Skip to main content
Cureus logoLink to Cureus
. 2025 Aug 7;17(8):e89579. doi: 10.7759/cureus.89579

Optimizing Cardiovascular Outcomes With Bisoprolol: An Evidence-Based Perspective

Kamal Sharma 1,, Sunil Sathe 2, Bhupen Desai 3, Subhash Manchanda 4, Jagdish Mohan 5, Manish Bansal 6, Nagamalesh U M 7, Abraham Oomman 8, Arindam Pande 9, Jay Shah 10, Johann Christopher 11, Sachin Patil 12, Jabir Abdullakutty 13, Akshay Bafna 14, Sarita Rao 15
Editors: Alexander Muacevic, John R Adler
PMCID: PMC12414114  PMID: 40922852

Abstract

The cardiovascular continuum is the developmental process of cardiovascular diseases (CVDs) leading to heart failure (HF) and sudden cardiac death. Beta-blockers (BBs) are at the forefront of managing conditions along this continuum, ranging from cardiovascular (CV) risk factors to heart failure. In particular, bisoprolol proved to be a highly cardio-selective BB with a favourable pharmacokinetic profile, demonstrating long-term safety, good tolerability, and proven efficacy in reducing cardiac events, including arrhythmias and mortality in patients with heart failure with reduced ejection fraction (HFrEF). This evidence-based perspective showcased numerous clinical studies revealing the utility of bisoprolol in managing patients with CVDs, particularly HFrEF and stable angina. It also included safety evidence for bisoprolol in patients with renal and hepatic dysfunction, etc. Expert opinions from leading cardiologists across India further reinforce the role of bisoprolol as a first-line therapy in managing HFrEF and stable angina, making its usage suitable for HFrEF and angina patients with special emphasis on comorbidities, such as chronic kidney disease (CKD), chronic obstructive pulmonary disease (COPD), and diabetes.

Keywords: beta blocker, bisoprolol, cardiovascular disease, heart failure, hfref: heart failure with reduced ejection fraction

Introduction and background

Introduction

The cardiovascular continuum was first described in 1991 by Dzau and Braunwald as a new strategy for the management of cardiovascular diseases (CVD) [1]. The cardiovascular disease continuum begins with risk factors for cardiovascular diseases (CVDs), chronic stable angina, myocardial infarction (MI), and asymptomatic left ventricular dysfunction, and extends to refractory heart failure (HF) and death (Figure 1) [2]. Angina pectoris and HF are among the most prevalent CVDs.

Figure 1. The cardiovascular continuum.

Figure 1

Reference no. [2].

HF accounts for a large proportion of global health problems and is expected to affect approximately 64 million people worldwide [3]. In India, HF with reduced ejection fraction (HFrEF) is the most common type of HF, as shown by the Trivandrum HF registry (THFR) [4]. Similarly, the Kerala Acute Heart Failure Registry (CSI-KHFR) claims that 67.5% of patients with acute heart failure (AHF) exhibit reduced ejection fraction, primarily in younger populations [5].

Understanding the underlying pathophysiology is crucial for guiding targeted therapies. Central to the progression of HF are three neurohormonal systems: the sympathetic nervous system (SNS), renin-angiotensin-aldosterone system (RAAS), and natriuretic peptide system (NPS) [6]. These systems initially help maintain adequate cardiac output by increasing the heart rate (HR) and blood volume. However, chronic activation leads to adverse cardiac remodelling and progression to HF [7]. According to the CSI-KHFR, in patients with HF, the mortality rate is reported to be 59% at 5 years, and sudden cardiac death is the reason in 46% of the cases, showing a poor prognosis associated with this condition [5].

