Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 1.
Published in final edited form as: Circ Heart Fail. 2015 Jan;8(1):221–228. doi: 10.1161/CIRCHEARTFAILURE.114.001598

Nitrate’s Effect on Activity Tolerance in Heart Failure with Preserved Ejection Fraction (NEAT-HFpEF) Trial: Rationale and Design

Rosita Zakeri 1, James A Levine 2, Gabriel A Koepp 2, Barry A Borlaug 1, Julio A Chirinos 3, Martin LeWinter 4, Peter VanBuren 4, Victor G Dávila-Román 5, Lisa de las Fuentes 5, Prateeti Khazanie 6, Adrian Hernandez 6, Kevin Anstrom 6, Margaret M Redfield 1
PMCID: PMC4304404  NIHMSID: NIHMS646715  PMID: 25605640

HEART FAILURE WITH PRESERVED EJECTION FRACTION

The prevalence of heart failure (HF) with preserved ejection fraction (HFpEF) is increasing1. In patients with HFpEF, the burden of symptoms, functional decline, and mortality is high2, and health-related quality of life is poor3. Physicians caring for these patients currently have limited therapeutic options beyond diuresis and management of comorbid conditions. Hence there remains an immediate and critical need for therapies to alleviate symptoms and meaningfully improve quality of life for patients with HFpEF.

ROLE OF NITRATE THERAPY IN HEART FAILURE

Long acting nitrates are used as the cornerstone of anti-anginal therapy and have demonstrated beneficial effects for treatment of patients with heart failure and reduced ejection fraction (HFrEF). In randomized studies, sustained increases in treadmill exercise time4, 5 and peak oxygen consumption6 have been observed at 3 months after initiation of nitrate therapy in patients with HFrEF, including those already treated with angiotensin converting enzyme inhibitors (ACEI)5. Attenuation of pathological left ventricular (LV) remodelling and improved LV systolic function have also been reported5. Although no study has directly examined the effects of nitrate monotherapy on survival in HF, symptom relief is a key management goal in patients with HFpEF, whose primary chronic symptom is often exercise limitation7.

Practice guidelines for the management of chronic HF from the American College of Cardiology/American Heart Association (ACC/AHA)8 and Heart Failure Society of America (HFSA)9 advocate a potential role for nitrates in diminishing symptoms in HFpEF but acknowledge the lack of supportive data and the risk of excessive nitrate-induced hypotension in elderly HFpEF patients. Therefore, it is desirable that a randomized, controlled evaluation of the efficacy and tolerance of nitrate therapy in HFpEF is performed in order to support its therapeutic application.

To address this lack of data and current clinical equipoise for nitrate therapy in HFpEF, the Nitrate’s Effect on Activity Tolerance in Heart Failure with Preserved Ejection Fraction (NEAT-HFpEF) trial (http://clinicaltrials.gov NCT02053493) is being conducted within the National Heart, Lung, and Blood Institute-sponsored HF clinical research network. Cognizant of the primary goal to reduce symptom burden and improve quality of life, NEAT-HFpEF will simultaneously assess a new paradigm of using patient-centric data, i.e. data emanating from and of immediate relevance to patients’ daily living, as the primary efficacy endpoint. Thus NEAT-HFpEF is expected to provide important information regarding nitrate’s safety and therapeutic benefit as well as the feasibility of a novel endpoint, with potential for wider application to future HF studies.

RATIONALE FOR NITRATE THERAPY IN HFPEF

Hemodynamic Effects

A fundamental hemodynamic derangement in HFpEF is pathologic elevation in LV filling pressure, at rest or on exertion7, 10. Commonly used organic nitrates, isosorbide dinitrate (ISDN), isosorbide-5-mononitrate (ISMN), and nitroglycerin, reduce ventricular preload by increasing peripheral venous capacitance, reducing LV filling pressure and wall stress11, 12. At higher doses, dilatation of pulmonary and systemic resistance vessels occurs13, particularly in patients with high arterial pressures14. Coronary artery disease is prevalent in HFpEF, and symptoms of angina may occur in patients without angiographically apparent coronary disease15. Nitrate-induced coronary vasodilatation may improve subendocardial perfusion, which could benefit HFpEF patients for whom ischemia is a contributory factor. Nitrate induced pre-load reduction may also be beneficial in HFpEF where the steep diastolic pressure-volume relationship confers marked increases in LV filling pressures, even at low stroke volumes (SV) and low work rate, prompting early cessation of exercise7. Preload reduction may therefore be expected to improve exercise capacity in HFpEF. Furthermore, nitrates may reduce wave reflections in the arterial tree16, 17, which increase left ventricular late systolic load and wall stress18 and impair diastolic relaxation19.

However, nitrate-induced hemodynamic effects may also be blunted or deleterious in HFpEF. While nitrates reduce arterial impedance and increase SV without causing hypotension in patients with HFrEF, a steeper end-systolic pressure volume relationship in HFpEF means SV increases less and systolic LV pressure decreases more in response to a decrease in preload or afterload2022. In fact, Schwartzenberg et al. observed a reduction in SV among 35% of HFpEF patients following infusion of sodium nitroprusside, suggesting greater vulnerability to excessive preload reduction22. Because deficient SV reserve contributes to exercise limitation in patients with HFpEF23 excessive venodilation from nitrates might offset any beneficial effects on filling pressures, coronary vasodilation or relief of pericardial constraint24. Moreover, HFpEF patients are frequently elderly and may have autonomic dysfunction, chronotropic incompetence and altered baroreflex sensitivity, all of which may exaggerate hypotension with load changes and thus heighten nitrate intolerance25. Finally, whether the potentially favorable effects of nitrates on wave reflections reported in unselected hypertensive subjects also occur in patients with HFpEF is unknown.

Endothelial Effects of Nitrates

Nitrate vasorelaxant responses are thought to be mediated by the formation of nitric oxide (NO) or a closely related entity26. NO activates soluble guanylyl cyclase (sGC) in vascular smooth muscle, prompting synthesis of the second messenger cyclic guanosine monophosphate (cGMP). Downstream activation of cGMP effector proteins, including cGMP-dependent protein kinase (PKG), leads to a reduction in intracellular calcium, and thus to vasodilation. Endothelial-dependent vasodilation is impaired in patients with HFpEF, compared with healthy age-matched controls, and correlates with greater symptoms and poorer exercise capacity23. Exogenous NO delivery or enhancement of endogenous NO biosynthesis may therefore improve endothelial function, as has been observed in HFrEF27.

However, nitrates could also paradoxically worsen endothelial function. Studies in normal humans and experimental animals have shown endothelial dysfunction resulting from chronic nitrate therapy, attributed to the generation of reactive oxygen species (ROS) and local endothelin activation28, 29.

Myocardial Effects of Nitrates

Increased NO bioavailability with nitrates, and thus cGMP/PKG signaling, may acutely improve LV diastolic function and ameliorate myocardial hypertrophic remodeling. Low PKG activity has been implicated in the development of myocardial hypertrophy, delayed relaxation and increased passive stiffness30. Van Heerebeek et al. confirmed both low cGMP content and low PKG activity in myocardial tissue obtained from HFpEF patients, compared with samples from patients with HFrEF and aortic stenosis31. cGMP-phosphodiesterase (PDE5) expression, a mediator of cGMP hydrolysis, was similar between groups. This observation may provide insight into the lack of effect of PDE5 inhibition in HFpEF32 as enhanced cGMP hydrolysis does not appear to mediate the unique reduction in myocardial cGMP in HFpEF31. Rather, nitrotyrosine content (reflecting oxidative/nitrosative stress) was highest in HFpEF31, suggesting that a reduction in NO-stimulated cGMP synthesis due to ROS scavenging of NO and oxidation of the NO target, soluble guanylyl cyclase, underlies low myocardial cGMP content and reduced PKG activity in HFpEF. Activation of cGMP-PKG may also acutely improve diastolic function via phosphorylation of titin33. Acute titin phosphorylation via exogenous administration of PKG results in dramatic reduction in cardiomyocyte stiffness in vitro31. While direct augmentation of PKG improves myocardial diastolic properties in vivo, whether chronic nitrate therapy will enhance cGMP, PKG activity and myocardial diastolic function in HFpEF is unclear.

NITRATE RESISTANCE

Therapeutic responses to nitrates are variable and larger doses are needed to elicit hemodynamic and endothelial responses in patients with HFrEF compared to patients without HF (‘nitrate resistance’)34. Furthermore, early systemic effects, including reflex neurohumoral activation and volume expansion, may lead to reversal of initial hemodynamic benefits (‘nitrate pseudotolerance’)34.

Prolonged exposure is widely recognized to induce true nitrate tolerance34, 35. This is thought to involve vascular processes such as impaired nitrate biotransformation, increased ROS production with impaired clearance, sGC desensitization to NO, enhanced sensitivity to endogenous vasoconstrictors, and increased cGMP phophodiesterase activity, all of which inactivate nitrate vasodilator effects34. The extent of tolerance is somewhat dose-related and low-doses or intermittent dosing regimens with low-nitrate or nitrate-free intervals may be sufficient to prevent its occurrence35, 36. Although, a combination of hydralazine and nitrates is suggested to reduce nitrate tolerate and is utilized in HFrEF, the additional vasodilation and afterload reduction imparted by hydralazine may prove excessive in HFpEF due to the aforementioned differences in pressure-volume relationships, and may mask a beneficial effect of lone nitrate therapy in HFpEF. Furthermore, tolerance differs between nitrate preparations, as once daily ISMN was shown to be devoid of tolerance in patients with coronary artery disease37, 38. Therefore, once daily lone ISMN therapy has been selected for NEAT-HFpEF.

ASSESSMENT OF SYMPTOM BURDEN IN HF

To determine whether nitrates are effective at reducing symptom burden, selection of an appropriate “functional” end-point is preferable to conventional disease-related outcomes. Functional performance refers to the ability to perform day-to-day activities to: “meet basic needs, fulfil usual roles, and maintain health and wellbeing.”39 Such activities often require much less than maximal exertion and may not be accurately summarized by the peak exertional measures used in recent HF trials. Likewise, intermittent evaluation of submaximal exercise capacity, such as the 6MWD, provides low density data which are subject to coaching effects and may underestimate the true burden of disease, for example when patients voluntarily reduce activity to avoid symptoms. Accelerometer-assessed activity is a novel endpoint which may circumvent these limitations by providing high density, quantitative data from continuous assessment of physical activity during usual daily life.

Prior recent studies in patients with HFrEF, have demonstrated correlations between accelerometer data and New York Heart Association (NYHA) functional class40, 6 minute walk distance (6MWD)41, 42, peak oxygen consumption40, 43, and estimated (Seattle Heart Failure Model) or observed mortality risks44, 45. Studies have also shown increases in accelerometer-assessed activity after cardiac resynchronization therapy, confirming its ability to reflect therapeutic response41, 46, 47. Validity, analytical issues, and compliance with externally worn accelerometer devices have been addressed in clinical trials for patients with COPD, among whom age and activity level are likely comparable to elderly patients with HFpEF4851. Notably, patients who may be too frail to undergo comprehensive cardiopulmonary exercise testing will still be eligible for a study using accelerometry, thereby addressing concerns about the spectrum of disease severity in recent HFpEF trials32, 52.

Importantly, cardiopulmonary diseases, or even therapy, are not the only determinants of physical activity. Behavioral and psychological factors as well as the patient’s immediate environment and social support, may have an impact on habitual physical activity. Thus, while accelerometer-assessed physical activity represents a novel, feasible, and patient-centric method to detect a clinically relevant response to nitrate therapy in ambulatory patients with HFpEF, whether enhanced exercise capacity will translate into increased daily physical activity in HFpEF patients remains uncertain .

RATIONALE AND DESIGN OF NEAT-HFPEF

NEAT-HFpEF is a multicenter, randomized (1:1), double-blind, placebo-controlled, crossover study designed to test the hypothesis that once daily extended-release ISMN, at a maximally tolerated dose (30–120mg), improves daily physical activity in patients with HFpEF. Daily activity will be assessed by two hip-worn tri-axial accelerometers and the primary endpoint will be a within-patient comparison of 14-day averaged arbitrary accelerometer units (AAU14) achieved during the ISMN treatment phase, compared with placebo. Because the ability to carry out usual daily activities is a pervasive marker of symptom burden, and yet may be highly variable between patients, the novel endpoint and crossover design used in NEAT-HFpEF are uniquely suited to address the primary hypothesis while taking heed of nitrate pharmacology and pragmatic sample size.

Study protocol and dose-titration schedule

Approximately 110 patients with chronic stable HF and preserved ejection fraction (EF≥50%) will be enrolled. Specific entry criteria stipulate that activity limitation is primarily due to HF symptoms (Table). Eligible participants undergo baseline assessment (echocardiography, blood sampling for biomarkers including cGMP and N-terminal pro-brain type natriuretic peptide [NT-proBNP], 6MWD, symptom questionnaires: Minnesota Living with Heart Failure Questionnaire [MLWHFQ] and the Kansas City Cardiomyopathy Questionnaire [KCCQ]) followed by training in accelerometer use. Participants are subsequently randomized to one of two treatment groups, placebo first with crossover to ISMN or ISMN first with crossover to placebo, stratified by clinical site (permuted block randomization) to ensure equal distribution of participants per arm per site.

Table.

NEAT-HFpEF Eligibility criteria

Inclusion criteria
  1. Age ≥ 50 years

  2. Symptoms of dyspnea (II–IV) without non-cardiac or ischemic etiology

  3. EF ≥ 50%

  4. One of the following within the last 12 months
    • Previous hospitalization for HF or
    • Catheterization documented elevated filling pressures at rest (LVEDP≥15 or PCWP≥20) or with exercise (PCWP≥25) or
    • Elevated NT-proBNP (> 400 pg/ml) or BNP (> 200 pg/ml) or
    • Echo evidence of diastolic dysfunction / elevated filling pressures (at least two)
      • E/A > 1.5 + decrease in E/A of > 0.5 with Valsalva
      • Deceleration time ≤ 140 ms
      • PVs<PVd (sinus rhythm)
      • E/e’≥15
      • LA enlargement (≥ moderate)
      • PASP > 35 mmHg
      • Evidence of LVH
        • LV mass/BSA ≥ 96 (♀) or ≥ 116 (♂) g/m2
        • RWT ≥ 0.43 (♂ or ♀)
        • Posterior wall thickness ≥ 0.9 (♀) or 1.0 (♂) cm
  5. No chronic nitrate therapy or not using (≤ 1× week) intermittent sublingual GTN

  6. Ambulatory (not wheelchair/scooter dependent)

  7. Heart failure is primary factor limiting activity as indicated by answering # 2 to the following question:

    My ability to be active is most limited by:
    1. Joint, foot, leg, hip or back pain
    2. Shortness of breath and/or fatigue and/or chest pain
    3. Unsteadiness or dizziness
    4. Lifestyle, weather, or I just don’t like to be active
  8. Does not regularly swim or do water aerobics as primary form of exercise

Primary Exclusion Criteria
  1. Recent (< 3 months) hospitalization for heart failure or acute coronary syndrome

  2. Previous adverse reaction to the nitrates which necessitated withdrawal of therapy

  3. Therapy with phosphodiesterase type 5 (PDE-5) inhibitors

  4. Documentation of previous EF < 50%

  5. Hemodynamic instability, rare causes of HFpEF, significant primary valve disease or severe hematologic, liver or kidney disease.

All participants enter a 2-week ‘run-in’ phase during which accelerometer data obtained reflects baseline physical activity off nitrates. Thereafter, participants commence once daily placebo or oral ISMN according to the dose titration schedule in Figure 1. At each titration step, study staff will discuss tolerability and determine safety to proceed with up-titration. In the case of drug intolerance, participants will be instructed to down-titrate to the previously tolerated dose or discontinue as necessary. At the end of phase 1, repeat assessment is performed (blood sampling, 6MWD and symptom questionnaires) and all participants receive a new accelerometer device and study drug supply and begin a 2-week drug-free washout phase. Dose-titration is then performed as before. Participants are called weekly and encouraged to be active within the limitations imposed by their HF symptoms. After completion of phase 2 patients undergo a final in-person assessment as at the end of phase 1.

Figure 1.

Figure 1

Study protocol and dose-titration schedule for NEAT-HFpEF. * or maximally tolerated dose; BL, Baseline; WO, Washout; wk, week

Crossover study design considerations

As per the crossover study design, each participant will serve as their own control. Avoiding between-participant variation in estimating the intervention effect enables a smaller sample size and timely completion. External social and behavioral related factors influencing exercise capacity would also be expected to vary little during the trial, thus the within-subject comparisons will remain robust. The modest total treatment duration (4 weeks) was selected to minimize potential bias due to period effects or remodeling (carry-over effect). In particular, nitrates have a rapid onset and offset of action and the 2-week washout phase was determined based on nitrate pharmacokinetics and literature review17, 38, 5355. The key hypothesis to be tested in NEAT-HFpEF is that nitrate hemodynamic effects provide acute symptom relief in HFpEF and that symptom relief will translate into an increase in activity levels. Importantly, NEAT-HFpEF is not designed to evaluate chronic remodeling effects associated with nitrate therapy and, in the event of a negative outcome, should not preclude further investigation into these.

ENDPOINTS

Primary Endpoint

As outlined above, the primary endpoint for NEAT-HFpEF will be a within-patient comparison of accelerometer-assessed physical activity averaged over 14-days between the ISMN and placebo phases.

Technical details

Patient factors

Each patient will wear two external, hip worn (belt-attached) accelerometer devices (Figure 2) throughout the study duration, which will also provide variability data. The devices will not display activity or step counts to the patient. Device settings are pre-programmed and the accelerometers will remain switched on throughout the entire measurement period to minimize patient handling. Baseline training and weekly study calls will address participant-specific strategies to enhance steadfast use including during sleep. Participants will be advised to remove the belt only during bathing or swimming.

Figure 2.

Figure 2

Figure 2

The NEAT-HFpEF accelerometer device.

Accelerometer measures

The tri-axial accelerometer (piezoelectric) sensor measures physical activity in terms of acceleration (movement) values along the vertical (x), anteroposterior (y) and mediolateral (z) axes over time. The accelerometer unit samples these measurements 16 times per second, and the raw output generated, i.e. the change in voltage vector from one sample to the next is converted into a digital series of numbers known as Kionix-based arbitrary accelerometer units (AAU; Kionix is the integrated circuit accelerometer supplier) according to the formula: (x1x2)2+(y1y2)2+(z1z2)2where subscript1 = time point 1, subscript2 = time point 2, and time points one and two are 1/16th of a second apart. Data are continuously recorded and cumulative AAUs are stored in 15-minute epochs (assigned recording periods) providing a total of 96 data points per day. In NEAT-HFpEF these AAU data points will be averaged over a period of 14 days (area under the curve integration) to provide a single measure of AAU14, reflecting the average accelerometer-assessed physical activity during habitual free-living conditions, for each individual, for each phase. All raw data collected will be available for analysis, irrespective of activity level. This is important as some commercially available devices eliminate data corresponding to walking speeds less than approximately 1.5 miles per hour and would therefore be unsuitable for sedentary persons with HFpEF.

At present, there are no consistent cutoffs for classifying accelerometer-assessed activity units into recognized intensity levels for direct clinical interpretation56. Furthermore, NEAT-HFpEF represents the first study within a dedicated HFpEF population to use accelerometer-assessed physical activity as a primary endpoint. Therefore, in addition to within-patient comparisons with and without nitrate therapy, NEAT-HFpEF will also define the typical AAU14 value and standard deviation relevant for an HFpEF population and future studies.

Secondary Endpoints

NEAT-HFpEF is powered to detect a clinically meaningful change in the 6MWD, the KCCQ score, and the MLHFQ score. Perceived dyspnea and effort (Borg) scores during 6MWD, and changes in plasma NTproBNP concentration will also be assessed. Additional accelerometer endpoints will include: hours active, the slope of daily-averaged AAU during study drug administration, and area under the curve for daily-averaged AAU during study drug administration. Patient preference for study phase will also be assessed via a standardized questionnaire.

Tertiary Endpoints

Tertiary endpoints will include the quotients of 6MWD and associated Borg score (integrated measure of performance and symptoms), plasma cGMP concentration and, where available, accelerometer assessed dose-response to nitrate therapy.

Subgroup Analysis

Prespecified subgroup exploratory analyses include comparison of nitrate efficacy according to treatment (or absence of treatment) with agents reported to reduce nitrate tolerance (renin angiotensin aldosterone antagonists35, 57, carvedilol58, statins59, or hydralazine35, 60), plasma NT-proBNP level, systolic blood pressure, and presence or absence of coronary artery disease. An analysis confined to patients on “on drug” will also be performed.

STATISTICAL CONSIDERATIONS

Analyses will be conducted on a modified intention-to-treat basis and include all randomized participants who complete both phases. The primary endpoint is based on the within-patient comparison of AAU14 achieved during the maximally-tolerated dose period of ISMN versus placebo, i.e. using accelerometer data obtained during weeks 5–6 versus weeks 11–12. For missing data corresponding to periods when the device-belt is not worn (bathing or water activities) the imputation approach of Catellier et al. will be used to create a pseudo-complete dataset61. The imputation plan will also account for potential differences in activity between weekdays and weekends. The primary analysis will involve a mixed model with fixed effect terms for the sequence, period (study phase) and treatment62. A random effect term will be included to account for the correlated measurement within each participant63. Baseline characteristics (demographics, echocardiographic data, biomarkers, and questionnaire scores) will be used for adjusted and subgroup analyses. Data from the first phase (baseline to maximum tolerated dose) and second phase (washout to maximum tolerated dose) will be presented separately in a sensitivity analysis, thereby including any patients who may have only completed one phase. For secondary and tertiary endpoints, continuous outcomes will be assessed using mixed models, as for the primary analysis; binary outcomes will be assessed using chi-square tests and Fisher’s exact test, for unadjusted comparisons.

Sample Size and Power

As the primary endpoint has not previously been examined in the proposed study population, the justification for sample size is based on two key secondary endpoints: the overall summary score from the KCCQ and 6MWD.

Data from the recently completed Xanthine Oxidase Inhibition for Hyperuricemic Heart Failure Patients (EXACT-HF) trial64 suggest the within-patient standard deviation (SD) for KCCQ overall summary score is approximately 17 points. A clinically significant difference is considered to be 5 points and a moderately large clinical difference, 10points65, 66 Assuming a two-sided type I error (α) of 0.05, based on a crossover ANOVA63 and a 17-point SD in KCCQ summary score, a total of 94 participants (47 per sequence), would have 80% power to detect a difference of 5 points in KCCQ summary score. 6MWD data from the RELAX trial32 suggest the within-patient SD is ≈ 90 meters. Based on a clinically meaningful difference of 43 meters67, a sample size greater than 60 participants (30 per sequence) would have greater than 90% power to detect this difference in this 2*2 crossover design. The NEAT-HFpEF sample size is larger than that used in previous crossover studies which established nitrate efficacy for HFrEF and angina5, 5355, 6870.

We have also previously measured accelerometer-assessed activity as the AAU14 at baseline, 3 months and 6 months in 49 elderly sedentary volunteers receiving no intervention71. The average within-patient SD between baseline (mean 4462 AAU) and 3 months (mean 4496 AAU) was 337 AAU. If the baseline AAU14 and the within-patient variability in HFpEF patients are similar to those observed in healthy elderly sedentary persons, NEAT-HFpEF would have 90% power to detect a difference between the intervention and placebo phases of 114 AAU (approximately 2.5% of the baseline measurement).

SAFETY

Potential inadvertent unblinding of treatment due to nitrate-related side effects including headache, and rarely, lightheadedness and syncope is recognized., Once daily extended release ISMN has been chosen as the study drug for NEAT-HFpEF, with gradual up-titration from a low dose at weekly intervals in order to minimize the risk of severe side effects and enhance tolerability. ISMN is the active metabolite of ISDN and at 60 or 120mg dose has a plasma elimination half-life of approximately 6 hours38, thus the dosing regimen in NEAT-HFpEF will provide a low-nitrate interval per 24 hours. Co-administration of phosphodiesterase-5 inhibitors will be prohibited due to the risk of excessive hypotension.

CONCLUSIONS

While expert consensus guidelines recommend consideration of nitrates in HFpEF, there are currently no data supporting this recommendation. The NEAT-HFpEF trial will address this critical question and determine whether nitrate pharmacodynamic effects may be leveraged for acute symptom relief among ambulatory patients with HFpEF. Moreover, in using accelerometer-assessed activity as the primary endpoint, NEAT-HFpEF will ascertain the true impact of nitrate therapy on patients’ physical activity during daily living and establish the clinical utility of this novel and patient-centric endpoint for future HF trials.

Acknowledgments

Sources of Funding

This work was supported by grants from the National Heart, Lung and Blood Institute (coordinating center: U10 HL084904; regional clinical centers: U10 HL110312, U109 HL110337, U10 HL110342, U10 HL110262, U10 HL110297, U10 HL110302, U10 HL110309, U10 HL110336, U10 HL110338).

http://Clinicaltrials.gov Identifier: NCT02053493, http://clinicaltrials.gov

Footnotes

Disclosures

None.

References

  • 1.Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in prevalence and outcome of heart failure with preserved ejection fraction. N Engl J Med. 2006;355:251–259. doi: 10.1056/NEJMoa052256. [DOI] [PubMed] [Google Scholar]
  • 2.Smith GL, Masoudi FA, Vaccarino V, Radford MJ, Krumholz HM. Outcomes in heart failure patients with preserved ejection fraction: Mortality, readmission, and functional decline. J Am Coll Cardiol. 2003;41:1510–1518. doi: 10.1016/s0735-1097(03)00185-2. [DOI] [PubMed] [Google Scholar]
  • 3.Hoekstra T, Lesman-Leegte I, van Veldhuisen DJ, Sanderman R, Jaarsma T. Quality of life is impaired similarly in heart failure patients with preserved and reduced ejection fraction. Eur J Heart Fail. 2011;13:1013–1018. doi: 10.1093/eurjhf/hfr072. [DOI] [PubMed] [Google Scholar]
  • 4.Leier CV, Huss P, Magorien RD, Unverferth DV. Improved exercise capacity and differing arterial and venous tolerance during chronic isosorbide dinitrate therapy for congestive heart failure. Circulation. 1983;67:817–822. doi: 10.1161/01.cir.67.4.817. [DOI] [PubMed] [Google Scholar]
  • 5.Elkayam U, Johnson JV, Shotan A, Bokhari S, Solodky A, Canetti M, Wani OR, Karaalp IS. Double-blind, placebo-controlled study to evaluate the effect of organic nitrates in patients with chronic heart failure treated with angiotensin-converting enzyme inhibition. Circulation. 1999;99:2652–2657. doi: 10.1161/01.cir.99.20.2652. [DOI] [PubMed] [Google Scholar]
  • 6.Franciosa JA, Goldsmith SR, Cohn JN. Contrasting immediate and long-term effects of isosorbide dinitrate on exercise capacity in congestive heart failure. Am J Med. 1980;69:559–566. doi: 10.1016/0002-9343(80)90468-4. [DOI] [PubMed] [Google Scholar]
  • 7.Borlaug BA, Nishimura RA, Sorajja P, Lam CS, Redfield MM. Exercise hemodynamics enhance diagnosis of early heart failure with preserved ejection fraction. Circ Heart Fail. 2010;3:588–595. doi: 10.1161/CIRCHEARTFAILURE.109.930701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS, Ganiats TG, Jessup M, Konstam MA, Mancini DM, Michl K, Oates JA, Rahko PS, Silver MA, Stevenson LW, Yancy CW. 2009 focused update incorporated into the acc/aha 2005 guidelines for the diagnosis and management of heart failure in adults: A report of the american college of cardiology foundation/american heart association task force on practice guidelines: Developed in collaboration with the international society for heart and lung transplantation. Circulation. 2009;119:e391–479. doi: 10.1161/CIRCULATIONAHA.109.192065. [DOI] [PubMed] [Google Scholar]
  • 9.Heart Failure Society Of A. Evaluation and management of patients with heart failure and preserved left ventricular ejection fraction. Journal of cardiac failure. 2006;12:e80–85. doi: 10.1016/j.cardfail.2005.11.016. [DOI] [PubMed] [Google Scholar]
  • 10.Penicka M, Bartunek J, Trakalova H, Hrabakova H, Maruskova M, Karasek J, Kocka V. Heart failure with preserved ejection fraction in outpatients with unexplained dyspnea: A pressure-volume loop analysis. J Am Coll Cardiol. 2010;55:1701–1710. doi: 10.1016/j.jacc.2009.11.076. [DOI] [PubMed] [Google Scholar]
  • 11.Battock DJ, Levitt PW, Steele PP. Effects of isosorbide dinitrate and nitroglycerin on central circulatory dynamics in coronary artery disease. Am Heart J. 1976;92:455–458. doi: 10.1016/s0002-8703(76)80044-0. [DOI] [PubMed] [Google Scholar]
  • 12.Elkayam U. Nitrates in the treatment of congestive heart failure. Am J Cardiol. 1996;77:41C–51C. doi: 10.1016/s0002-9149(96)00188-9. [DOI] [PubMed] [Google Scholar]
  • 13.Packer M, Medina N, Yushak M, Lee WH. Comparative effects of captopril and isosorbide dinitrate on pulmonary arteriolar resistance and right ventricular function in patients with severe left ventricular failure: Results of a randomized crossover study. Am Heart J. 1985;109:1293–1299. doi: 10.1016/0002-8703(85)90354-0. [DOI] [PubMed] [Google Scholar]
  • 14.Haber HL, Simek CL, Bergin JD, Sadun A, Gimple LW, Powers ER, Feldman MD. Bolus intravenous nitroglycerin predominantly reduces afterload in patients with excessive arterial elastance. J Am Coll Cardiol. 1993;22:251–257. doi: 10.1016/0735-1097(93)90841-n. [DOI] [PubMed] [Google Scholar]
  • 15.Hwang SJ, Melenovsky V, Borlaug BA. Implications of coronary artery disease in heart failure with preserved ejection fraction. J Am Coll Cardiol. 2014;63:2817–2827. doi: 10.1016/j.jacc.2014.03.034. [DOI] [PubMed] [Google Scholar]
  • 16.Yaginuma T, Avolio A, O’Rourke M, Nichols W, Morgan JJ, Roy P, Baron D, Branson J, Feneley M. Effect of glyceryl trinitrate on peripheral arteries alters left ventricular hydraulic load in man. Cardiovasc Res. 1986;20:153–160. doi: 10.1093/cvr/20.2.153. [DOI] [PubMed] [Google Scholar]
  • 17.Stokes GS, Barin ES, Gilfillan KL. Effects of isosorbide mononitrate and aii inhibition on pulse wave reflection in hypertension. Hypertension. 2003;41:297–301. doi: 10.1161/01.hyp.0000049622.07021.4f. [DOI] [PubMed] [Google Scholar]
  • 18.Chirinos JA, Segers P, Gillebert TC, Gupta AK, De Buyzere ML, De Bacquer D, St John-Sutton M, Rietzschel ER. Arterial properties as determinants of time-varying myocardial stress in humans. Hypertension. 2012;60:64–70. doi: 10.1161/HYPERTENSIONAHA.112.190710. [DOI] [PubMed] [Google Scholar]
  • 19.Gillebert TC, Lew WY. Influence of systolic pressure profile on rate of left ventricular pressure fall. The American journal of physiology. 1991;261:H805–813. doi: 10.1152/ajpheart.1991.261.3.H805. [DOI] [PubMed] [Google Scholar]
  • 20.Chen CH, Nakayama M, Nevo E, Fetics BJ, Maughan WL, Kass DA. Coupled systolic-ventricular and vascular stiffening with age: Implications for pressure regulation and cardiac reserve in the elderly. J Am Coll Cardiol. 1998;32:1221–1227. doi: 10.1016/s0735-1097(98)00374-x. [DOI] [PubMed] [Google Scholar]
  • 21.Kawaguchi M, Hay I, Fetics B, Kass DA. Combined ventricular systolic and arterial stiffening in patients with heart failure and preserved ejection fraction: Implications for systolic and diastolic reserve limitations. Circulation. 2003;107:714–720. doi: 10.1161/01.cir.0000048123.22359.a0. [DOI] [PubMed] [Google Scholar]
  • 22.Schwartzenberg S, Redfield MM, From AM, Sorajja P, Nishimura RA, Borlaug BA. Effects of vasodilation in heart failure with preserved or reduced ejection fraction implications of distinct pathophysiologies on response to therapy. J Am Coll Cardiol. 2012;59:442–451. doi: 10.1016/j.jacc.2011.09.062. [DOI] [PubMed] [Google Scholar]
  • 23.Borlaug BA, Olson TP, Lam CS, Flood KS, Lerman A, Johnson BD, Redfield MM. Global cardiovascular reserve dysfunction in heart failure with preserved ejection fraction. J Am Coll Cardiol. 2010;56:845–854. doi: 10.1016/j.jacc.2010.03.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ludbrook PA, Byrne JD, Kurnik PB, McKnight RC. Influence of reduction of preload and afterload by nitroglycerin on left ventricular diastolic pressure-volume relations and relaxation in man. Circulation. 1977;56:937–943. doi: 10.1161/01.cir.56.6.937. [DOI] [PubMed] [Google Scholar]
  • 25.Borlaug BA, Redfield MM. Diastolic and systolic heart failure are distinct phenotypes within the heart failure spectrum. Circulation. 2011;123:2006–2013. doi: 10.1161/CIRCULATIONAHA.110.954388. ; discussion 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ignarro LJ, Lippton H, Edwards JC, Baricos WH, Hyman AL, Kadowitz PJ, Gruetter CA. Mechanism of vascular smooth muscle relaxation by organic nitrates, nitrites, nitroprusside and nitric oxide: Evidence for the involvement of s-nitrosothiols as active intermediates. The Journal of pharmacology and experimental therapeutics. 1981;218:739–749. [PubMed] [Google Scholar]
  • 27.Schwarz M, Katz SD, Demopoulos L, Hirsch H, Yuen JL, Jondeau G, LeJemtel TH. Enhancement of endothelium-dependent vasodilation by low-dose nitroglycerin in patients with congestive heart failure. Circulation. 1994;89:1609–1614. doi: 10.1161/01.cir.89.4.1609. [DOI] [PubMed] [Google Scholar]
  • 28.Thomas GR, DiFabio JM, Gori T, Parker JD. Once daily therapy with isosorbide-5-mononitrate causes endothelial dysfunction in humans: Evidence of a free-radical-mediated mechanism. J Am Coll Cardiol. 2007;49:1289–1295. doi: 10.1016/j.jacc.2006.10.074. [DOI] [PubMed] [Google Scholar]
  • 29.Oelze M, Knorr M, Kroller-Schon S, Kossmann S, Gottschlich A, Rummler R, Schuff A, Daub S, Doppler C, Kleinert H, Gori T, Daiber A, Munzel T. Chronic therapy with isosorbide-5-mononitrate causes endothelial dysfunction, oxidative stress, and a marked increase in vascular endothelin-1 expression. European heart journal. 2013;34:3206–3216. doi: 10.1093/eurheartj/ehs100. [DOI] [PubMed] [Google Scholar]
  • 30.Paulus WJ, Tschope C. A novel paradigm for heart failure with preserved ejection fraction: Comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol. 2013;62:263–271. doi: 10.1016/j.jacc.2013.02.092. [DOI] [PubMed] [Google Scholar]
  • 31.van Heerebeek L, Hamdani N, Falcao-Pires I, Leite-Moreira AF, Begieneman MP, Bronzwaer JG, van der Velden J, Stienen GJ, Laarman GJ, Somsen A, Verheugt FW, Niessen HW, Paulus WJ. Low myocardial protein kinase g activity in heart failure with preserved ejection fraction. Circulation. 2012;126:830–839. doi: 10.1161/CIRCULATIONAHA.111.076075. [DOI] [PubMed] [Google Scholar]
  • 32.Redfield MM, Chen HH, Borlaug BA, Semigran MJ, Lee KL, Lewis G, LeWinter MM, Rouleau JL, Bull DA, Mann DL, Deswal A, Stevenson LW, Givertz MM, Ofili EO, O’Connor CM, Felker GM, Goldsmith SR, Bart BA, McNulty SE, Ibarra JC, Lin G, Oh JK, Patel MR, Kim RJ, Tracy RP, Velazquez EJ, Anstrom KJ, Hernandez AF, Mascette AM, Braunwald E. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: A randomized clinical trial. Jama. 2013;309:1268–1277. doi: 10.1001/jama.2013.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kruger M, Kotter S, Grutzner A, Lang P, Andresen C, Redfield MM, Butt E, dos Remedios CG, Linke WA. Protein kinase g modulates human myocardial passive stiffness by phosphorylation of the titin springs. Circ Res. 2009;104:87–94. doi: 10.1161/CIRCRESAHA.108.184408. [DOI] [PubMed] [Google Scholar]
  • 34.Munzel T, Daiber A, Gori T. Nitrate therapy: New aspects concerning molecular action and tolerance. Circulation. 2011;123:2132–2144. doi: 10.1161/CIRCULATIONAHA.110.981407. [DOI] [PubMed] [Google Scholar]
  • 35.Gupta D, Georgiopoulou VV, Kalogeropoulos AP, Marti CN, Yancy CW, Gheorghiade M, Fonarow GC, Konstam MA, Butler J. Nitrate therapy for heart failure: Benefits and strategies to overcome tolerance. JACC Heart failure. 2013;1:183–191. doi: 10.1016/j.jchf.2013.03.003. [DOI] [PubMed] [Google Scholar]
  • 36.Elkayam U, Roth A, Mehra A, Ostrzega E, Shotan A, Kulick D, Jamison M, Johnston JV, Rahimtoola SH. Randomized study to evaluate the relation between oral isosorbide dinitrate dosing interval and the development of early tolerance to its effect on left ventricular filling pressure in patients with chronic heart failure. Circulation. 1991;84:2040–2048. doi: 10.1161/01.cir.84.5.2040. [DOI] [PubMed] [Google Scholar]
  • 37.Chrysant SG, Glasser SP, Bittar N, Shahidi FE, Danisa K, Ibrahim R, Watts LE, Garutti RJ, Ferraresi R, Casareto R. Efficacy and safety of extended-release isosorbide mononitrate for stable effort angina pectoris. Am J Cardiol. 1993;72:1249–1256. doi: 10.1016/0002-9149(93)90292-k. [DOI] [PubMed] [Google Scholar]
  • 38.Gunasekara NS, Noble S. Isosorbide 5-mononitrate: A review of a sustained-release formulation (imdur) in stable angina pectoris. Drugs. 1999;57:261–277. doi: 10.2165/00003495-199957020-00016. [DOI] [PubMed] [Google Scholar]
  • 39.Coyne KS, Allen JK. Assessment of functional status in patients with cardiac disease. Heart Lung. 1998;27:263–273. doi: 10.1016/s0147-9563(98)90038-3. [DOI] [PubMed] [Google Scholar]
  • 40.Jehn M, Schmidt-Trucksass A, Hanssen H, Schuster T, Halle M, Koehler F. Association of physical activity and prognostic parameters in elderly patients with heart failure. J Aging Phys Act. 2011;19:1–15. doi: 10.1123/japa.19.1.1. [DOI] [PubMed] [Google Scholar]
  • 41.Kadhiresan VA, Pastore J, Auricchio A, Sack S, Doelger A, Girouard S, Spinelli JC. A novel method–the activity log index–for monitoring physical activity of patients with heart failure. Am J Cardiol. 2002;89:1435–1437. doi: 10.1016/s0002-9149(02)02364-0. [DOI] [PubMed] [Google Scholar]
  • 42.Jehn M, Schmidt-Trucksaess A, Schuster T, Hanssen H, Weis M, Halle M, Koehler F. Accelerometer-based quantification of 6-minute walk test performance in patients with chronic heart failure: Applicability in telemedicine. J Card Fail. 2009;15:334–340. doi: 10.1016/j.cardfail.2008.11.011. [DOI] [PubMed] [Google Scholar]
  • 43.van den Berg-Emons R, Balk A, Bussmann H, Stam H. Does aerobic training lead to a more active lifestyle and improved quality of life in patients with chronic heart failure? Eur J Heart Fail. 2004;6:95–100. doi: 10.1016/j.ejheart.2003.10.005. [DOI] [PubMed] [Google Scholar]
  • 44.Shoemaker MJ, Curtis AB, Vangsnes E, Dickinson MG, Paul R. Analysis of daily activity data from implanted cardiac defibrillators: The minimum clinically important difference and relationship to mortality/life expectancy. World J Cardiovasc Dis. 2012;2:129–135. [Google Scholar]
  • 45.Conraads VM, Spruit MA, Braunschweig F, Cowie MR, Tavazzi L, Borggrefe M, Hill MR, Jacobs S, Gerritse B, van Veldhuisen DJ. Physical activity measured with implanted devices predicts patient outcome in chronic heart failure. Circ Heart Fail. 2014;7:279–287. doi: 10.1161/CIRCHEARTFAILURE.113.000883. [DOI] [PubMed] [Google Scholar]
  • 46.Kawabata M, Fantoni C, Regoli F, Raffa S, Pastori F, Fratini S, Prentice J, Klein HU, Auricchio A. Activity monitoring in heart failure patients with cardiac resynchronization therapy. Circ J. 2007;71:1885–1892. doi: 10.1253/circj.71.1885. [DOI] [PubMed] [Google Scholar]
  • 47.Gilliam FR, 3rd, Kaplan AJ, Black J, Chase KJ, Mullin CM. Changes in heart rate variability, quality of life, and activity in cardiac resynchronization therapy patients: Results of the hf-hrv registry. Pacing Clin Electrophysiol. 2007;30:56–64. doi: 10.1111/j.1540-8159.2007.00582.x. [DOI] [PubMed] [Google Scholar]
  • 48.Lores V, Garcia-Rio F, Rojo B, Alcolea S, Mediano O. Recording the daily physical activity of copd patients with an accelerometer: An analysis of agreement and repeatability. Archivos de bronconeumologia. 2006;42:627–632. doi: 10.1016/s1579-2129(07)60004-4. [DOI] [PubMed] [Google Scholar]
  • 49.Hecht A, Ma S, Porszasz J, Casaburi R. Methodology for using long-term accelerometry monitoring to describe daily activity patterns in copd. COPD. 2009;6:121–129. doi: 10.1080/15412550902755044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Jehn M, Schmidt-Trucksass A, Meyer A, Schindler C, Tamm M, Stolz D. Association of daily physical activity volume and intensity with copd severity. Respir Med. 2011;105:1846–1852. doi: 10.1016/j.rmed.2011.07.003. [DOI] [PubMed] [Google Scholar]
  • 51.Waschki B, Spruit MA, Watz H, Albert PS, Shrikrishna D, Groenen M, Smith C, Man WD, Tal-Singer R, Edwards LD, Calverley PM, Magnussen H, Polkey MI, Wouters EF. Physical activity monitoring in copd: Compliance and associations with clinical characteristics in a multicenter study. Respir Med. 2012;106:522–530. doi: 10.1016/j.rmed.2011.10.022. [DOI] [PubMed] [Google Scholar]
  • 52.Edelmann F, Wachter R, Schmidt AG, Kraigher-Krainer E, Colantonio C, Kamke W, Duvinage A, Stahrenberg R, Durstewitz K, Loffler M, Dungen HD, Tschope C, Herrmann-Lingen C, Halle M, Hasenfuss G, Gelbrich G, Pieske B. Effect of spironolactone on diastolic function and exercise capacity in patients with heart failure with preserved ejection fraction: The aldo-dhf randomized controlled trial. JAMA. 2013;309:781–791. doi: 10.1001/jama.2013.905. [DOI] [PubMed] [Google Scholar]
  • 53.Wisenberg G, Roks C, Nichol P, Goddard MD. Sustained effect of and lack of development of tolerance to controlled-release isosorbide-5-mononitrate in chronic stable angina pectoris. The American journal of cardiology. 1989;64:569–576. doi: 10.1016/0002-9149(89)90480-3. [DOI] [PubMed] [Google Scholar]
  • 54.Svendsen JH, Aldershvile J, Abildgaard U, Amtorp O. Efficacy of controlled-release isosorbide-5-mononitrate as adjunctive treatment to beta-blocking agents in patients with stable angina pectoris. J Cardiovasc Pharmacol. 1989;14:358–363. doi: 10.1097/00005344-198909000-00002. [DOI] [PubMed] [Google Scholar]
  • 55.Rinaldi CA, Linka AZ, Masani ND, Avery PG, Jones E, Saunders H, Hall RJ. Randomized, double-blind crossover study to investigate the effects of amlodipine and isosorbide mononitrate on the time course and severity of exercise-induced myocardial stunning. Circulation. 1998;98:749–756. doi: 10.1161/01.cir.98.8.749. [DOI] [PubMed] [Google Scholar]
  • 56.Matthew CE. Calibration of accelerometer output for adults. Medicine and science in sports and exercise. 2005;37:S512–522. doi: 10.1249/01.mss.0000185659.11982.3d. [DOI] [PubMed] [Google Scholar]
  • 57.Pizzulli L, Hagendorff A, Zirbes M, Fehske W, Ewig S, Jung W, Luderitz B. Influence of captopril on nitroglycerin-mediated vasodilation and development of nitrate tolerance in arterial and venous circulation. American heart journal. 1996;131:342–349. doi: 10.1016/s0002-8703(96)90364-6. [DOI] [PubMed] [Google Scholar]
  • 58.Watanabe H, Kakihana M, Ohtsuka S, Sugishita Y. Randomized, double-blind, placebo-controlled study of carvedilol on the prevention of nitrate tolerance in patients with chronic heart failure. J Am Coll Cardiol. 1998;32:1194–1200. doi: 10.1016/s0735-1097(98)00392-1. [DOI] [PubMed] [Google Scholar]
  • 59.Liuni A, Luca MC, Di Stolfo G, Uxa A, Mariani JA, Gori T, Parker JD. Coadministration of atorvastatin prevents nitroglycerin-induced endothelial dysfunction and nitrate tolerance in healthy humans. J Am Coll Cardiol. 2011;57:93–98. doi: 10.1016/j.jacc.2010.07.037. [DOI] [PubMed] [Google Scholar]
  • 60.Munzel T, Kurz S, Rajagopalan S, Thoenes M, Berrington WR, Thompson JA, Freeman BA, Harrison DG. Hydralazine prevents nitroglycerin tolerance by inhibiting activation of a membrane-bound nadh oxidase. A new action for an old drug. The Journal of clinical investigation. 1996;98:1465–1470. doi: 10.1172/JCI118935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Catellier DJ, Hannan PJ, Murray DM, Addy CL, Conway TL, Yang S, Rice JC. Imputation of missing data when measuring physical activity by accelerometry. Med Sci Sports Exerc. 2005;37:S555–562. doi: 10.1249/01.mss.0000185651.59486.4e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mills EJ, Chan AW, Wu P, Vail A, Guyatt GH, Altman DG. Design, analysis, and presentation of crossover trials. Trials. 2009;10:27. doi: 10.1186/1745-6215-10-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Senn S. Cross-over trials in clinical research. (2) 2002 [Google Scholar]
  • 64.Givertz MM, Mann DL, Lee KL, Ibarra JC, Velazquez EJ, Hernandez AF, Mascette AM, Braunwald E. Xanthine oxidase inhibition for hyperuricemic heart failure patients: Design and rationale of the exact-hf study. Circ Heart Fail. 2013;6:862–868. doi: 10.1161/CIRCHEARTFAILURE.113.000394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Spertus J, Peterson E, Conard MW, Heidenreich PA, Krumholz HM, Jones P, McCullough PA, Pina I, Tooley J, Weintraub WS, Rumsfeld JS. Monitoring clinical changes in patients with heart failure: A comparison of methods. Am Heart J. 2005;150:707–715. doi: 10.1016/j.ahj.2004.12.010. [DOI] [PubMed] [Google Scholar]
  • 66.Flynn KE, Lin L, Moe GW, Howlett JG, Fine LJ, Spertus JA, McConnell TR, Pina IL, Weinfurt KP. Relationships between changes in patient-reported health status and functional capacity in outpatients with heart failure. Am Heart J. 2012;163:88–94. e83. doi: 10.1016/j.ahj.2011.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.O’Keeffe ST, Lye M, Donnellan C, Carmichael DN. Reproducibility and responsiveness of quality of life assessment and six minute walk test in elderly heart failure patients. Heart. 1998;80:377–382. doi: 10.1136/hrt.80.4.377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Handler CE, Sullivan ID. Double-blind randomised crossover trial comparing isosorbide dinitrate cream and oral sustained-release tablets in patients with angina pectoris. Int J Cardiol. 1985;7:149–157. doi: 10.1016/0167-5273(85)90356-0. [DOI] [PubMed] [Google Scholar]
  • 69.Uusitalo A. Long term efficacy of a controlled-release formulation of isosorbide 5-mononitrate (imdur) in angina patients receiving beta-blockers. Drugs. 1987;33(Suppl 4):111–117. doi: 10.2165/00003495-198700334-00020. [DOI] [PubMed] [Google Scholar]
  • 70.Parker JD, Parker JO. Nitrate therapy for stable angina pectoris. N Engl J Med. 1998;338:520–531. doi: 10.1056/NEJM199802193380807. [DOI] [PubMed] [Google Scholar]
  • 71.Thompson WG, Kuhle CL, Koepp GA, McCrady-Spitzer SK, Levine JA. “Go4life” exercise counseling, accelerometer feedback, and activity levels in older people. Arch Gerontol Geriatr. 2014;58:314–319. doi: 10.1016/j.archger.2014.01.004. [DOI] [PubMed] [Google Scholar]

RESOURCES