Advances in detection, prevention and treatment of heart failure in type 2 diabetes: part II

ALICE C COWLEY, ABHISHEK DATTANI, EMER M BRADY, GERRY P MCCANN, GAURAV S GULSIN

Department of Cardiovascular Science, University of Leicester and the NIHR Biomedical Research Centre, Glenfield Hospital, Leicester, UK

Address for correspondence: Dr Gaurav S Gulsin
Department of Cardiovascular Science, University of Leicester and the NIHR Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
E-mail: gg149@leicester.ac.uk

Abstract

This review is the second of two that aim to cover the advances in heart failure (HF) prevention, detection and treatment relevant to people with type 2 diabetes (T2DM). Part I focuses on HF classification and prevention, specifically lifestyle changes and primary preventative techniques including smoking cessation, physical activity, weight loss, lipid and glucose control. This concluded: 1) intensive blood glucose control is not in itself a necessary or sufficient treatment target for HF prevention, and a multifaceted preventative approach is likely to have a greater effect; 2) the most compelling evidence for HF risk reduction is for sodium glucose co- transporter 2 inhibitors although glucagon-like peptide 1 receptor agonists may also have a role; and 3) patients likely to derive most benefit are those at highest risk of developing overt HF, which probably represent the majority of people with T2DM. Part II of this review will cover early detection of cardiac dysfunction and treatment of established heart failure. Particular emphasis is placed on heart failure with preserved ejection fraction.

Br J Diabetes 2024;24(1):24-29

https://doi.org/10.15277/bjd.2024.442

Key words: type 2 diabetes, heart failure, heart failure with reduced ejection fraction, heart failure with preserved ejection fraction

Stages C and D – treatment of established heart failure

Diabetes is a complex multisystem disorder, often accompanied by multimorbidity. In many instances, pharmacological management can be challenging due to the interaction between multi-organ dysfunction, drug contraindications or side effects, and variations in guidelines. Wherever possible, cases should be discussed within a multidisciplinary team to ensure that optimum therapies are instituted.1

In the vast majority of cases, management of symptomatic heart failure (HF) in people with type 2 diabetes (T2DM) is the same as for people without T2DM. Goals of treatment via a patient-centred approach are: 1) avoidance of signs and symptoms of congestion, to improve exercise tolerance and quality of life; and 2) rapid initiation, up-titration and maintenance of guideline-directed foundational HF medications to prevent HF hospitalisation and lengthen survival. A detailed description of HF management is beyond the scope of this article and has been extensively reviewed elsewhere.1,2 Herein we summarise only the most recent developments. Due to differences in the efficacy of available treatments, stratification of HF based on left ventricle ejection fraction (LVEF) is necessary; in accordance with most large-scale clinical trials, we define heart failure with preserved ejection fraction (HFpEF) as those with an LVEF >40% and HFrEF as those with an LVEF <40%. Heart failure with improved ejection fraction (HFimpEF) is defined as a baseline LVEF <40%, >10% improvement in LVEF and subsequent LVEF measured at LVEF >40%.3

Heart failure with preserved ejection fraction

Lack of efficacy of traditional heart failure medications
HFpEF is the predominant manifestation of HF in T2DM, accounting for up to 83% of people with T2DM and newly identified HF.4

Until as recently as 2021, none of the established HF medications used to treat HFrEF had been convincingly shown to improve clinical outcomes in people with HFpEF. For example, in 2019 the hotly anticipated PARAGON-HF trial (44% of participlants had diabetes) of the angiotensin-receptor neprilysin-inhibitor sacubitril-valsartan did not demonstrate a reduction in cardiovascular (CV) death or HF hospitalisation in patients with HFpEF (with an LVEF >45%) compared to valsartan.5 Emerging real-world data suggest that beta blockers may in fact be harmful in patients with HFpEF, particularly those with higher ejection fraction (EF).6 Lastly, the mineralocorticoid receptor antagonist spironolactone demonstrated no reduction in CV death and HF hospitalisation in a similar HFpEF cohort to PARAGON-HF,7 although there is some debate regarding the study findings due to inconsistencies in trial data from Russia and Georgia and there may yet be some benefit of spironolactone in HFpEF.8 All in all, treatment options for HFpEF were extremely limited. The FIDELITY pooled analysis investigated the effect of the non-steroidal mineralocorticoid receptor antagonist finerenone, versus placebo, in patients with T2DM and chronic kidney disease.9 Alongside renal protective effects, there was a significant improvement in the composite CV outcome, driven by a reduction in HF hospitalisation.9 Finerenone is not yet established as an alternative to eplerenone or spironolactone, but may be an important consideration for future research.

Sodium glucose co-transporter 2 inhibitors
Publication of the EMPEROR-Preserved and DELIVER trials of the sodium glucose co-transporter 2 inhibitors (SGLT2i) empagliflozin and dapagliflozin, respectively, in people with chronic HFpEF have since changed the landscape of HF management.10,11 Both trials similarly found that treatment with SGLT2i was associated with lower rates of HF hospitalisation compared to placebo, irrespective of the presence of diabetes (Table 1).

Unsurprisingly, there has been intense interest in the mechanisms by which SGLT2i exert their rapid CV effects,12,13 for significant benefits appear in less than two weeks following treatment initiation.14 Several small mechanistic studies utilising advanced cardiac imaging techniques have been published, in which the effects of SGLT2i appear pleiotropic. Small reductions in left ventricle (LV) mass (~5%) and myocardial extracellular volume have been described,15-17 but no convincing improvements in myocardial energetics or blood flow have been demonstrated.18-20 We posit that the modest but multifactorial effects of SGLT2i on lowering blood glucose, body weight and blood pressure, coupled with a diuretic effect, work in tandem targeting several HF pathways.21 In any case, alongside loop diuretics to treat congestion, SGLT2i are now regarded as a foundational treatment in HFpEF and should be prescribed in all eligible patients.

1223 Cowley Table 1

Glucagon-like peptide-1 agonists
Although small trials have shown that lifestyle-mediated weight loss improves exercise capacity in obesity-related HFpEF,22 only modest body weight reductions (~5 to 10%) are achieved even with intensive lifestyle modification and long-term sustainability is limited.23 Alternatively, bariatric surgery achieves more marked (>15%) and sustained weight loss (in up to a fifth of patients),24 remission of diabetes (in up to one third of patients)25 and reductions in downstream major adverse CV events (including HF hospitalisation) in obese patients with CV disease.26 Large-scale availability and fitness for surgery of HF patients with multimorbidity, however, hinders widespread feasibility of bariatric surgery. The emergence of glucagon-like peptide-1 receptor agonists (GLP-1 RA) as safe, tolerable and efficacious weight loss pharmacotherapies has brought new hope for the treatment of obesity and its related complications, including HFpEF.

Obesity-related HFpEF has emerged as a distinct pathological entity, mediated by a combination of direct deleterious effects on cardiac structure and function combined with systemic multi-organ damage.27 Patients with obesity- related HFpEF have more fluid retention, worse symptom burden and lower exercise capacity than those with normal weight HFpEF.28 The STEP-HFpEF Trial was a multicentre, international, placebo-controlled, randomised trial aiming to determine whether treatment with the GLP-1 RA semaglutide, in addition to weight loss, would improve symptom burden and exercise capacity in obesity-related HFpEF, but the trial excluded people with diabetes.29 Compared with placebo, participants in the semaglutide arm experienced an anticipated and marked reduction in body weight (mean change -13.3%, compared with -2.6% for placebo), consistent with previous weight loss trials of semaglutide.30 Crucially, greater improvements in HF symptoms (Kansas City Cardiomyopathy Questionnaire clinical summary score, KCCQ-CSS, the co- primary outcome measure together with percent weight reduction) and exercise capacity (six-minute walk test, a secondary outcome measure) were observed in the active treatment arm compared with the placebo arm. The absolute improvement in HF symptoms with semaglutide was especially promising: KCCQ-CSS increased from ~59 to ~76 points overall (median overall change 16 points), representing a moderate to large clinical improvement in symptoms.31 These improvements in symptoms were far in excess of the change in KCCQ scores in the DELIVER and EMPEROR-Preserved trials, where only small increases of between ~2 to 5 points, respectively, were observed. Less impressive in STEP-HFpEF was the increase in six-minute walk distance, which increased from 316 to 338 metres in the semaglutide arm: a mere 7% improvement in exercise capacity. Although the study did not include people with T2DM, an ongoing trial is looking specifically at a diabetic cohort (STEP HFpEF DM).32 Weight loss treatments in overweight and obese individuals are likely to play a major role in treatment of HFpEF in the future.

