Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Protocol of BEYOND trial: Clinical BEnefit of sodium-glucose cotransporter-2 (SGLT-2) inhibitors in rhYthm cONtrol of atrial fibrillation in patients with diabetes mellitus

  • Kyuhyun Lee ,

    Contributed equally to this work with: Kyuhyun Lee, Soo Kyoung Lee, Juyeon Lee, Bo Kyung Jeon

    Roles Writing – original draft

    Affiliation College of Medicine, Ewha Womans University, Seoul, Republic of Korea

  • Soo Kyoung Lee ,

    Contributed equally to this work with: Kyuhyun Lee, Soo Kyoung Lee, Juyeon Lee, Bo Kyung Jeon

    Roles Writing – original draft

    Affiliation College of Medicine, Ewha Womans University, Seoul, Republic of Korea

  • Juyeon Lee ,

    Contributed equally to this work with: Kyuhyun Lee, Soo Kyoung Lee, Juyeon Lee, Bo Kyung Jeon

    Roles Writing – original draft

    Affiliation College of Medicine, Ewha Womans University, Seoul, Republic of Korea

  • Bo Kyung Jeon ,

    Contributed equally to this work with: Kyuhyun Lee, Soo Kyoung Lee, Juyeon Lee, Bo Kyung Jeon

    Roles Project administration, Writing – review & editing

    Affiliation Department of Cardiology, Ewha Womans University Medical Center, Seoul, Republic of Korea

  • Tae-Hoon Kim,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Yonsei University Health System, Seoul, Republic of Korea

  • Hee Tae Yu,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Yonsei University Health System, Seoul, Republic of Korea

  • Jung Myung Lee,

    Roles Conceptualization, Investigation, Methodology, Visualization

    Affiliation Kyung Hee University, Medical Center, Seoul, Republic of Korea

  • Jin-Kyu Park,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Department of Cardiology, Hanyang University Seoul Hospital, Seoul, Republic of Korea

  • Yong-Soo Baek,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Inha University College of Medicine and Inha University Hospital, Incheon, Republic of Korea

  • Dong Hyeok Kim,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Department of Cardiology, Ewha Womans University Medical Center, Seoul, Republic of Korea

  • Jaemin Shim,

    Roles Conceptualization, Methodology, Project administration, Validation

    Affiliation Korea University College of Medicine, Anam Hospital, Seoul, Republic of Korea

  • Boyoung Joung,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Yonsei University Health System, Seoul, Republic of Korea

  • Moon-Hyoung Lee,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Yonsei University Health System, Seoul, Republic of Korea

  • Hui-Nam Pak,

    Roles Conceptualization, Investigation, Methodology, Validation

    Affiliation Yonsei University Health System, Seoul, Republic of Korea

  • Junbeom Park

    Roles Conceptualization, Funding acquisition, Project administration, Supervision, Writing – review & editing

    parkjb@ewha.ac.kr

    Affiliation Department of Cardiology, Ewha Womans University Medical Center, Seoul, Republic of Korea

Abstract

SGLT-2 inhibitor, traditionally used for glycemic control, has several beneficial effects that can help manage heart failure (HF). SGLT-2 inhibitors reduce the risk of cardiovascular mortality in patients with HF. As atrial fibrillation (AF) is closely associated with HF and diabetes mellitus (DM) is a risk factor for AF, we assume that SGLT-2 inhibitors will also show therapeutic benefits regarding AF, especially for rhythm control. This trial has a multicenter, prospective, open, blinded endpoint design. It is a 1:1 randomized and controlled study. A total of 716 patients who are newly diagnosed of AF and DM within 1 year will be enrolled from 7 tertiary medical centers. The trial is designed to compare the effects of SGLT-2 inhibitors and other oral hypoglycemic agents on atrial rhythm control in patients with AF and DM. The primary outcome is the recurrence of AF within a year (including post-antiarrhythmic drugs (AAD) or ablation). The secondary outcomes are the ablation rate within a year, change in AF burden, size of the left atrium, NT-proBNP, the AF symptom score, and the quality of life. This trial will prospectively evaluate the effect and safety of SGLT-2 inhibitors on AF rhythm control in patients with DM. It will provide an invaluable dataset on rhythm control in AF with DM for future studies and offer novel information to assist in clinical decisions. (BEYOND trial, ClinicalTrials.gov number: NCT05029115. https://clinicaltrials.gov/ct2/show/NCT05029115).