Another cardiovascular (CV) ailment, angina pectoris, results from an unbalanced supply-to-demand ratio of oxygen in the myocardium. Angina is not a standalone diagnosis but a symptom of the underlying cause of coronary artery disease (CAD), which can lead to myocardial ischemia and infarction. The pathophysiology of angina is multifactorial, involving epicardial coronary artery obstruction usually due to atherosclerosis, microvascular dysfunction, endothelial dysfunction, and dynamic plaque behavior [8]. In patients with angina, increased HR shortens myocardial diastole and consequently increases oxygen demand [9]. While HR suppression is considered a critical component in alleviating ischemic symptoms, addressing the broader pathophysiologic contributor is essential. Beta-blockers (BBs) by reducing myocardial oxygen demand, exert anti-ischemic effects and also provide broader cardiovascular benefits, including controlling heart rate, and improve survival in HF [10]. Both the THFR [4] and CSI-KHFR [5] registries highlight BB as one of the four cornerstone disease-modifying therapies shown to significantly improve long-term outcomes in patients with HFrEF.

Building on this rationale, bisoprolol, a cardioselective beta blocker (BB), plays a well-established role in the management of HFrEF and stable angina pectoris, primarily by reducing sympathetic overactivity and myocardial oxygen demand through inhibition of the overactive sympathetic nervous system (SNS) and renin-angiotensin-aldosterone system (RAAS). These characteristics also make bisoprolol a rational choice for long-term management of CVDs, especially in the Indian context, where comorbidities such as chronic kidney disease (CKD), diabetes, and chronic obstructive pulmonary disease (COPD) are common.

Objective of the meeting

Recognizing the evolving clinical landscape and real-world considerations of bisoprolol in cardiovascular care, this evidence-based perspective paper synthesizes insights from a forum of Indian cardiologists. The objective of this opinion paper was to provide a focused review on the clinical utility of BBs, particularly bisoprolol, not only in the later stages of the cardiovascular continuum, like HFrEF, but also in earlier phases, such as stable angina, which remains the central focus of this discussion. Additionally, the paper seeks to explore its potential application in patients with commonly encountered comorbidities, acknowledging the multifaceted nature of patient care in routine clinical practice.

Review

Methodology

A series of structured expert group meetings were conducted with 56 leading cardiologists from across India representing all major regions-North (16), East (6), West (19) and South (15) Discussions focused on the positioning of bisoprolol among BBs in guideline-directed medical therapy (GDMT), its use in patients with comorbid conditions, and specific patient profiles favoring bisoprolol. Key discussion points included receptor selectivity, dosing considerations, and real-world applicability across diverse patient subgroups.

Beta blockers

Beta blockers (BBs) primarily act on β1 adrenergic receptors in cardiac myocytes and juxtaglomerular cells of the kidney and work by blocking β1 adrenoreceptors, leading to reduced HR and consequent reduction in myocardial oxygen demand and cardiac workload. This makes them a critical component in the management of multiple CVDs, including HFrEF and stable angina pectoris [10]. Additionally, non-selective BBs block β2 receptors, mainly found in vascular and airway smooth muscle, potentially causing bronchoconstriction and vasoconstriction, thus requiring caution in patients with asthma or peripheral artery disease [11].Bisoprolol

Bisoprolol is a cardioselective β1-adrenergic receptor antagonist and is among the GDMTs for HFrEF that are intended to reduce the mortality rate and hospital readmissions based on the 2022 AHA/ACC/HFSA guidelines [12]. It helps in reducing morbidity and mortality post-myocardial infarction (MI) and the occurrence of stroke and CAD [13]. Bisoprolol's favorable pharmacokinetic (PK) profile supports its use as a BB of choice within the GDMT framework. It is well-absorbed with 90% oral bioavailability. Maximum plasma levels are attained approximately 3 hrs after a dose of 10 mg bisoprolol [14]. Approximately 50% of bisoprolol is metabolized by the liver, mainly involving CYP3A4, and is converted into inactive metabolites, while the other 50% is excreted unchanged by the kidneys [15]. Its elimination half-life is 10-11 hours in healthy individuals, extending to 18 hours in those with renal impairment and 13 hours in individuals with hepatic cirrhosis, supporting once-daily dosing [16]. Due to its high oral bioavailability and low first-pass metabolism, bisoprolol has linear pharmacokinetics over a wide range of dosage regimens [17]. Comparison of PK properties of bisoprolol vs other BBs is mentioned in Table 1.

Table 1. Comparative profile of beta blockers .

Parameter Bisoprolol Carvedilol Metoprolol Succinate Nebivolol
β1 Selectivity [18] High (β1-selective) (Affinity (pKD) β1=7.43-7.98; β2=5.42-6.70; β3=5.04-5.67) Non-selective (β1, β2) + α1-blocking (Affinity (pKD) β1=8.75-9.26; β2=8.96-10.06 β3=6.61-8.30) Moderate (β1-selective) (Affinity (pKD) β1=7.26-7.36; β2=5.49-6.89; β3=5.00-5.16) High (β1-selective) (Affinity (pKD) β1=8.79-9.17; β2=6.55-7.96 β3=5.66-7.04)
Vasodilatory Properties [19] None Yes (via α1-blockade) None Yes (via nitric oxide release)
Half-life (hours) [19] 9–12 7–10 3–4 10–30( depending on CYP polymorphism)
Metabolism [20] 50% hepatic, 50% renal; no dose adjustment in hepatic/renal impairment Hepatic (CYP2D6); caution in hepatic impairment Hepatic (CYP2D6); caution in hepatic impairment Hepatic (CYP2D6); caution in hepatic impairment
Impact on Glucose Metabolism [21] Neutral May improve insulin sensitivity; favorable in diabetics May worsen glycemic control Improves insulin sensitivity; favorable in diabetics
Impact on Lipid Metabolism  [22] Neutral May improve lipid profile May adversely affect lipid profile May improve lipid profile
Suitability in Diabetes [23], [24] Suitable Preferred due to metabolic benefits Use with caution Preferred due to metabolic benefits
Suitability in COPD/Asthma [25] Preferred due to β1-selectivity Use with caution; non-selective β-blockade may exacerbate symptoms Use with caution; less β1-selective Preferred due to β1-selectivity
Dosing Frequency Once daily Twice daily Once daily Once daily

Bisoprolol, with its low pharmacokinetic (PK) variability, offers more consistent drug exposure across individuals, reducing the need for frequent dose adjustments, making it a more reliable option than other BBs [26]. However, dose adjustment may be needed in elderly patients, as discontinuation is more common than in large mortality trials, primarily due to symptomatic hypotension limiting dose escalation [27]. In patients with severe renal or hepatic dysfunction, slow titration and close monitoring are advised due to extended half-life and accumulation potential [28].

Use of BBs in the management of HFrEF

In HFrEF, chronic β-adrenergic receptor activation leads to excessive catecholamine stimulation, myocardial apoptosis and fibrosis. BBs are fundamental in treating HFrEF due to their ability to attenuate the action of catecholamines, thereby improving myocardial function and reducing cardiac sympathetic overdrive [29]. Furthermore, effective use of BBs can help enhance the symptoms of HFrEF, minimize hospital readmissions, and increase survival duration [29]. Both Indian and international guidelines support the use of BBs (bisoprolol, carvedilol, or metoprolol) as a first-line treatment for HFrEF [7,12,30,31] (Table 2). Table 3 summarizes key clinical trials evaluating the efficacy of different BBs in HFrEF, including bisoprolol, carvedilol, and metoprolol, highlighting their comparative impact on mortality, hospitalization, and overall outcomes. Collectively, these trials confirmed the efficacy of BB therapy in HFrEF patients. The MERIT-HF trial demonstrated the clinical benefits of metoprolol CR/XL (controlled release/ extended release) in HFrEF patients, reducing mortality significantly by 34% [32]. Similarly, the COPERNICUS trial showed a 35% mortality reduction with carvedilol in HFrEF patients [33]. The US Carvedilol trial reported a 65% relative risk reduction (RRR) in mortality and CV hospitalization with carvedilol [34]. Bisoprolol is a recommended evidence-based BBs in guidelines for the management of HFrEF [12,31,35,36] (Table 2) and has shown significant efficacy and safety in patients with HFrEF through several randomized controlled trials (CIBIS, CIBIS II, and CIBIS III) (Table 4). The initial Cardiac Insufficiency Bisoprolol Study (CIBIS) evaluated 641 patients with HFrEF, and bisoprolol was associated with 20% relative risk reduction in mortality compared to placebo (relative risk 0.80, CI: 0.56, 1.15; p=0.22); however the result did not reach statistical significance largely due to being underpowered, with a small sample size and lower-than-expected event rates. However, bisoprolol significantly improved the patient’s New York Heart Association (NYHA) class (p=0.04) [37]. The subsequent CIBIS II trial [38], which included a larger cohort of 2647 patients, was stopped early due to a significant mortality benefit with bisoprolol. Treatment with bisoprolol led to 34 % RRR in all-cause mortality (HR: 0.66;95% CI 0.54-0.81; p < 0.0001) and 44% RRR in sudden death ( HR: 0.56;95% CI 0.39-0.80; p = 0.0011) compared to placebo [38]. This survival benefit was observed across all tolerated dose levels, and withdrawal was associated with a significant increase in mortality rate in the bisoprolol group with a relative hazard (RH) of 2.13 (95% CI=1.43-3.17, p=0.0002) [39]. Although post-hoc analyses are exploratory and not sufficient to drive guideline-level recommendations, findings from post-hoc analyses of the CIBIS II trials indicated consistent benefits of bisoprolol across various patient subgroups, including patients with or without diabetes, with or without renal impairment, with or without severe HF (NYHA class IV) symptoms, and patients with age ≥71 years Vs. <71 [40]. The third trial, CIBIS III, evaluated the sequence of initiating HF therapy with either bisoprolol or enalapril, an angiotensin-converting enzyme inhibitor (ACEi). The trial observed no significant differences in clinical outcomes between the initiation of bisoprolol or ACEi and the addition of another drug. However, it highlighted the importance of dose optimization, demonstrating that the therapy initiated first was more likely to be titrated to at least half of the maximal dose [41]. A meta-analysis of the CIBIS I and CIBIS II trials also proved the effectiveness of bisoprolol in reducing overall death, cardiovascular death, and hospitalizations [42].

Table 2. Guideline recommendation for use of beta blockers for HFrEF.

COR: Class of recommendation; LOE: Level of evidence; BP: Blood pressure; HF: Heart failure, HFrEF: Heart failure with reduced ejection fraction, BB: Beta blocker, OD: Once daily, CV: Cardiovascular.

Guideline Recommendation COR LOE
European Society of Cardiology (ESC) 2021 [31] BB is recommended for patients with stable HFrEF to reduce the risk of HF hospitalization and death I A
American Heart Association (AHA)/ACC/Heart Failure Society of America (HFSA) 2022 [12]   In patients with HFrEF, with current or previous symptoms, use of 1 of the 3 BBs proven to reduce mortality (i.e. bisoprolol, carvedilol, sustained-release metoprolol succinate) is recommended to reduce mortality and hospitalizations. I     A
Start evidence-based BB at their initial dose; bisoprolol to start at 1.25 mg OD; consider increasing dose every 2 weeks until maximum tolerated or targeted dose (10 mg OD) is achieved.
Delphi Survey- based Consensus from Asia-Pacific 2023 [29] BB are recommended as first-line treatment in patients with HFrEF to reduce all-cause mortality, CV mortality, and HF-related hospitalization. Consistent benefits have been observed by age, sex, cause (i.e., ischemic vs. non-ischemic cardiomyopathy), degree of symptoms, and degree of systolic dysfunction. They also improved left ventricular function.
India Consensus 2020  [43] At discharge from hospital: BB reduces the risk of all-cause and CV mortality but increase the risk of bradycardia and hypotension; once BP is stable, BB can be safely administered. The dose should be carefully increased to reduce the HR to around 70 beats per minute

Table 3. Summary of major beta blocker trials in HFrEF.