Heart failure with reduced ejection fraction
Four cornerstones of therapy and avoidance of treatment inertia
The four cornerstones of pharmacological therapy for HFrEF (Figure 1) are now well recognised,33,34 with beneficial effects of therapy being demonstrated within just 30 days of initiation.33,35,36 The cumulative impact of treatment with all four drug classes in HFrEF has been estimated to represent an absolute risk reduction in all-cause mortality of over 25%, with a number needed to treat of just four patients.37 A principal aspect of contemporary HF treatment is early implementation of guideline-directed medical therapy; far too many patients are not prescribed beneficial disease-modifying therapies that have a cumulative impact on clinical outcomes (Figure 1). Avoidance of treatment inertia, therefore, cannot be emphasised enough. The recent STRONG-HF study demonstrated that rapid uptitration of guideline-directed therapy, after an acute admission with decompensated HF, improved HF symptoms and quality of life whilst reducing the risk of HF readmission and all-cause death at 180 days.38 Deferring treatment of an angiotensin-converting enzyme inhibitor, beta blocker and aldosterone antagonist for one year leads to an absolute increase in mortality of 12 in 100 patients, according to a meta- analysis of randomised controlled trials. This is consistent witha 1% increase in absolute mortality per month delay for ‘low-risk’ HF patients.39

1223 Cowley Figure 1

Despite recognition that treatment inertia in HFrEF has marked impact on patient outcomes, observational studies highlight the scale of the problem. The CHAMP-HF study assessed initiation of therapy for chronic HF in the US and found that just 1% of eligible patients were receiving target doses of guideline-directed therapy at the time.40 More recently, the EVOLUTION-HF study, which evaluated prescribing patterns in the US, Japan and Sweden following HF hospitalisation, noted that delays in prescribing disease-modifying agents were more pronounced for novel therapies and in patients with co- morbidities such as T2DM.41 Alongside education and increased awareness for patients and clinicians, adapting organisational structures and providing an integrated multidisciplinary approach to follow-up can help to tackle clinical inertia.42

Finally, adherence to prescribed medications can often be overlooked. In a cohort of people with T2DM attending primary care, 28.1% did not adhere to medications to treat diabetes, hypertension or lipid-lowering therapies.43 Similarly, in HF cohorts, non-adherence has been reported at between 33.3- 45.9%, and has been shown to have negative effects on clinical outcomes.44,45 For a person living with diabetes, 90% of their disease management is self-care,46 and therefore patient engagement and education is pivotal to providing effective care. Meta-analyses evaluating a variety of strategies to improve adherence, including disease and medication education, self-monitoring and interactions with the multi-disciplinary team, have shown improved mortality and risk of HF hospitalisation in comparison to control cohorts.47,48

Despite the known side effects of therapy, HFrEF is a progressive and high-risk condition where the risks of not initiating appropriate medications are great, especially in people with T2DM.49 Our overarching recommendation for HFrEF treatment in T2DM is to initiate treatment in a timely manner and utilise combination therapy, which is more effective than uptitration of single drug classes.33-35,50 Medication adherence is a key part of HF self-care.47

Heart failure with improved ejection fraction

1223 Cowley key messages

Patients who have historically had a reduced LVEF that has completely (LVEF >50%) or partially (LVEF 40-50%) improved are considered a separate subgroup. It is well recognised that LVEF can improve in HFrEF, but there are limited evidence- based data for these patients.4,51 An expert consensus from the Journal of the American College of Cardiology has defined HF with recovered LVEF as: 1) documentation of baseline LVEF<40%; 2) >10% improvement in LVEF; and 3) subsequent measurement LVEF >40%.4

Despite improvements in morbidity and mortality, HF hospitalisations, exercise tolerance and HF biomarkers, a significant proportion of patients with an HFimpEF remain at risk of deterioration, and it is difficult to identify which patients are at the highest risk.4,51 Molecular changes that occur in adverse LV remodelling remain dysregulated in the recovered ventricle.4 Therefore these patients should be considered as in ‘remission’ rather than cured,4 and current guidance recommends that disease-modifying therapies are continued after LVEF recovers.3,4

Conclusions

Type 2 diabetes and HF are inextricably linked, such that consideration of HF prevention, detection and treatment should remain at the forefront of management of all people with T2DM. We have outlined crucial preventive strategies across the A to D spectrum of HF that could mitigate disease development and progression. In those at risk of developing symptomatic HF (Stages A and B), emphasis should be placed on aggressive risk factor control and early initiation of SGLT2i. In those with established HF (Stages C and D), particularly those with reduced ejection fraction, rapid commencement and maintenance of foundational HF therapies avoids treatment inertia and improves outcomes. Although major advances in HF have been made, there remain multiple outstanding challenges and clinical outcomes are unacceptably poor in people with T2DM, most notably related to early detection of Stage B HF. Urgent work is still needed to facilitate early initiation of preventive treatments.

© 2024. This work is openly licensed via CC BY 4.0

© 2024. This work is openly licensed via CC BY 4.0.

This license enables reusers to distribute, remix, adapt, and build upon the material in any medium or format, so long as attribution is given to the creator. The license allows for commercial use. CC BY includes the following elements: BY – credit must be given to the creator.

Copyright ownership The author(s) retain copyright.

Conflict of interest None.

Funding AC, EMB, and GPM received funding from the National Institute for Health and Care Research (NIHR) United Kingdom through a Research Professorship award (RP-2017-08-ST2-007). AD received funding from the British Heart Foundation through a Clinical Research Training Fellowship (FS/CRTF/20/24069). GSG is funded by the NIHR, through a Clinical Lectureship.

Acknowledgements Figure created using biorender.com.

References

  1. Heidenreich PA, Bozkurt B, Aguilar D, et al. 2022 AHA/ACC/HFSA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association joint committee on clinical practice guidelines. Circulation 2022; 145(18):e895-e1032. https://doi.org/10.1161/CIR.00000000001063
  2. McDonagh TA, Metra M, Adamo M, et al. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 2021;42(36):3599-726. https://doi.org/10.1093/eurheartj/ehab368
  3. Wilcox JE, Fang JC, Margulies KB, Mann DL. Heart failure with recovered left ventricular ejection fraction: JACC scientific expert panel. J Am Coll Cardiol 2020;76(6):719-34. https://doi.org/10.j.jacc.2020.05.075
  4. Boonman-de Winter LJ, Rutten FH, Cramer MJ, et al. High prevalence of previously unknown heart failure and left ventricular dysfunction in patients with type 2 diabetes. Diabetologia 2012;55(8):2154-62. https://doi.org/10.1007/s00125-012-2579-0
  5. Solomon SD, McMurray JJV, Anand IS, et al. Angiotensin–neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med 2019;381(17):1609-20. https://.doi.org/10.1056/NEJMoa1908655
  6. Arnold SV, Silverman DN, Gosch K, et al. Beta-blocker use and heart failure outcomes in mildly reduced and preserved ejection fraction. JACC Heart Fail 2023;11(8 Pt 1):893-900. https://doi.org/10.1016/j.jchf.2023.03.017
  7. Pitt B, Pfeffer MA, Assmann SF, et al. Spironolactone for heart failure with preserved ejection fraction. N Engl J Med 2014;370(15):1383-92. https://doi.org/10.1056/NEJMoa1313731
  8. Pfeffer MA, Claggett B, Assmann SF, et al. Regional variation in patients and outcomes in the Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist (TOPCAT) trial. Circulation 2015;131(1):34-42. https://doi.org/10.1161/CIRCULATIONAHA.114.013255
  9. Agarwal R, Filippatos G, Pitt B, et al. Cardiovascular and kidney outcomes with finerenone in patients with type 2 diabetes and chronic kidney disease: the FIDELITY pooled analysis. European Heart Journal 2022;43(6):474-84. https://doi.org/10.1093/eurheartj/ehab777
  10. Anker SD, Butler J, Filippatos G, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 2021;385(16):1451-61.https://doi.org/10.1056/NEJMoa2107038
  11. Solomon SD, McMurray JJV, Claggett B, et al. Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction. N Engl J Med 2022;387(12):1089-98. https://doi.org/10.1056/NEJMoa2206286
  12. Packer M, Butler J, Zannad F, et al. Effect of empagliflozin on worsening heart failure events in patients with heart failure and preserved ejection fraction: EMPEROR-Preserved trial. Circulation 2021; 144(16):1284-94. https://doi.org/10.1161/CIRCULATIONAHA.121.056824
  13. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015; 373(22):2117-28. https://doi.org/10.1056/NEJMoa1504720
  14. Vaduganathan M, Claggett BL, Jhund P, et al. Time to clinical benefit of dapagliflozin in patients with heart failure with mildly reduced or preserved ejection fraction. JAMA Cardiol 2022;7(12):1259-63. https://doi.org/10.1001/jamacardio.2022.3750
  15. Verma S, Mazer CD, Yan AT, et al. Effect of empagliflozin on left ventricular mass in patients with type 2 diabetes mellitus and coronary artery disease: the EMPA-HEART CardioLink-6 randomized clinical trial. Circulation 2019;140(21):1693-702. https://doi.org/10.1161/CIIRCULATIONAHA.119.042375
  16. Brown AJM, Gandy S, McCrimmon R, Houston JG, Struthers AD, Lang A randomized controlled trial of dapagliflozin on left ventricular hypertrophy in people with type two diabetes: the DAPA-LVH trial. Eur Heart J 2020;41(36):3421032. https://doi.org/10.1093/eurheartj/ehaa419
  17. Mason T, Coelho-Filho OR, Verma S, et al. Empagliflozin reduces myocardial extracellular volume in patients with type 2 diabetes and coronary artery disease. JACC Cardiovasc Imaging 2021;14(6):1164-73.https://doi.org/10.1016/j.jcmg.2020.10.017
  18. Hundertmark MJ, Adler A, Antoniades C, et al. Assessment of cardiac energy metabolism, function, and physiology in patients with heart failure taking empagliflozin: the randomized, controlled EMPA-VISION trial. Circulation 2023;147:1654-69. https://doi.org/10.1161/CIRCULATIONAHA.122.062021
  19. Jurgens M, Schou M, Hasbak P, et al. Effects of empagliflozin on myocardial flow reserve in patients With type 2 diabetes mellitus: the SIMPLE trial. J Am Heart Assoc 2021;10(15):e020418. https://doi.org/10.1161/JAHA.120.020418
  20. Thirunavukarasu S, Jex N, Chowdhary A, et al. Empagliflozin treatment Is associated with improvements in cardiac energetics and function and reductions in myocardial cellular volume in patients with type 2 diabetes. Diabetes 2021;70(12):2810-22. https://doi.org/10.2337db21-0270
  21. Sattar N, Petrie MC, Zinman B, Januzzi JL, Jr. Novel diabetes drugs and the cardiovascular specialist. J Am Coll Cardiol 2017;69(21):2646-56.https://doi.org/10.1016/j.jacc.2017.04.014
  22. Kitzman DW, Brubaker P, Morgan T, et al. Effect of caloric restriction or aerobic exercise training on peak oxygen consumption and quality of life in obese older patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 2016;315(1):36-46. https://doi.org/10.1001/jama.2015.17346
  23. Papamargaritis D, le Roux CW, Holst JJ, Davies MJ. New therapies for obesity. Cardiovasc Res 2024;119(18):2825-42. https://doi.org/10.1093/cvr/cvac176
  24. Sjostrom L, Peltonen M, Jacobson P, et al. Bariatric surgery and long- term cardiovascular events. JAMA 2012;307(1):56-65. https://doi.org/10.1001/jama.2011.1914
  25. Sjostrom L, Peltonen M, Jacobson P, et al. Association of bariatric surgery with long-term remission of type 2 diabetes and with microvascular and macrovascular complications. JAMA 2014; 311(22):2297-304. https://doi.org/10.1001/jama.2014.5988
  26. Doumouras AG, Wong JA, Paterson JM, et al. Bariatric surgery and cardiovascular outcomes in patients with obesity and cardiovascular disease: a population-based retrospective cohort study. Circulation 2021; 143(15):1468-80. https://doi.org/10.1161/CIRCULATIONAHA.120.052386
  27. Borlaug BA, Jensen MD, Kitzman DW, Lam CSP, Obokata M, Rider OJ. Obesity and heart failure with preserved ejection fraction: new insights and pathophysiological targets. Cardiovasc Res 2023; 118(18):3434-50. https://doi.org/10.1093/cvr/cvac120
  28. Reddy YNV, Lewis GD, Shah SJ, et al. Characterization of the obese phenotype of heart failure with preserved ejection fraction: a RELAX trial ancillary study. Mayo Clin Proc 2019;94(7):1199-209. https://doi.org/10.1016/j.mayocp.2018.11.037
  29. Kosiborod MN, Abildstrom SZ, Borlaug BA, et al. Semaglutide in patients with heart failure with preserved ejection fraction and obesity. N Engl J Med 2023;389(12):1069-84. https://doi.org/10.1056/NEJMoa2306963
  30. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med 2021; 384(11):989. https://doi.org/10.1056/NEJMoa2032183
  31. Stogios N, Fezza G, Wong JV, Ross HJ, Farkouh ME, Nolan RP. Current challenges for using the Kansas City Cardiomyopathy Questionnaire to obtain a standardized patient-reported health status outcome. Eur J Heart Fail 2021;23(2):205-07. https://doi.org/10.1002/ejhf.2139
  32. Kosiborod MN, Abildstrøm SZ, Borlaug BA, et al. Design and baseline characteristics of STEP-HFpEF program evaluating semaglutide in patients with obesity HFpEF phenotype. JACC Heart Fail 2023; 11(8 Pt 1):1000-10. https://doi.org/10.1016/j.jchf.2023.05.010
  33. Nikolaus Marx MF, Katharina Schütt, Dirk Müller-Wieland, et al and ESC scientific document group. 2023 ESC guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J 2023;44:4043-4140. https://doi.org/10.1093/eurheartj/ehad192
  34. Miller RJH, Howlett JG, Fine NM. A novel approach to medical management of heart failure with reduced ejection fraction. Can J Cardiol 2021;37(4):632-43. https://doi.org/10.1016/j.cjca.2020.12.028
  35. McMurray JJV, Packer M. How should we sequence the treatments for heart failure and a reduced ejection fraction?: a redefinition of evidence-based medicine. Circulation 2021;143(9):875-7. https://doi.org/10.1161/CIRCULATIONAHA.120.052926
  36. Velazquez EJ, Morrow DA, Devore AD, et al. Angiotensin–neprilysin inhibition in acute decompensated heart failure. N Engl J Med 2019; 380(6):539-48. https://doi.org/10.1056/NEJMoa1812851
  37. Fonarow GC, Yancy CW, Hernandez AF, Peterson ED, Spertus JA, Heidenreich PA. Potential impact of optimal implementation of evidence-based heart failure therapies on mortality. Am Heart J 2011; 161(6):1024-30.e3. https://doi.org/10.1016/j.ahj.2011.01.027
  38. Mebazaa A, Davison B, Chioncel O, et al. Safety, tolerability and efficacy of up-titration of guideline-directed medical therapies for acute heart failure (STRONG-HF): a multinational, open-label, randomised, trial. Lancet 2022;400(10367):1938-52. https://doi.org/10.1016/S0140-6736(22)02076-1
  39. Zaman S, Zaman SS, Scholtes T, et al. The mortality risk of deferring optimal medical therapy in heart failure: a systematic comparison against norms for surgical consent and patient information leaflets. Eur J Heart Fail 2017;19(11):1401-9. https://doi.org/10.1002/ejhf.838
  40. Greene SJ, Butler J, Albert NM, et al. Medical therapy for heart failure with reduced ejection fraction: the CHAMP-HF registry. J Am Coll Cardiol 2018;72(4):351-66. https://doi.org/10.1016/j.jacc.2018.04.070
  41. Savarese G, Kishi T, Vardeny O, et al. Heart failure drug treatment-- inertia, titration, and discontinuation: a multinational observational study (EVOLUTION HF). JACC Heart Fail 2023;11(1):1-14.https://doi.org/10.1016/j.jchf.2022.08.009
  42. Verhestraeten C, Heggermont WA, Maris M. Clinical inertia in the treatment of heart failure: a major issue to tackle. Heart Failure Reviews 2021;26(6):1359-70. https://doi.org/10.1007/s10741-020-09979-z
  43. Patel P, Gupta P, Burns A, et al. Biochemical urine testing of adherence to cardiovascular medications reveals high rates of nonadherence in people attending their annual review for type 2 diabetes. Diabetes Care 2019;42(6):1132-5. https://doi.org/10.2337/dc18-1453
  44. Sze S, Squire I, Gupta P, et al. 128 Medication non-adherence assessed using liquid chromatography tandem mass spectrometry in patients with chronic heart failure. Heart 2023;109:A145-A6. https://doi.org/10.1136/heartjnl-2023-BCS.128
  45. Gupta P, Voors AA, Patel P, et al. Non‐adherence to heart failure medications predicts clinical outcomes: assessment in a single spot urine sample by liquid chromatography‐tandem mass spectrometry (results of a prospective multicentre study). Eur J Heart Fail 2021; 23(7):1182-90. https://doi.org/10.1002/ejhf.2160
  46. Powers M. Every person with diabetes needs ongoing self-management education and support. National Institue of Diabetes and Digestive and Kidney Diseases, 2019. [Available from: https://www.niddk.nih.gov/health-information/professionals/diabetes- discoveries-practice/diabetes-self-management-education-support]
  47. Ruppar TM, Cooper PS, Mehr DR, Delgado JM, Dunbar‐Jacob JM. Medication adherence interventions improve heart failure mortality and readmission rates: systematic review and meta‐analysis of controlled trials. J Am Heart Assoc 2016;5(6):e002606. https://doi.org/10.1161/JAHA.115.002606
  48. Unverzagt S, Meyer G, Mittmann S, Samos F-A, Unverzagt M, Prondzinsky R. Improving treatment adherence in heart failure. Deutsches Ärzteblatt Int 2016;113(25):423-30. https://doi.org/10.3238/arztebl.2016.0423
  49. Greene SJ, Fonarow GC. Clinical inertia and medical therapy for heart failure: the unintended harms of ‘first, do no harm’. Eur Journal Heart Fail 2021;23(8):1343-5. https://doi.org/10.1002/ejhf.2283
  50. Bauersachs J. Heart failure drug treatment: the fantastic four. Eur Heart J 2021;42(6):681-3. https://doi.org/10.1093/eurheartj/ehaa1012
  51. Chen X, Wu M. Heart failure with recovered ejection fraction: current understanding and future prospects. Am J Med Sci 2023;365(1):1-8. https://doi.org/10.j.amjms.2022.07.018
  52. Fonarow GC, Greene SJ. Rapid and intensive guideline-directed medical therapy for heart failure. J Am Coll Cardiol 2023;81(22):2145-8.https://doi.org/10.1016/j.jacc.2023.04.006