Introduction

Atrial fibrillation (AF) is a common cause of cardiac arrhythmia, and its prevalence increases with age [1,2]. In the United States alone, the prevalence of AF is expected to increase to 12.1 million in 2030 from 5.2 million in 2010 [3]. In addition, owing to the growth in the global prevalence of diabetes mellitus (DM) [4], it has become necessary to manage both AF and DM simultaneously. The Action in Diabetes and Vascular Disease: preterAx and diamicroN-MR Controlled Evaluation (ADVANCE) study showed that AF is relatively common in DM and is associated with a considerably increased risk of cardiovascular events and death in patients with DM [5]. According to the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation (ORBIT-AF) registry, DM is not only a risk factor for AF, but also associated with worse AF symptoms and a lower quality of life [6].

AF and congestive heart failure (CHF) are commonly encountered disease entities that share common risk factors such as hypertension, DM, ischemic heart disease, and valvular heart disease [7,8]. AF and CHF are so intertwined that many questions related to their pathophysiologic relationship are still unknown [9]. It has been suggested that heart failure (HF) increases the risk of AF by elevating atrial filling pressures [10,11], inducing alterations in intracellular calcium [12,13], and disturbing the neuro-endocrinological balance [7]. The Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure (DAPA-HF) trial and The Empagliflozin Outcome Trial in Patients with Chronic Heart Failure and Reduced Ejection Fraction (EMPEROR-Reduced) trial revealed that two sodium–glucose cotransporter 2 (SGLT-2) inhibitors, dapagliflozin and empagliflozin, reduce the risk of cardiovascular death or HF exacerbation, regardless of the presence or absence of DM [14,15]. Besides glucose-lowering, SGLT-2 inhibitors appear to have several other beneficial effects as well.

Recent meta-analyses and retrospective studies have reported that the use of SGLT-2 inhibitors in type 2 DM patients with or without HF, as well as in type 2 DM patients with HF, reduces the incidence rate of AF, atrial flutter, and other cardiovascular events [1619]. However, few randomized controlled studies have reported the relationship between SGLT-2 and AF, and therefore, reports of detailed prognostic characteristics such as change of AF burden and rhythm control other than the incidence of AF are rare. This trial aimed to determine whether SGLT-2 inhibitors have an advantageous influence on rhythm control of AF and, hence, reduce the adverse outcomes of AF through prospective, randomized, and controlled methods.

Materials and methods

Study hypothesis and primary outcome

The primary hypothesis of this study is that the SGLT-2 inhibitor is superior to other oral hypoglycemic medications for rhythm control in patients with AF and DM, diagnosed within the prior year. A step-up treatment will be carried out when the participant’s AF recurs. The recurrence rate of AF will be investigated after performing stepwise rhythm control therapies including anti-arrhythmic drugs (AAD) and ablation. AF is defined as per the 2019 AHA/ACC/HRS Guideline, i.e., irregular R–R intervals (when atrioventricular conduction is present), absence of distinct repeating P waves, and irregular atrial activity [20]. It is checked by 24-h Holter ECG every 3 months and is considered significant when it lasts for > 30 s [21].

Secondary outcomes

Secondary outcomes are listed in Table 1. AF-free survival will be compared between the two groups as the first of the secondary outcome, analyzed using the Kaplan–Meier method, and documented on curve. AF burden at 3-month follow-up visit, 12-month follow-up visit in those who did not step up and used AAD only for rhythm control, immediately before trying ablation, and at 12-month follow-up visit in those who stepped up and underwent ablation in both the SGLT-2 inhibitor group and the control group will be compared. AF burden is defined as the atrial fibrillation time percentage documented on 24-h Holter ECG [22]. Ablation rate, which is the percentage of patients undergoing ablation, within a year will also be compared between the two groups to identify the efficacy of symptom and rhythm control using SGLT-2 inhibitors. Sinus rhythm is considered stable when either a standard ECG or a 24-h Holter ECG showed no episode of clinically relevant arrhythmia, including AF, at the time of check-up [23]. The proportion of participants with stable sinus rhythm in all participants, regardless of with or without ablation, is compared between the two groups at baseline, 6-, and 12-month follow-up visits.

As dilatation of the left atrium (LA) is associated with AF in both pathophysiologic and prognostic perspectives, we will use the LA size as a secondary outcome. Left atrial remodeling induces electrical modification, which is associated with AF. LA structural remodeling is characterized by atrial dilatation [24]. An increase in the LA size at baseline is associated with progressive LA enlargement and AF recurrence. Hence, the LA size can be considered a prognostic factor for spontaneous conversion of AF [25]. As an indicator of LA dilatation, the left atrial diameter (anterior–posterior) will be measured by transthoracic echocardiography (TTE) at baseline and 12-month follow-up visit [26]. N-terminal pro-B-type natriuretic peptide (NT-proBNP) is often elevated in AF and is higher in severe AF [27,28]. As it is a significant secondary outcome, we will measure the level of NT-proBNP in blood and compare their values in the two groups at baseline and 12-month follow-up visit.

The symptom score is compared by using the EHRA score. It is measured at baseline, 3-, 6-, 9-, and 12-month of follow-up visits. The ratio of asymptomatic participants will be compared between the two groups. To compare the quality of life between the two groups, EuroQoL five-dimensional instrument (EQ-5D, www.euroqol.org) and SF-12 (a shortened version of SF-36, www.sf-36.org) Health Survey will be used, and assessments will be conducted between the data on baseline and 12-month follow-up visit.

Adverse events

Adverse events are assessed and judged twice a year by an independent committee. More than half of the votes should be matched to finalize the decision. Adverse events are listed in Table 2. Any unexpected medical events or laboratory findings are also defined as adverse events. Atrial arrhythmia alone is not classified as an adverse event. Instead, it is considered as the primary and/or secondary outcome.

Study design and participants

This is a multicenter study with a prospective, open, blinded endpoint design (Fig 1). It is a 1:1 randomized and controlled study that compares the rhythm control effect of SGLT-2 inhibitor therapy to other oral hypoglycemic agents in patients with AF and DM (Fig 2).

thumbnail
Fig 1. Completed SPIRIT schedule of trial.

The rough schedule of BEYOND trial is summarized in a table. If AF is recurred during the follow-up period, patient will get a catheter or CRYO-balloon ablation and have an additional one-year follow-up period after ablation.

https://doi.org/10.1371/journal.pone.0280359.g001

thumbnail
Fig 2. Flow chart of the trial.

Brief flow of the BEYOND trial described as a schematic diagram. Patients who enroll this trial will be randomly administrated SGLT-2 inhibitor or other DM medication while applying stepwise rhythm control strategy for AF.

https://doi.org/10.1371/journal.pone.0280359.g002

Patients diagnosed with AF within a year, who have never undergone any ablation treatment, newly diagnosed with DM, and with ages between 18 and 80 years will be selected as the study participants. They will be enrolled in 7 tertiary medical centers from the Division of Cardiology and the Division of Endocrinology. As patients diagnosed with AF also tend to check for DM to calculate the CHAD2-VASc score, those who have been diagnosed as AF in the prior year and newly diagnosed with DM in the follow-up can be recruited for the trial in the Division of Cardiology. To identify patients for enrollment in the Division of Endocrinology, every patient newly diagnosed with DM during the enrollment period will be asked for 12-lead ECG and 24-h Holter ECG; those with AF > 10 s in 12-lead ECG or >30 s in 24-h Holter ECG can be recruited for the trial. All participants must meet the required inclusion (Table 3) and exclusion criteria (Table 4) at the point of randomization. The participants should otherwise be healthy and use only oral hypoglycemic agents, not subcutaneous agents, for glycemic control.

The rhythm control strategy for AF will be followed, independent of the type of hypoglycemic agents received. This strategy includes two sessions (Fig 2). In session 1, an AAD will be assigned to every participant and taken for at least 3 months. If patients have persistent AF, cardioversion will be performed with AAD. During the follow-up, those with recurrent AF should step up to session 2, which involves treatment with radiofrequency ablation (RFA) or CRYO-balloon ablation (4 pulmonary vein isolation; 4PVI). For participants of session 2, the follow-up will begin with the moment RFA or cryotherapy is applied, which will be regarded as 0-month; follow-up will be conducted every 3 months for the next 12 months. The rhythm control strategy will be conducted as per the clinical guidelines of 2020 ESC [30] and 2019 AHA/ACC/HRS guidelines [20] for AF.

Treatment: DM control (allocation)

The subjects will receive oral hypoglycemic agents for glucose control; the type of agents will be determined by randomization. Patients using SGLT-2 inhibitors to control their serum glucose will be specified as the case group, while those using other hypoglycemic agents (e.g., metformin, meglitinides, sulfonylureas, DPP-4 inhibitors, GLP-1 receptor agonists, α-glucosidase inhibitors, and thiazolidinediones) to control their serum glucose will be classified as the control group. The number, type, and dosage of the agents will be individualized at the discretion of the physician in accordance with the 2021 ADA guidelines [31], considering factors such as the kidney function, age, and cardiovascular status.

Follow-up and detection of AF recurrence

After their recruitment, the participants will receive baseline and evaluation studies under scrutiny, which include history taking, physical examination, blood sampling, urine sampling, 12-lead ECG, 24-h Holter ECG, TTE, EHRA, EQ-5D, and SF-12. The follow-up schedule will begin with the prescription of oral hypoglycemic agents (Table 5). During each intervention, the following steps were conducted: (1) history taking for the review of AF and DM status, adverse events, and other specific illnesses on every visit; (2) physical examination to evaluate any DM complication, AF, NYHA score, neurologic exam, systolic and diastolic blood pressure; (3) blood sampling for CBC, BUN/Cr, electrolyte, coagulation profile, HbA1c, fasting glucose, AST/ALT, lipid battery, and NT-proBNP; (4) urine sampling for dip stick urinalysis with microscopy and measuring proteinuria (especially for albuminuria); and (5) TTE to measure the LA size and left ventricular ejection fraction (LVEF). Any other additive assessing procedure may be conducted and recorded during the clinical course of the patients.

During patient monitoring, if AF recurs and the patient complains of its symptoms, RFA or cryoablation (4PVI) will be performed at the discretion of the physician in accordance with the 2020 ESC [30] 2019 AHA/ACC/HRS guidelines [20] for AF. For patients with ablation, 24-h Holter ECG monitoring every 3 months for an additional year will be performed from that point on. If severe adverse events occur, the patient will be excluded from the study after a committee meeting. The main concern of the review is the recurrence of AF, which is the primary outcome of this trial.

Statistical plan, sample size, and power determination

In this study, 716 patients will be enrolled from 7 tertiary medical centers. The sample size was determined based on the primary outcome, i.e., the SGLT-2 inhibitors should show 40% reduction in AF after a year of treatment [32] and 50% of AAD users should step up to the ablation therapy [33,34]. Assuming 10% follow-up loss, the sample size of 716 patients (358 in each group) is expected to achieve 80% power and an alpha-level of 5% to detect the difference. The study is powered to demonstrate the superior additive effect of SGLT-2 inhibitors over other hypoglycemic agents using the Chi square method. AF recurrence rate and ratio of sinus rhythm on 24-h holter EKG will be analyzed using Chi square test, and left atrial size, NT-pro BNP and quality of life (AFEQT) score on 12-month final follow-up will be analyzed by student t-test. Disease-free survival and overall survival during the follow-up period will be calculated using Kaplan-Meier method. Mono-variable and multi-variable cox regression analysis will be used to calculate hazard ratio of SGLT-2 group.

To reduce the selection bias, all subjects will be randomized to allocate the hypoglycemic agents according to random number table. Missing data is planned to be controlled using multiple imputation.

Study status and organization

Ewha Womans University Mokdong Hospital is responsible for this trial, and the Institutional Review Board (IRB) of Ewha Womans University Mokdong Hospital approved this research plan, including the written consent form (S2, S3 and S4 File). In addition to Ewha Womans University Mokdong Hospital, at least seven other tertiary hospitals in South Korea are now preparing for IRB approval of each hospital with the same research plan.

This study complied with the fundamental spirit of the clinical trial management standards of the Helsinki Declaration (revised 2013) and the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use–Guidelines for Good Clinical Practice (ICH-GCP). The study was initiated after approval of the IRB in each medical center, and after approval, centers recruited patients and the therapies were administered following the appropriate study protocol and clinical standards.

Patients who met the inclusion criteria and voluntarily agreed to enroll in the study provided written informed consent, which was reviewed and approved by the IRB of each participating hospital after sufficient explanation about the research process. Personal identifiers were kept confidential by the researcher, and the research data was recorded with initials and research subject identification information was coded.

All adverse events reported by the subjects during a 3-month follow-up were reported at meetings between researchers four times a year and evaluated by an independent committee. Interruption of participation was decided when the above adverse reaction was a considerable side effect of the SGLT-2 inhibitors (dapagliflozin and empagliflozin), as informed by the Ministry of Food and Drug Safety, or was a serious adverse event unrelated to causality.

Discussion

Recently, SGLT-2 inhibitors have been shown to exhibit beneficial effects beyond glucose control. SGLT-2 inhibitors were originally developed as hypoglycemic agents. They target SGLT-2 protein, which is responsible for 90% reabsorption of filtered glucose in the proximal convoluted tubules of the kidneys [35], resulting in diuresis. Previous studies have shown that SGLT-2 inhibitors suppress sympathetic overactivity [36] and oxidative stress [37]. DAPA-HF and EMPEROR-Reduced trial demonstrated that SGLT-2 inhibitors can reduce cardiovascular mortality in patients with HF [14,15]. Considering that AF and CHF are mutual risk factors, we designed a clinical trial based on the rationale that SGLT-2 inhibitors might be able to improve the clinical outcome of AF with DM by establishing the rhythm control of AF.

We will compare the recurrence rate of AF of the two groups after performing AAD administration and hypoglycemic treatment for at least 3 months as a primary outcome and evaluating the degree of rhythm control of AF. Because enlisting patients who were previously diagnosed with DM and whose glucose levels are currently well controlled in the trial can give rise to grave ethical issues, we recruited patients newly diagnosed with AF or DM within 1 year. We decided not to choose any specific SGLT-2 inhibitor agents and their dosage right away; instead, we decided to recommend physicians to utilize the maximum tolerable doses.

Several systemic reviews and meta-analyses have described advantages of catheter ablation over AAD in maintaining sinus rhythm [3842] and improving the quality of life [43]. The CABANA trial showed that catheter ablation was superior in reducing the recurrence rate of AF compared to the AAD-only therapy [44]. Assuming that catheter ablation will improve the clinical outcome and lower the AF recurrence rate compared to that achieved with the AAD-only treatment in patients with AF and DM, we will add the following procedures after the random assignment of SGLT-2 inhibitors to minimize the bias caused by study protocols: First, the patient takes the assigned medicine for at least 3 months. Second, if AF recurrence is detected and the patient manifests clinical symptoms at follow-up visits, the physician will make a clinical decision on catheter ablation. AF recurrence after ablation (RFA and 4PVI) within a year will be measured by using the established protocol as the primary outcome among patients who have underwent ablation. In this setting, however, comparing the AF burden between the case (SGLT-2 inhibitor) and control (non-SGLT-2 inhibitor) groups at 12 months of trial would disturb the study results. Thus, the recurrence of AF is set as the primary outcome in each step (AAD and ablation).

Previous studies, including DAPA-HF, EMPORER-Reduced, and DECLARE-TIMI 58, have focused on the efficacy of SGLT-2 inhibitors in reducing the risk of adverse cardiovascular events as the primary endpoint in patients with DM, HF, or AF. However, the effects of SGLT-2 inhibitors on arrhythmia, especially AF, were only described by subgroup post-hoc analyses or meta-analyses, so they usually focused on the AF incidence rate and could not analyze other specific properties of AF [16,32,45]. Thus, our trial was advantageous because it allowed the measurement of AF control prospectively in patients with AF and DM.

Very few studies have quantitatively analyzed the AF burden, represented as the percentage of AF time during the monitoring period, because continuous ambulatory monitoring is needed to calculate the exact AF burden [22]. There are some ongoing clinical trials such as DAPA-AF that attempt to quantitatively evaluate the AF burden using cardiac implantable electronic devices (CIED). On the contrary, we plan to measure the AF burden after randomization of SGLT-2 inhibitors, before and after ablation, by utilizing 24-h Holter ECG, which is an excellent choice in terms of accessibility. Furthermore, our study results and information about the population with AF and DM will provide valuable data for subsequent studies in the future.

The LA size and blood level of NT-proBNP are set as the secondary endpoints, as we speculate that SGLT-2 inhibitors will delay the pathophysiologic course of AF. Remodeling of LA by itself acts as an underlying substrate for AF. In addition, progression of AF further deteriorates the LA function and accelerates LA remodeling, thereby continuing the vicious cycle [46,47]. Degree of abnormality of the LA structure and function can be used to determine the positive association with the electrical burden of AF [47]. Therefore, we plan to evaluate the progress of LA remodeling by measuring the degree of LA enlargement, i.e., change in the LA size by utilizing TTE. In addition, considering that NT-proBNP is often increased in AF and shows positive correlation with the clinical severity of AF [27,28], NT-proBNP can be used for assessing the severity of AF. Finally, the quality of life of all patients will be precisely measured.

The trial has some limitations as well. First, it primarily focuses on the effect of SGLT-2 inhibitors in patients newly diagnosed with AF and DM within 1 year. As a result, long-term effects of SGLT-2 inhibitors cannot be analyzed in this trial. Nonetheless, the short-term effects of SGLT-2 inhibitors are meaningful because these inhibitors reduced the incidence of atrial arrhythmia within 1 year compared to that in the control group [32] and the diuretic effect of SGLT-2 inhibitors appeared to be most prominent in first three months of administration only [48]. Second, a certain SGLT-2 inhibitor product is not unified in this trial. Physicians can choose the specific product and the maximum tolerable dose based on their clinical decisions and settings. To control the bias, a subgroup analysis including stratification on products will be conducted as post-hoc analysis.

Conclusions

Data on the effect of SGLT-2 inhibitors derived from the trials that solely target patients with AF and DM have limited applicability. The proposed trial will evaluate the efficacy and safety of SGLT-2 inhibitors on the AF rhythm control in patients with AF and DM. The results generated by this trial will provide an invaluable dataset on rhythm control in AF with DM for future studies and offer novel information to assist in clinical decisions.

Supporting information

S2 File. Study protocol which was approved by institutional review board of Ewha Womans University Mokdong Hospital (original Korean version).

https://doi.org/10.1371/journal.pone.0280359.s002

(PDF)

S3 File. Study protocol which was approved by institutional review board of Ewha Womans University Mokdong Hospital (English-transcribed version).

https://doi.org/10.1371/journal.pone.0280359.s003

(DOCX)

S4 File. Approval latter from institutional review board of Ewha Womans University Mokdong Hospital (original Korean version).

https://doi.org/10.1371/journal.pone.0280359.s004

(PDF)

S5 File. Approval latter from institutional review board of Ewha Womans University Mokdong Hospital (English-transcribed version).

https://doi.org/10.1371/journal.pone.0280359.s005

(PDF)

References

  1. 1. Benjamin EJ WP, D’Agostino RBSilbershatz H, Kannel WB, Levy D. Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation. 1998;98(10):946–52. pmid:9737513
  2. 2. January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC Jr., et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014;130(23):e199–267. Epub 2014/04/01. pmid:24682347; PubMed Central PMCID: PMC4676081.
  3. 3. Colilla S, Crow A, Petkun W, Singer DE, Simon T, Liu X. Estimates of current and future incidence and prevalence of atrial fibrillation in the U.S. adult population. Am J Cardiol. 2013;112(8):1142–7. Epub 2013/07/09. pmid:23831166.
  4. 4. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Research and Clinical Practice. 2019;157:107843. pmid:31518657
  5. 5. Du X, Ninomiya T, De Galan B, Abadir E, Chalmers J, Pillai A, et al. Risks of cardiovascular events and effects of routine blood pressure lowering among patients with type 2 diabetes and atrial fibrillation: results of the ADVANCE study. European heart journal. 2009;30(9):1128–35. pmid:19282274
  6. 6. Echouffo-Tcheugui JB, Shrader P, Thomas L, Gersh BJ, Kowey PR, Mahaffey KW, et al. Care patterns and outcomes in atrial fibrillation patients with and without diabetes: ORBIT-AF registry. Journal of the American College of Cardiology. 2017;70(11):1325–35. pmid:28882229
  7. 7. Kareti KR, Chiong JR, Hsu SS, Miller AB. Congestive heart failure and atrial fibrillation: rhythm versus rate control. J Card Fail. 2005;11(3):164–72. Epub 2005/04/07. pmid:15812742.
  8. 8. Heist EK, Ruskin JN. Atrial fibrillation and congestive heart failure: risk factors, mechanisms, and treatment. Prog Cardiovasc Dis. 2006;48(4):256–69. Epub 2006/03/07. pmid:16517247.
  9. 9. Anter E, Jessup M, Callans DJ. Atrial fibrillation and heart failure: treatment considerations for a dual epidemic. Circulation. 2009;119(18):2516–25. Epub 2009/05/13. pmid:19433768.
  10. 10. SOLTI F, VECSEY T, KÉKESI V, JUHÁSZ-NAGY A. The effect of atrial dilatation on the genesis of atrial arrhythmias. Cardiovascular Research. 1989;23(10):882–6. pmid:2620315
  11. 11. Frank Bode AK, Raymond L. Woosley, and Michael R. Franz. Gadolinium Decreases Stretch-Induced Vulnerability to Atrial Fibrillation. Circulation. 2000;101(18):2200–5. pmid:10801762
  12. 12. Dirk J. Beuckelmann MMN, MD; and Erland Erdmann, MD. Intracellular Calcium Handling in Isolated Ventricular Myocytes From Patients With Terminal Heart Failure. Circulation. 1992;85(3):1046–55. pmid:1311223
  13. 13. Ohkusa T, Ueyama T, Yamada J, Yano M, Fujumura Y, Esato K, et al. Alterations in cardiac sarcoplasmic reticulum Ca2+regulatory proteins in the atrial tissue of patients with chronic atrial fibrillation. Journal of the American College of Cardiology. 1999;34(1):255–63. pmid:10400019
  14. 14. McMurray JJV, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med. 2019;381(21):1995–2008. Epub 2019/09/20. pmid:31535829.
  15. 15. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413–24. Epub 2020/09/01. pmid:32865377.
  16. 16. Wang M, Zhang Y, Wang Z, Liu D, Mao S, Liang B. The effectiveness of SGLT2 inhibitor in the incidence of atrial fibrillation/atrial flutter in patients with type 2 diabetes mellitus/heart failure: a systematic review and meta-analysis. Journal of Thoracic Disease. 2022;14(5):1620. pmid:35693625
  17. 17. Li W-j, Chen X-q, Xu L-l, Li Y-q, Luo B-h. SGLT2 inhibitors and atrial fibrillation in type 2 diabetes: a systematic review with meta-analysis of 16 randomized controlled trials. Cardiovascular diabetology. 2020;19(1):1–14.
  18. 18. Bonora BM, Raschi E, Avogaro A, Fadini GP. SGLT-2 inhibitors and atrial fibrillation in the Food and Drug Administration adverse event reporting system. Cardiovascular diabetology. 2021;20(1):1–8.
  19. 19. Okunrintemi V, Mishriky BM, Powell JR, Cummings DM. Sodium‐glucose co‐transporter‐2 inhibitors and atrial fibrillation in the cardiovascular and renal outcome trials. Diabetes, Obesity and Metabolism. 2021;23(1):276–80. pmid:33001548
  20. 20. January CT, Wann LS, Calkins H, Chen LY, Cigarroa JE, Cleveland JC, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration With the Society of Thoracic Surgeons. Circulation. 2019;140(2):e125–e51. pmid:30686041
  21. 21. Steinberg JS, O’Connell H, Li S, Ziegler PD. Thirty-Second Gold Standard Definition of Atrial Fibrillation and Its Relationship With Subsequent Arrhythmia Patterns: Analysis of a Large Prospective Device Database. Circ Arrhythm Electrophysiol. 2018;11(7):e006274. Epub 2018/07/14. pmid:30002065.
  22. 22. Chen LY, Chung MK, Allen LA, Ezekowitz M, Furie KL, McCabe P, et al. Atrial Fibrillation Burden: Moving Beyond Atrial Fibrillation as a Binary Entity: A Scientific Statement From the American Heart Association. Circulation. 2018;137(20). pmid:29661944
  23. 23. Plewan A, Lehmann G, Ndrepepa G, Schreieck J, Alt EU, Schomig A, et al. Maintenance of sinus rhythm after electrical cardioversion of persistent atrial fibrillation; sotalol vs bisoprolol. Eur Heart J. 2001;22(16):1504–10. Epub 2001/08/03. pmid:11482924.
  24. 24. Casaclang-Verzosa G, Gersh BJ, Tsang TS. Structural and functional remodeling of the left atrium: clinical and therapeutic implications for atrial fibrillation. J Am Coll Cardiol. 2008;51(1):1–11. Epub 2008/01/05. pmid:18174029.
  25. 25. Valerio Zacà MD MGM, Sergio Mondillo MD, Marta Focardi MD, Piercarlo Ballo MD, Francesco Guerrini MD. Left atrial enlargement as a predictor of recurrences in lone paroxysmal atrial fibrillation. Can J Cardiol. 2007;23(11):869–72. PubMed Central PMCID: PMC2651363. pmid:17876377
  26. 26. Badano LP, Miglioranza MH, Mihăilă S, Peluso D, Xhaxho J, Marra MP, et al. Left Atrial Volumes and Function by Three-Dimensional Echocardiography. Circulation: Cardiovascular Imaging. 2016;9(7):e004229. pmid:27412658
  27. 27. Buttner P, Schumacher K, Dinov B, Zeynalova S, Sommer P, Bollmann A, et al. Role of NT-proANP and NT-proBNP in patients with atrial fibrillation: Association with atrial fibrillation progression phenotypes. Heart Rhythm. 2018;15(8):1132–7. Epub 2018/04/01. pmid:29604419.
  28. 28. Hijazi Z, Wallentin L, Siegbahn A, Andersson U, Christersson C, Ezekowitz J, et al. N-terminal pro-B-type natriuretic peptide for risk assessment in patients with atrial fibrillation: insights from the ARISTOTLE Trial (Apixaban for the Prevention of Stroke in Subjects With Atrial Fibrillation). J Am Coll Cardiol. 2013;61(22):2274–84. Epub 2013/04/09. pmid:23563134.
  29. 29. Cheung AK, Chang TI, Cushman WC, Furth SL, Hou FF, Ix JH, et al. KDIGO 2021 Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney International. 2021;99(3):S1–S87. pmid:33637192
  30. 30. Hindricks G, Potpara T, Dagres N, Arbelo E, Bax JJ, Blomström-Lundqvist C, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. European Heart Journal. 2020;42(5):373–498. pmid:32860505
  31. 31. Association AD. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2021. Diabetes Care. 2021;44(Supplement 1):S15–S33. pmid:33298413
  32. 32. Fernandes GC, Fernandes A, Cardoso R, Penalver J, Knijnik L, Mitrani RD, et al. Association of SGLT2 inhibitors with arrhythmias and sudden cardiac death in patients with type 2 diabetes or heart failure: A meta-analysis of 34 randomized controlled trials. Heart Rhythm. 2021. Epub 2021/03/25. pmid:33757845.
  33. 33. Andrade JG, Deyell MW, Verma A, Macle L, Champagne J, Leong-Sit P, et al. Association of Atrial Fibrillation Episode Duration With Arrhythmia Recurrence Following Ablation: A Secondary Analysis of a Randomized Clinical Trial. JAMA Network Open. 2020;3(7):e208748-e. pmid:32614422
  34. 34. Andrade JG, Wells GA, Deyell MW, Bennett M, Essebag V, Champagne J, et al. Cryoablation or Drug Therapy for Initial Treatment of Atrial Fibrillation. N Engl J Med. 2021;384(4):305–15. Epub 2020/11/17. pmid:33197159.
  35. 35. Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Current Opinion in Endocrinology & Diabetes and Obesity. 2016:1. pmid:27898586
  36. 36. Sano M. A new class of drugs for heart failure: SGLT2 inhibitors reduce sympathetic overactivity. Journal of Cardiology. 2018;71(5):471–6. pmid:29415819
  37. 37. Li C, Zhang J, Xue M, Li X, Han F, Liu X, et al. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovascular Diabetology. 2019;18(1). pmid:30710997
  38. 38. Al-Khatib SM, Allen LaPointe NM, Chatterjee R, Crowley MJ, Dupre ME, Kong DF, et al. Rate-and rhythm-control therapies in patients with atrial fibrillation: a systematic review. Annals of internal medicine. 2014;160(11):760–73. pmid:24887617
  39. 39. Bonanno C, Paccanaro M, La Vecchia L, Ometto R, Fontanelli A. Efficacy and safety of catheter ablation versus antiarrhythmic drugs for atrial fibrillation: a meta-analysis of randomized trials. Journal of Cardiovascular Medicine. 2010;11(6):408–18. pmid:19834326
  40. 40. Calkins H, Reynolds MR, Spector P, Sondhi M, Xu Y, Martin A, et al. Treatment of atrial fibrillation with antiarrhythmic drugs or radiofrequency ablation: two systematic literature reviews and meta-analyses. Circulation: Arrhythmia and Electrophysiology. 2009;2(4):349–61. pmid:19808490
  41. 41. Piccini JP, Lopes RD, Kong MH, Hasselblad V, Jackson K, Al-Khatib SM. Pulmonary vein isolation for the maintenance of sinus rhythm in patients with atrial fibrillation: a meta-analysis of randomized, controlled trials. Circulation: Arrhythmia and Electrophysiology. 2009;2(6):626–33. pmid:20009077
  42. 42. Khan AR, Khan S, Sheikh MA, Khuder S, Grubb B, Moukarbel GV. Catheter Ablation and Antiarrhythmic Drug Therapy as First- or Second-Line Therapy in the Management of Atrial Fibrillation. Circulation: Arrhythmia and Electrophysiology. 2014;7(5):853–60. pmid:25110162
  43. 43. Siontis KC, Ioannidis JPA, Katritsis GD, Noseworthy PA, Packer DL, Hummel JD, et al. Radiofrequency Ablation Versus Antiarrhythmic Drug Therapy for Atrial Fibrillation: Meta-Analysis of Quality of Life, Morbidity, and Mortality. JACC Clin Electrophysiol. 2016;2(2):170–80. Epub 2016/04/01. pmid:29766867.
  44. 44. Poole JE, Bahnson TD, Monahan KH, Johnson G, Rostami H, Silverstein AP, et al. Recurrence of Atrial Fibrillation After Catheter Ablation or Antiarrhythmic Drug Therapy in the CABANA Trial. Journal of the American College of Cardiology. 2020;75(25):3105–18. pmid:32586583
  45. 45. Zelniker TA, Bonaca MP, Furtado RHM, Mosenzon O, Kuder JF, Murphy SA, et al. Effect of Dapagliflozin on Atrial Fibrillation in Patients With Type 2 Diabetes Mellitus. Circulation. 2020;141(15):1227–34. pmid:31983236
  46. 46. Delgado V, Di Biase L, Leung M, Romero J, Tops LF, Casadei B, et al. Structure and Function of the Left Atrium and Left Atrial Appendage. Journal of the American College of Cardiology. 2017;70(25):3157–72. pmid:29268928
  47. 47. Gupta DK, Shah AM, Giugliano RP, Ruff CT, Antman EM, Grip LT, et al. Left atrial structure and function in atrial fibrillation: ENGAGE AF-TIMI 48. European Heart Journal. 2014;35(22):1457–65. pmid:24302269
  48. 48. Thewjitcharoen Y, Yenseung N, Malidaeng A, Nakasatien S, Chotwanvirat P, Krittiyawong S, et al. Effectiveness of long-term treatment with SGLT2 inhibitors: real-world evidence from a specialized diabetes center. Diabetology & Metabolic Syndrome. 2017;9(1). pmid:29213337