AM, Horton J, Fleg JL, Fonarow GC, Keteyian S, Goldberg L, Wolfel G, Handberg EM, Bensimhon D, Illiou MC, Vest M, Ewald G, Blackburn G, Leifer E, Cooper L, Kraus WE, HF-ACTION Investigators Modest increase in peak VO2 is related to better clinical outcomes in chronic heart failure patients: results from heart failure and a controlled trial to investigate outcomes of exercise training. Circ Heart Fail. 2012;5:579–585. doi: 10.1161/CIRCHEARTFAILURE.111.965186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Redfield MM, Borlaug BA, Lewis GD, Mohammed SF, Semigran MJ, Lewinter MM, Deswal A, Hernandez AF, Lee KL, Braunwald E, Heart Failure Clinical Research Network PhosphdiesteRasE-5 Inhibition to Improve CLinical Status and EXercise Capacity in Diastolic Heart Failure (RELAX) trial: rationale and design. Circ Heart Fail. 2012;5:653–659. doi: 10.1161/CIRCHEARTFAILURE.112.969071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Arena R, Humphrey R, Peberdy MA. Relationship between the Minnesota Living With Heart Failure Questionnaire and key ventilatory expired gas measures during exercise testing in patients with heart failure. J Cardiopulm Rehabil. 2002;22:273–277. doi: 10.1097/00008483-200207000-00010. [DOI] [PubMed] [Google Scholar]
  • 22.Davies LC, Wensel R, Georgiadou P, Cicoira M, Coats AJ, Piepoli MF, Francis DP. Enhanced prognostic value from cardiopulmonary exercise testing in chronic heart failure by non-linear analysis: oxygen uptake efficiency slope. Eur Heart J. 2006;27:684–690. doi: 10.1093/eurheartj/ehi672. [DOI] [PubMed] [Google Scholar]
  • 23.Abbate A, Kontos MC, Grizzard JD, Biondi-Zoccai GG, Van Tassell BW, Robati R, Roach LM, Arena RA, Roberts CS, Varma A, Gelwix CC, Salloum FN, Hastillo A, Dinarello CA, Vetrovec GW, VCU-ART Investigators Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction (Virginia Commonwealth University Anakinra Remodeling Trial [VCU-ART] Pilot study). Am J Cardiol. 2010;105:1371–1377. doi: 10.1016/j.amjcard.2009.12.059. [DOI] [PubMed] [Google Scholar]
  • 24.Poole DC, Hirai DM, Copp SW, Musch TI. Muscle oxygen transport and utilization in heart failure: implications for exercise (in)tolerance. Am J Physiol Heart Circ Physiol. 2012;302:H1050–1063. doi: 10.1152/ajpheart.00943.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Anjan VY, Loftus TM, Burke MA, Akhter N, Fonarow GC, Gheorghiade M, Shah SJ. Prevalence, clinical phenotype, and outcomes associated with normal B-type natriuretic peptide levels in heart failure with preserved ejection fraction. Am J Cardiol. 2012;110:870–876. doi: 10.1016/j.amjcard.2012.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mehra MR, Uber PA, Park MH, Scott RL, Ventura HO, Harris BC, Frohlich ED. Obesity and suppressed B-type natriuretic peptide levels in heart failure. J Am Coll Cardiol. 2004;43:1590–1595. doi: 10.1016/j.jacc.2003.10.066. [DOI] [PubMed] [Google Scholar]
  • 27.Maeder MT, Rickenbacher P, Rickli H, Abbühl H, Gutmann M, Erne P, Vuilliomenet A, Peter M, Pfisterer M, Brunner-La Rocca HP, the TIME-CHF Investigators N-terminal pro brain natriuretic peptide-guided management in patients with heart failure and preserved ejection fraction: findings from the Trial of Intensified versus standard Medical therapy in Elderly patients with Congestive Heart Failure (TIME-CHF). Eur J Heart Fail. 2013 May 8; doi: 10.1093/eurjhf/hft076. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 28.McMurray JJ, Kjekshus J, Gullestad L, et al. Effects of statin therapy according to plasma high-sensitivity C-reactive protein concentration in the Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA): a retrospective analysis. Circulation. 2009;120:2188–2196. doi: 10.1161/CIRCULATIONAHA.109.849117. [DOI] [PubMed] [Google Scholar]
  • 29.Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1β inhibition and the prevention of recurrent cardiovascular events: Rationale and Design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J. 2011;162:597–605. doi: 10.1016/j.ahj.2011.06.012. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES