/    /  II.5 Clinical Evaluations – Endpoints
Translational Pathway for Transcatheter Aortic Valves

Clinical Evaluations – Endpoints

Author

Ori Ben-Yehuda, MD

play-sharp-fill

Ori Ben-Yehuda, MD

Introduction

Endpoint selection is of utmost importance and an essential part of trial design. Clearly, the endpoints should be clinically relevant and provide an opportunity to demonstrate the presence of a meaningful impact of the intervention. Cardiovascular study endpoints can be divided to “hard” endpoints such as mortality, myocardial infarction, and stroke; intermediate endpoints such as hospitalizations for heart failure; and “soft” endpoints which can include symptoms, quality of life measures (e.g., New York Heart Association [NYHA] class), and surrogate measures previously proven to correlate with clinically relevant endpoints (e.g., ejection fraction [EF]). Earlier transcatheter aortic valve replacement (TAVR) trials, such as the early PARTNER (1,2) or CoreValve (3) trials, compared TAVR to surgical or medical only treatments and included inoperable patients or those at high surgical risk. Understandably, the population in these trials had high rates of “hard” endpoints and statistically proven differences could be demonstrated with a reasonable sample size. Current and next phase TAVR trials are enrolling lower-risk patients, some asymptomatic or at an earlier stage of aortic valve disease. Since event rates may be lower and effect size smaller, a large sample size will be needed to prove benefit. The primary method to reduce sample size is to use “soft,” or surrogate, endpoints and/or to combine several endpoints to one composite endpoint with its inherent limitations (4-6). A somewhat different approach, combining endpoints in a hierarchical clinically intuitive way can be achieved by the Finkelstein and Schoenfeld method (7) or the Pocock Win Ratio (8).

In this section, we will review how innovative approaches to clinical trial design can inform future trials in aortic stenosis therapy.

Aortic Stenosis

Aortic stenosis (AS) is the most common type of valvular heart disease in the western world (9). Our current understanding of disease progression in AS relies on small datasets, some more than 50 years old. In the 1930s, the cardinal features of AS — angina pectoris, syncope, and heart failure (HF) — were recognized as indicating rapid deterioration after a long asymptomatic period (10-12). The seminal work of Ross and Braunwald, cited repeatedly to this day, matched different survival periods to the onset of angina, syncope, or HF, highlighting the importance of intervention once symptoms developed (13).  Earlier intervention in select patients is an area of emerging interest, tied to a more detailed understanding of the natural history of AS. Current guidelines (14-16) recommend aortic valve replacement in symptomatic patients with severe AS (Class I recommendation) or asymptomatic patients with severe AS and EF <50%. Asymptomatic patients may also be referred for intervention in the presence of a pathological stress test (Class IIa) or in patients undergoing other cardiac surgery (Class I for severe AS and Class IIa for moderate AS). Advances in medicine throughout the years and better management of risk factors, coupled with the introduction of balloon valvuloplasty in 1986 (17) and TAVR in 2002 (18) may alter the risk-benefit ratio and favor earlier intervention. While TAVR is a valid alternative to surgical aortic valve replacement (SAVR) in high- and intermediate-risk symptomatic patients (16), the question of whether the reduced risk of TAVR should affect the timing of AS intervention and expand the indication for intervention to asymptomatic and low-risk patients is being determined in ongoing trails detailed below.

Current TAVR Trials

Several trials are trying to assess the utility of TAVR in lower-risk populations than those studied thus far. Endpoints and statistical analysis vary between trials. EARLY TAVR (Evaluation of Transcatheter Aortic Valve Replacement Compared to Surveillance for Patients With Asymptomatic Severe Aortic Stenosis; NCT03042104) is enrolling 1,109 asymptomatic patients with severe AS to either TAVR or regular clinical surveillance. The nonhierarchical primary endpoint is a composite of all-cause death, stroke, or unplanned cardiovascular hospitalization. The secondary endpoint is death or disabling stroke. While asymptomatic, the study population is expected to be older, with baseline comorbidities, and a Society of Thoracic Surgeons (STS) score that is not necessarily low, and so event rates are expected to be high enough for a reasonable sample size trial. On the other hand, the PARTNER 3 trial (NCT02675114) is randomizing 1,328 patients with severe AS and low surgical risk (STS <4) to TAVR or SAVR. The primary endpoint is the composite of all-cause mortality, all stroke, and rehospitalization. Since event rates are not expected to be high in this population, the composite endpoint will be evaluated by a noninferiority analysis based on a relative noninferiority margin of 35%. A noninferiority design provides an opportunity to utilize “hard” endpoints such as mortality and stroke with a reasonable relative margin. Finally, the TAVR UNLOAD trial (NCT02661451) is enrolling 600 patients with moderate AS and HF randomized to either TAVR or optimal medical therapy. The primary outcome is a hierarchical composite of death, disabling stroke, hospitalizations and Kansas City Cardiomyopathy Questionnaire (KCCQ) change from baseline. Combining these endpoints increases event rates and allows enrollment of a smaller sample size with sufficient power to demonstrate an expected significant difference. The hierarchical ordering provides further clinical validity, as described below.

While device versus device trials have been infrequent, they may highlight important differences in secondary endpoints such as paravalvular leak, rate of permanent pacemaker implantation, and vascular complications.

Surrogate Endpoints

Patients with AS suffer from exercise intolerance (usually due to HF or chest pain), overall poor quality of life (QOL), and frequent hospitalizations. Mortality is also increased in this population. However, running clinical trials testing new devices aimed at showing a reduction in mortality rates has become increasingly difficult due to requirements for a large patient population and a long duration of follow-up. Justifiably, a search for more prevalent, yet clinically meaningful, surrogate or “soft” endpoints has produced numerous candidates. The advantages of using surrogate endpoints include not only a higher number of events with every patient contributing during a shorter follow up, but also capturing clinically relevant endpoints affecting the everyday life of patients such as QOL or functional capacity measures. The clear caveat of using “soft” endpoints is that association is not necessarily causal (19). Moreover, some endpoints are difficult to combine into one composite endpoint.

Possible surrogate endpoints for TAVR trials include: 1) imaging endpoints, such as echocardiography (valvular function, EF) and magnetic resonance imaging (e.g., left ventricular [LV] hypertrophy reduction, infarct size); 2) functional capacity endpoints (e.g., 6-minute walking test [6MWT], exercise treadmill test, cardiopulmonary exercise test); 3) quality of life measures with such examples as generic questionnaires such as SF-36/SF-12 (20), EQ-5 (21), and disease specific ones such as Seattle angina questionnaire (22), Minnesota Living with Heart Failure Questionnaire (MLWHFQ) (23), and KCCQ (24,25); and 4) HF worsening assessed by rates of hospitalizations/hospitalization equivalents, and biomarkers (e.g., N-terminal pro-B-type natriuretic peptide [NT-proBNP], STS).

Imaging

Valvular intervention trials regularly utilize imaging to assess device performance. Long-term echocardiographic follow-up provides important information when comparing trial arms. Nevertheless, to reach a clinically meaningful and valid conclusion, a trial must contain a clinically meaningful endpoint and cannot rely solely on imaging. Imaging endpoints may also be combined into a single composite endpoint. EARLY TAVR, for example, uses a combination of imaging measurements as a secondary endpoint termed “Integrated measure of LV health,” which includes LV global longitudinal strain, LV mass index, and left atrial volume index.

Functional Capacity Measures

Rigorous assessment of exercise tolerance as an endpoint is useful because it not only assesses the impact on a primary limitation faced by the study population but also correlates with mortality and rates of hospitalizations. Moreover, exercise measures such as cardiopulmonary exercise testing (CPET)/peak oxygen consumption (peak VO2) seem to outperform subjective measures (e.g., NYHA class, KCCQ) in predicting future “hard” outcomes. These outcomes have been validated in HF trials and should be suitable for trials assessing interventions for aortic valve disease. They are frequently accompanied by HF symptoms.

New York Heart Association Classification

The NYHA classification, first presented in 1928 (26) and extensively incorporated in HF guidelines (27), is often used for patient classification and risk stratification. While clearly having prognostic implications, this traditional four group division is subjective and limited in assessing more subtle changes in health status that may still be clinically meaningful.

Exercise Treadmill Test (ETT)

Physical fitness assessed by an ETT has long been shown to be a predictor of mortality (28). ETT was further incorporated into the guidelines with recommendations for AVR in asymptomatic patients with a positive test. Furthermore, ETT may also be used for risk stratification and prognostication in asymptomatic patients with AS (29). The use of ETT as a comparison tool between trial arms or to compare patients before and after intervention has been limited and impacted by comorbidities such as respiratory and orthopedic disease.

6-Minute Walking Test

The 6MWT in use today has been repeatedly modified since its inception as a 15-minute-run field test (30). A shorter 12-minute walk test was first used for the assessment of chronic bronchitis (31) and later shortened to 6 minutes to assess respiratory disease (32) and heart failure (33). The test is also frequently used in the evaluation and management of pulmonary hypertension (34). The primary measure is distance walked in a hallway during an uninterrupted 6-minute period. The test is most useful in those with limited exercise tolerance and in those limited by symptoms since patients are allowed brea ks (timer continues), a significant advantage over regular ETT assessment in the population enrolled in TAVR trials. The 6MWT has been shown to provide added prognostic information to the Euroscore in patients undergoing aortic valve replacement (35). In the TAVR population, the test seems sensitive enough to demonstrate improved function following TAVR and adds to risk stratification (36-39). A sub-analysis of the PARTNER trial showed that 6MWT improved following TAVR and that baseline 6MWT did not predict procedural outcomes but did predict long-term mortality (40). 6MWT is useful in medically managed patients with aortic stenosis as well (41). With proven prognostic implications, 6MWT seems a plausible, safe, and clinically meaningful surrogate marker in the population enrolled in TAVR trials.

Cardiopulmonary Exercise Test

CPET is an important tool in assessing functional capacity and carries prognostic implications (42). The test can be performed on a treadmill or a bicycle and several different protocols have been established. In HF, the cornerstone study by Mancini et. al. established the validity of peak VO2 as a strong prognostic factor (43) and set the foundation for defining the cutoff value of 14ml/kg/min in heart transplant candidates (44). Other informative parameters from CPET (albeit less substantiated than peak VO2) include exercise capacity, the ventilatory anaerobic threshold and ventilatory expired gas parameters (VE/Vco2 slope). CPET has been assessed in patients with aortic stenosis, mainly for risk stratification and management guidance as compared to ETT and stress echocardiography (45). A baseline CPET that shows no hemodynamic compromise suggests conservative treatment is associated with a good prognosis (46).

In conclusion, impaired exercise capacity is a cardinal manifestation of HF and/or aortic valve disease and improvement in exercise capacity is a meaningful endpoint for patients. Exercise capacity (i.e., peak VO2 and 6MWT distance) closely relates to outcomes across the spectrum of cardiovascular disease, particularly HF. Measurements of exercise capacity are safe and increasingly simple to perform, and each has advantages and limitations (Table 1). Done properly with core lab oversight, these results are highly reproducible and permit ascertainment of clinically meaningful endpoints. Despite providing less prognostic information, 6MWT is frequently preferred over CPET due to ease of administration and the lack of expertise or equipment required.

 

Table 1. Functional Capacity Measures

Advantages Limitations
NYHA
  1. Well validated, widespread use and proven prognostic implications
  1. Subjective
  2. Limited in assessing subtle changes
Exercise Treadmill Test
  1. Useful in risk stratification and treatment decisions (guidelines)
  1. Limited use in AS population due to comorbidities (e.g., orthopedic restrictions)
6-Minute Walking Test
  1. Simple, easily administered
  2. Reflects activities of daily living
  3. High test-retest reliability (47)
  4. Independently predicts HF hospitalization and mortality (48)
  5. Changes reflect response to therapy (e.g., following LVAD implantation [49], ferric treatment [50], and CRT treatment [51])
  1. Some training effect
  2. No clear standardization of instructions and performance of the test
  3. Does not provide information about volitional effort or other limiting organ systems (e.g., orthopedic problems, Parkinson’s disease)
  4. Large standard deviation
  5. Represents varying degrees of maximum effort, performs less well in less sick HF patients
Cardiopulmonary Exercise Test
  1. Peak VO2 is currently the gold standard measure of cardiopulmonary function (52)
  2. Objective, more precise and reproducible than other measures of physical function. Can provide information on sub-maximal exercise capacity as well.
  3. Symptoms of exercise intolerance in AS, such as dyspnea and fatigue, result from a complex interplay of mechanisms originating from both the central and peripheral components of the oxygen transport system (ventricle-valve-vasculature relationship) and may be captured by CPET
  4. Correlates with all-cause mortality and all-cause hospitalization in HF patients (53)
  5. Demonstrated response to therapy (54-56)
  1. Equipment and expertise in performing the test are required. Core lab central analysis is recommended to ensure proper equipment calibration, validate procedures and adjudicate data in a blinded and consistent manner
  2. Some studies have shown significant inter-and intra-reader variability, and a clear threshold is not always discernable

AS = aortic stenosis; CPET = cardiopulmonary exercise test; CRT = cardiac resynchronization therapy; HF = heart failure; LVAD = left ventricular assist device; NYHA = New York Heart Association.

QOL Measures

Although difficult to measure, health is not just the absence of disease but also the presence of physical, mental, and social well-being (57). Quantifying HF has been largely based on the presence of symptoms (NYHA classification) and the consequences of the disease/symptoms (e.g., mortality and HF hospitalizations). Aside from reducing mortality (especially sudden death), treatment goals in AS are to improve quality of life by reducing symptoms. HF treatments individually proven to impact mortality/disease progression have been demonstrated to affect QOL, such as medications (LCZ696 [58] and valsartan [59] but neutral results in a meta-analysis of beta-blockers [60]) , cardiac resynchronization therapy (61), and LV assist device use (49). Several questionnaires were constructed to better quantify disease burden, such as the MLHFQ introduced in 1987 and repeatedly validated since (62,63), the KCCQ (24), and the less known Chronic HF Questionnaire (64). These and other assessment tools have been compared with conflicting results (65-67).

The well-validated and widely used KCCQ was formulated to “create a valid, sensitive, disease-specific health status measure for patients with congestive heart failure (CHF)” and claimed to better assess clinical improvement and subcategories of QOL compared to other questionnaires (24). The score has several domains including physical limitation, symptoms (frequency, severity and change), self-efficacy, social, and QOL and has also been published in a shortened format (25). The KCCQ has been shown to be more sensitive to clinical change (assessed by blinded cardiologists) than other measures, including NYHA classification and 6MWT (68). An analysis of 1,358 patients from EPHESUS (Eplerenone’s Neurohormonal Efficacy and Survival Study) demonstrated the value of serial KCCQ assessments in predicting mortality and the combined endpoint of cardiovascular death and hospitalization, after multivariable analysis (hazard ratio [HR]: 1.09; 95% confidence interval [CI]: 1.00 to 1.18; and HR: 1.11; 95% CI: 1.05 to 1.17 for each 5-point decrease in KCCQ, respectively) (69). In an analysis of the PARADIGM-HF trial (58), LCZ696 treatment in HF patients demonstrated higher KCCQ scores in surviving patients (70). Although no specific tool exists for quality of life assessment in patients with AS, current generic health assessment tools such as KCCQ and MLHFQ have been used successfully. A sub-analysis from the PARTNER trial demonstrated KCCQ was reliable, sensitive enough to capture a difference post-TAVR (uncaptured by NYHA), and prognostic (71). Current-era TAVR trials (e.g., TAVR UNLOAD, EARLY TAVR, and PARTNER 3) all include KCCQ as a secondary endpoint or part of the composite primary endpoint.

While QOL provides a methodological assessment than can be easily compared between trials, it is limited in that it is somewhat subjective and easily biased by missing data.

QOL improvement is an important treatment goal and should be clearly quantified to allow comparisons and support fine-tuning therapy. Several validated measurement tools are available, with the widespread use of the KCCQ in clinical trials, providing it with some advantage over other questionnaires.

HF Worsening -- Hospitalization, Hospitalization Equivalents, and Neurohormones

Any hospitalization, time to first hospitalization, or accumulating hospitalizations during follow-up are all clinically meaningful and acceptable endpoints. In the PARADIGM-HF trial, LCZ696 treatment significantly reduced the risk of hospitalization (58).  A sub-analysis of the PARADIGM-HF trial further demonstrated that combining HF hospitalization equivalents (outpatient intensification of HF therapy and emergency department visits) with HF hospitalizations and cardiovascular death into one composite endpoint contributed a significant number of events and shortened the time to achieve these event rates (72). Understandably, all current-era TAVR trials incorporate hospitalization (HF or valve related) or rehospitalization in the composite primary endpoint, increasing expected event rates by including a clinically important endpoint.

Current guidelines recommend measuring B-type natriuretic peptide (BNP) and NT-proBNP for HF diagnosis and for establishing prognosis (27). Several systematic reviews reported one or both peptides can independently predict mortality and/or HF hospitalization (73-78). In the PARADIGM-HF trial, NT-proBNP levels decreased with LCZ696 treatment and endpoint event rates were significantly higher in patients with NT-proBNP >1000 pg/ml (79). Consequently, clear definitions to correctly diagnosis HF worsening and provide a foundation for proper comparisons across trials have been published (80). Neurohormone levels change following TAVR (e.g., from 642 ± 634 pg/ml to 340 ± 253 pg/ml at 30 days in a small prospective study (37) could be used to suggest reduced wall stress and the effect of therapy. Moreover, several studies demonstrated the utility of measuring BNP or NT-proBNP for risk stratification and prognostic implications, including mortality (81,82).

Composite Endpoints and the Finkelstein Schoenfeld Method

The common use of composite endpoints, intended to increase statistical power and avoid type II error and to account for several effects of a studied intervention, has several limitations. First, pooling together several endpoints that are not considered equal and differ in their clinical importance is problematic. Second, traditional time-to-event statistics fail to capture events occurring after the first endpoint event occurred. Furthermore, death can cause a competing effect on the risk of nonfatal endpoints. Defining each component of the composite as an independent secondary endpoint has been recommended to address these concerns (6).

Several statistical methods aim to deal with the inherent limitations of using a composite endpoint: The Anderson-Gill, an extension of the Cox model, counts repeated events yet considers no dependence between events, which is not always true (83). The competing risk methods, an extension of survival analysis methods, consider the time lost for a potential event and its hazard risk, yet do not account for multiple events (84,85). The weighted composite endpoints, an extension of the regular time-to-event method, assigns a reduced (yet somewhat arbitrary) weight to a patient’s contribution to the cohort following a non-lethal event (86). The Finkelstein and Schoenfeld (FS) method compares each patient to every other patient, while accounting for both time to event and multiple events (7). The Win ratio, a further development of the FS method, also compares between patients, and not only generates a p value as a measure of significance, but also provides a measure of the relative effectiveness of the therapies (8).

FS Method and Pocock Win Ratio

The FS methodology (7) creates a hierarchy for all endpoints that can include clinically relevant hard and soft endpoints, for instance, mortality and quality of life. Each patient in Arm A is compared with each patient in Arm B. For each patient comparison and endpoint in the hierarchy, a win, loss, or tie is declared. If a win (and corresponding loss for the other patient) is declared, there is no need to continue to the next, lower tier, in the hierarchy. If a tie is declared, the comparison moves on to a lower tier. Then, the number of wins is counted in each group and statistically compared (e.g., by the Mann-Whitney test). This method provides the opportunity to increase power (and thus reduce the required sample size) by combining several endpoints yet ordering them in a predetermined, sensible hierarchy of importance to be clinically meaningful (unlike a “simple” composite endpoint). The FS methodology has been used repeatedly before, including in TAVR UNLOAD.

One limitation of the FS method is that the result is hard to interpret clinically, as it does not provide a measure of the magnitude of the difference between the study arms. The Win Ratio (also referred to as the Pocock Win Ratio) provides a ratio of the wins versus losses in the two arms and therefore an estimate of the effect size (8).

VARC-2 and Trial Design

Proper trial design incorporating several endpoints in a clinically meaningful hierarchy provides the opportunity to assess the effect of new therapy in the smallest sample size acceptable. The Valve Academic Research Consortium-2 or VARC-2 has published recommended trial design principles and endpoint definitions (87). The VARC-2 consensus document is meant to provide standardization of endpoint definitions for TAVR studies that will allow easy comparisons between different studies. The consortium acknowledged the need for composite endpoints for achievable sample sizes. The document emphasized the need for composite endpoints to include components that have a similar impact and are expected to trend in the same direction: for example, device success combines mortality and echocardiographic parameters. Clinical efficacy is defined as a composite of mortality, stroke, hospitalization, NYHA class, and echocardiographic parameters. The widespread implementation of these recommendations will help uniform comparisons between treatments and across various trials, and ultimately improve patient care. The use of validated surrogate endpoints, and hierarchical composite endpoints (both “hard” and “soft” endpoints), provide further tools to decrease sample size yet increase clinical validity by capturing several relevant aspects of patient care and treatment success.

References

  1. Leon MB, Smith CR, Mack M, et al.; PARTNER Trial Investigators. Transcatheter aortic-valve implantation for aortic stenosis in patients who cannot undergo surgery. N Engl J Med. 2010;363:1597-607.
  2. Smith CR, Leon MB, Mack MJ, et al.; PARTNER Trial Investigators. Transcatheter versus surgical aortic-valve replacement in high-risk patients. N Engl J Med. 2011;364:2187-98.
  3. Adams DH, Popma JJ, Reardon  MJ, et al. Transcatheter Aortic-Valve Replacement with a Self-Expanding Prosthesis. N Eng J Med. 2014;370:1790-8.
  4. Neaton JD, Gray G, Zuckerman BD, Konstam MA. Key issues in end point selection for heart failure trials: composite end points. J Card Fail. 2005;11:567-75.
  5. Califf RM, Harrelson-Woodlief L, Topol EJ. Left ventricular ejection fraction may not be useful as an end point of thrombolytic therapy comparative trials. 1990;82:1847-53.
  6. Freemantle N, Calvert M, Wood J, Eastaugh J, Griffin C. Composite outcomes in randomized trials: greater precision but with greater uncertainty? 2003;289:2554-9.
  7. Finkelstein DM, Schoenfeld DA. Combining mortality and longitudinal measures in clinical trials. Stat Med. 1999;18:1341-54.
  8. Pocock SJ, Ariti CA, Collier TJ, Wang D. The win ratio: a new approach to the analysis of composite endpoints in clinical trials based on clinical priorities. Eur Heart J. 2012;33:176-82.
  9. Vahanian A, Alfieri O, Andreotti F, et al. Guidelines on the management of valvular heart disease (version 2012): the Joint Task Force on the Management of Valvular Heart Disease of the European Society of Cardiology (ESC) and the European Association for Cardio-Thoracic Surgery (EACTS). Eur J Cardiothorac Surg. 2012;42:S1-44.
  10. Braunwald E. On the natural history of severe aortic stenosis. J Am Coll Cardiol. 1990;15:1018-20.
  11. Contratto AW, Levine SA. Aortic stenosis with special reference to angina pectoris and syncope*. Ann Intern Med. 1937;10:1636-53.
  12. Dry TJ, Willius FA. Calcareous disease of the aortic valve. Am Heart J. 1939;17:138-57.
  13. Ross J, Jr., Braunwald E. Aortic stenosis. 1968;38:61-7.
  14. Baumgartner H, Falk V, Bax JJ, et al. 2017 ESC/EACTS Guidelines for the management of valvular heart disease. Eur Heart J. 2017;38:2739-2791.
  15. Nishimura RA, Otto CM, Bonow RO, et al. 2014 AHA/ACC Guideline for the Management of Patients With Valvular Heart Disease: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2014;129:2440-92.
  16. Nishimura RA, Otto CM, Bonow RO, et al. 2017 AHA/ACC Focused Update of the 2014 AHA/ACC Guideline for the Management of Patients With Valvular Heart Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. 2017;135:e1159-95.
  17. Cribier A, Savin T, Saoudi N, Rocha P, Berland J, Letac B. Percutaneous transluminal valvuloplasty of acquired aortic stenosis in elderly patients: an alternative to valve replacement? 1986;1:63-7.
  18. Cribier A, Eltchaninoff H, Bash A, et al. Percutaneous Transcatheter Implantation of an Aortic Valve Prosthesis for Calcific Aortic Stenosis. First Human Case Description. 2002;106:3006-8.
  19. Fleming TR, DeMets DL. Surrogate end points in clinical trials: are we being misled? Ann Intern Med. 1996;125:605-13.
  20. McHorney CA, Ware JE, Jr., Raczek AE. The MOS 36-Item Short-Form Health Survey (SF-36): II. Psychometric and clinical tests of validity in measuring physical and mental health constructs. Med Care. 1993;31:247-63.
  21. EuroQol–a new facility for the measurement of health-related quality of life. Health Policy. 1990;16:199-208.
  22. Spertus JA, Winder JA, Dewhurst TA, et al. Development and evaluation of the Seattle Angina Questionnaire: a new functional status measure for coronary artery disease. J Am Coll Cardiol. 1995;25:333-41.
  23. Rector TS Kubo SH, Cohn JN. Patients’ Self-Assessment of Their Congestive Heart Failure: Content, Reliability and Validity of a New Measure, the Minnesota Living With Heart Failure Questionnaire. Heart Fail. 1987;3:198-219.
  24. Green CP, Porter CB, Bresnahan DR, Spertus JA. Development and evaluation of the Kansas City Cardiomyopathy Questionnaire: a new health status measure for heart failure. J Am Coll Cardiol. 2000;35:1245-55.
  25. Spertus JA, Jones PG. Development and Validation of a Short Version of the Kansas City Cardiomyopathy Questionnaire. Circ Cardiovasc Qual Outcomes. 2015;8:469-76.
  26. Pardee HEB NYHA. Criteria for the Classification & Diagnosis of Heart Disease. New York, NY:CB Hoober Inc;1928.
  27. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;62:e147-239.
  28. Ekelund LG, Haskell WL, Johnson JL, Whaley FS, Criqui MH, Sheps DS. Physical fitness as a predictor of cardiovascular mortality in asymptomatic North American men. The Lipid Research Clinics Mortality Follow-up Study. N Engl J Med. 1988;319:1379-84.
  29. Rafique AM, Biner S, Ray I, Forrester JS, Tolstrup K, Siegel RJ. Meta-analysis of prognostic value of stress testing in patients with asymptomatic severe aortic stenosis. Am J Cardiol. 2009;104:972-7.
  30. Balke B. A SIMPLE FIELD TEST FOR THE ASSESSMENT OF PHYSICAL FITNESS. REP 63-6. [Report] Civil Aeromedical Research Institute (US). 1963:1-8.
  31. McGavin CR, Gupta SP, McHardy GJ. Twelve-minute walking test for assessing disability in chronic bronchitis. Br Med J. 1976;1:822-3.
  32. Butland RJ, Pang J, Gross ER, Woodcock AA, Geddes DM. Two-, six-, and 12-minute walking tests in respiratory disease. Br Med J (Clin Res Ed). 1982;284:1607-8.
  33. Guyatt GH, Sullivan MJ, Thompson PJ, et al. The 6-minute walk: a new measure of exercise capacity in patients with chronic heart failure. Can Med Assoc J. 1985;132:919-23.
  34. Demir R, Küçükoğlu MS. Six-minute walk test in pulmonary arterial hypertension. Anatol J Cardiol. 2015;15:249-54.
  35. de Arenaza DP, Pepper J, Lees B, et al. Preoperative 6-minute walk test adds prognostic information to Euroscore in patients undergoing aortic valve replacement. 2010;96:113-7.
  36. Gotzmann M, Hehen T, Germing A, et al. Short-term effects of transcatheter aortic valve implantation on neurohormonal activation, quality of life and 6-minute walk test in severe and symptomatic aortic stenosis. 2010;96:1102-6.
  37. Gotzmann M, Bojara W, Lindstaedt M, et al. One-year results of transcatheter aortic valve implantation in severe symptomatic aortic valve stenosis. Am J Cardiol. 2011;107:1687-92.
  38. Mok M, Nombela-Franco L, Urena M, et al. Prognostic Value of Exercise Capacity as Evaluated by the 6-Minute Walk Test in Patients Undergoing Transcatheter Aortic Valve Implantation. J Am Coll Cardiol. 2013;61:897-8.
  39. Bagur R, Rodes-Cabau J, Dumont E, et al. Exercise capacity in patients with severe symptomatic aortic stenosis before and six months after transcatheter aortic valve implantation. Am J Cardiol. 2011;108:258-64.
  40. Green P, Cohen DJ, Généreux P, et al. Relation Between Six-Minute Walk Test Performance and Outcomes After Transcatheter Aortic Valve Implantation (from the PARTNER Trial). Am J Cardiol. 2013;112:700-6.
  41. Castillo-Moreno JA, Garcia-Escribano IA, Martinez-Pascual-de-Riquelme M, et al. Prognostic Usefulness of the 6-Minute Walk Test in Patients With Severe Aortic Stenosis. Am J Cardiol. 2016;118:1239-43.
  42. Albouaini K, Egred M, Alahmar A. Cardiopulmonary exercise testing and its application. Postgrad Med J. 2007;83:675-82.
  43. Mancini DM, Eisen H, Kussmaul W, Mull R, Edmunds LH, Jr., Wilson JR. Value of peak exercise oxygen consumption for optimal timing of cardiac transplantation in ambulatory patients with heart failure. 1991;83:778-86.
  44. Balady GJ, Arena R, Sietsema K, et al. Clinician’s Guide to cardiopulmonary exercise testing in adults: a scientific statement from the American Heart Association. 2010;122:191-225.
  45. Domanski O, Richardson M, Coisne A, et al. Cardiopulmonary exercise testing is a better outcome predictor than exercise echocardiography in asymptomatic aortic stenosis. Int J Cardiol. 2017;227:908-14.
  46. Le VD, Jensen GV, Kjoller-Hansen L. Prognostic Usefulness of Cardiopulmonary Exercise Testing for Managing Patients With Severe Aortic Stenosis. Am J Cardiol. 2017;120:844-9.
  47. Demers C, McKelvie RS, Negassa A, Yusuf S. Reliability, validity, and responsiveness of the six-minute walk test in patients with heart failure. Am Heart J. 2001;142:698-703.
  48. Bittner V, Weiner DH, Yusuf S, et al. Prediction of mortality and morbidity with a 6-minute walk test in patients with left ventricular dysfunction. SOLVD Investigators. 1993;270:1702-7.
  49. Rogers JG, Aaronson KD, Boyle AJ, et al. Continuous flow left ventricular assist device improves functional capacity and quality of life of advanced heart failure patients. J Am Coll Cardiol. 2010;55:1826-34.
  50. Anker SD, Comin Colet J, et al. Ferric carboxymaltose in patients with heart failure and iron deficiency. N Engl J Med. 2009;361:2436-48.
  51. Olsson LG, Swedberg K, Clark AL, Witte KK, Cleland JG. Six minute corridor walk test as an outcome measure for the assessment of treatment in randomized, blinded intervention trials of chronic heart failure: a systematic review. Eur Heart J. 2005;26:778-93.
  52. Cahalin LP, Chase P, Arena R, et al. A meta-analysis of the prognostic significance of cardiopulmonary exercise testing in patients with heart failure. Heart Fail Rev. 2013;18:79-94.
  53. Swank AM, Horton J, Fleg JL, et al. 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;5:579-85.
  54. Hamroff G, Katz SD, Mancini D, et al. Addition of angiotensin II receptor blockade to maximal angiotensin-converting enzyme inhibition improves exercise capacity in patients with severe congestive heart failure. 1999;99:990-2.
  55. Sarullo FM, Fazio G, Puccio D, et al. Impact of ”off-label” use of ivabradine on exercise capacity, gas exchange, functional class, quality of life, and neurohormonal modulation in patients with ischemic chronic heart failure. J Cardiovasc Pharmacol Ther. 2010;15:349-55.
  56. Young JB, Abraham WT, Smith AL, et al. Combined cardiac resynchronization and implantable cardioversion defibrillation in advanced chronic heart failure: the MIRACLE ICD Trial. 2003;289:2685-94.
  57. Testa MA, Simonson DC. Assessment of quality-of-life outcomes. N Engl J Med. 1996;334:835-40.
  58. McMurray JJV, Packer M, Desai AS, et al. Angiotensin–Neprilysin Inhibition versus Enalapril in Heart Failure. N Engl J Med. 2014;371:993-1004.
  59. Majani G, Giardini A, Opasich C, et al. Effect of valsartan on quality of life when added to usual therapy for heart failure: results from the Valsartan Heart Failure Trial. J Card Fail. 2005;11:253-9.
  60. Dobre D, van Jaarsveld CH, deJongste MJ, Haaijer Ruskamp FM, Ranchor AV. The effect of beta-blocker therapy on quality of life in heart failure patients: a systematic review and meta-analysis. Pharmacoepidemiol Drug Saf. 2007;16:152-9.
  61. Veazie PJ, Noyes K, Li Q, et al. Cardiac resynchronization and quality of life in patients with minimally symptomatic heart failure. J Am Coll Cardiol. 2012;60:1940-4.
  62. Rector TS, Cohn JN. Assessment of patient outcome with the Minnesota Living with Heart Failure questionnaire: reliability and validity during a randomized, double-blind, placebo-controlled trial of pimobendan. Pimobendan Multicenter Research Group. Am Heart J. 1992;124:1017-25.
  63. Bilbao A, Escobar A, García-Perez L, Navarro G, Quirós R. The Minnesota living with heart failure questionnaire: comparison of different factor structures. Health Qual Life Outcomes. 2016;14:23.
  64. Guyatt GH, Nogradi S, Halcrow S, Singer J, Sullivan MJ, Fallen EL. Development and testing of a new measure of health status for clinical trials in heart failure. J Gen Intern Med. 1989;4:101-7.
  65. Garin O, Ferrer M, Pont A, et al. Disease-specific health-related quality of life questionnaires for heart failure: a systematic review with meta-analyses. Qual Life Res. 2009;18:71-85.
  66. Garin O, Herdman M, Vilagut G, et al. Assessing health-related quality of life in patients with heart failure: a systematic, standardized comparison of available measures. Heart Fail Rev. 2014;19:359-67.
  67. Napier R, McNulty SE, Eton DT, Redfield MM, AbouEzzeddine O, Dunlay SM. Comparing Measures to Assess Health-Related Quality of Life in Heart Failure With Preserved Ejection Fraction. JACC Heart Fail. 2018;6:552-60.
  68. Spertus J, Peterson E, Conard MW, et al. Monitoring clinical changes in patients with heart failure: a comparison of methods. Am Heart J. 2005;150:707-15.
  69. Kosiborod M, Soto GE, Jones PG, et al. Identifying heart failure patients at high risk for near-term cardiovascular events with serial health status assessments. 2007;115:1975-81.
  70. Lewis EF, Claggett BL, McMurray JJV, et al. Health-Related Quality of Life Outcomes in PARADIGM-HF. Circ Heart Fail. 2017;10:e003430.
  71. Arnold SV, Spertus JA, Lei Y, et al. Use of the Kansas City Cardiomyopathy Questionnaire for monitoring health status in patients with aortic stenosis. Circ Heart Fail. 2013;6:61-7.
  72. Okumura N, Jhund PS, Gong J, et al. Importance of Clinical Worsening of Heart Failure Treated in the Outpatient Setting: Evidence From the Prospective Comparison of ARNI With ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure Trial (PARADIGM-HF). 2016;133:2254-62.
  73. Di Angelantonio E, Chowdhury R, Sarwar N, et al. B-type natriuretic peptides and cardiovascular risk: systematic review and meta-analysis of 40 prospective studies. 2009;120:2177-87.
  74. Doust JA, Pietrzak E, Dobson A, Glasziou P. How well does B-type natriuretic peptide predict death and cardiac events in patients with heart failure: systematic review. 2005;330:625.
  75. Maeda K, Tsutamoto T, Wada A, et al. High levels of plasma brain natriuretic peptide and interleukin-6 after optimized treatment for heart failure are independent risk factors for morbidity and mortality in patients with congestive heart failure. J Am Coll Cardiol. 2000;36:1587-93.
  76. Oremus M, Raina PS, Santaguida P, et al. A systematic review of BNP as a predictor of prognosis in persons with coronary artery disease. Clin Biochem. 2008;41:260-5.
  77. Balion C, Santaguida PL, Hill S, et al. Testing for BNP and NT-proBNP in the diagnosis and prognosis of heart failure. Evid Rep Technol Assess (Full Rep). 2006;142:1-147.
  78. Oremus M, Don-Wauchope A, McKelvie R, et al. BNP and NT-proBNP as prognostic markers in persons with chronic stable heart failure. Heart Fail Rev. 2014;19:471-505.
  79. Zile MR, Claggett BL, Prescott MF, et al. Prognostic Implications of Changes in N-Terminal Pro-B-Type Natriuretic Peptide in Patients With Heart Failure. J Am Coll Cardiol. 2016;68:2425-36.
  80. Hicks KA, Mahaffey KW, Mehran R, et al. 2017 Cardiovascular and Stroke Endpoint Definitions for Clinical Trials. 2018;137:961-72.
  81. Vale NC, Campante Teles R, Madeira S, et al. Post-procedural N-terminal pro-brain natriuretic peptide predicts one-year mortality after transcatheter aortic valve implantation. Rev Port Cardiol. 2018;37:67-73.
  82. Dahou A, Clavel MA, Capoulade R, et al. B-Type Natriuretic Peptide and High-Sensitivity Cardiac Troponin for Risk Stratification in Low-Flow, Low-Gradient Aortic Stenosis: A Substudy of the TOPAS Study. JACC Cardiovasc Imaging. 2018;11:939-47.
  83. Andersen PK, Gill RD. Cox’s Regression Model for Counting Processes: A Large Sample Study. Ann Stat. 1982;10:1100-20.
  84. Wolbers M, Koller MT, Stel VS, et al. Competing risks analyses: objectives and approaches. Eur Heart J. 2014;35:2936-41.
  85. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496-509.
  86. Capodanno D, Gargiulo G, Buccheri S, et al. Computing Methods for Composite Clinical Endpoints in Unprotected Left Main Coronary Artery Revascularization: A Post Hoc Analysis of the DELTA Registry. JACC Cardiovasc Interv. 2016;9:2280-8.
  87. Kappetein AP, Head SJ, Genereux P, et al. Updated standardized endpoint definitions for transcatheter aortic valve implantation: the Valve Academic Research Consortium-2 consensus document (VARC-2). Eur J Cardiothorac Surg. 2012;42:S45-60.
Hide picture