HR: Hazard ratio, RAAS: Renin-angiotensin-aldosterone system, RR: Relative risk, RRR: Relative risk reduction, SNS: Sympathetic nervous system,↓: Decrease or reduction,↑: Increase or elevation.

Trial & Drug Patients (n) Comparator HR / RR / RRR (95% CI) Key Findings
Bisoprolol – CIBIS I [37] 641 Placebo HR 0.80 [0.56–1.15], p=0.21 No mortality benefit; ↓ HF events
Bisoprolol – CIBIS II [38] 2,647 Placebo HR 0.66 [0.54–0.81], p<0.0001 ↓ All-cause & sudden death (HR 0.56 [0.39–0.80], p=0.0011)
Bisoprolol – CIBIS III [41] 1,010 Enalapril-first HR 0.94 [0.77–1.16] No difference in death/hospitalization for ACEI vs BB-first
Metoprolol CR/XL – MERIT-HF [32] 3,991 Placebo RR 0.66 [0.53–0.81], p=0.0062 ↓ All-cause death; ↓ sudden death (RR 0.59), ↓ HF death (RR 0.51)
Carvedilol – US Trial [34] 1,094 Placebo RRR 65% [39–80], p<0.001 ↓ Mortality; ↓ CV hospitalization
Carvedilol – COPERNICUS [33] 2,289 Placebo RRR 35% [19–48], p=0.0014 ↓ All-cause death; ↓ death/hospitalization (RRR 24%)

Table 4. Clinical evidence of Bisoprolol in HFrEF.

CIBIS: Cardiac Insufficiency Bisoprolol Study, HFrEF: Heart Failure with reduced Ejection Fraction, NYHA: New York Heart Association, LVEF: Left Ventricular Ejection Fraction, CHF: Chronic Heart Failure, ACEi: Angiotensin Converting Enzyme inhibitor, ARB: Angiotensin Receptor Blocker, BB: β Blocker, QD: Once a day, BID: Two times a day, HR: Hazard ratio, (HR <1.0 indicates reduced risk over time in the treatment group compared to control). CI: Confidence Interval, CV: Cardiovascular.

Study Population Number of patients Dosage of bisoprolol/ comparator Mean follow-up Primary end point results Other results
CIBIS I (Placebo-controlled RCT ) [37] N=641 symptomatic HFrEF patients (LVEF  <40%) NYHA class III or IV, Bisoprolol (n=320) or placebo (n=321) 1.25 mg daily and progressively increased to 5 mg daily 1.9 ± 0.1 years. All-cause mortality was lower with bisoprolol (16.6%) vs placebo (20.9%), but not statistically significant (RR 0.80; 95% CI 0.56–1.15; p=0.22) Bisoprolol significantly improved functional status, with fewer hospitalizations for cardiac decompensation (61 vs. 90; p<0.01) and more patients improving by ≥1 NYHA class (68 vs. 48; p=0.04)
CIBIS II [38] N=2647 symptomatic HFrEF patients NYHA class III or IV, (LVEF ≤35%) Bisoprolol (n=1327) or placebo (n=1320) 1.25 mg daily and progressively increased to 10 mg daily 1.3 years All-cause mortality was significantly lower with bisoprolol than placebo ( [11·8%] vs [17·3%] deaths with a HR of 0·66 (95% CI 0·54–0·81, p<0·0001) 20% less all-cause hospital admission in bisoprolol group than on placebo (p=0·0006)  Significant reduction in combined endpoint of CV death and admission to hospital for CV events ( p=0.0004) Significant reduction in sudden death by 44%
CIBIS III [41] N=1010 HFrEF patients (LVEF ≤35%) not receiving ACEi, BB, or ARB Bisoprolol (n=505) or enalapril (n=505) Bisoprolol (target dose 10 mg QD) or enalapril (target dose 10 mg BID) for 6 months, followed by their combination for 6–24 months 1–2.5 years All-cause mortality : bisoprolol- was noninferior to enalapril- treatment In intent-to treat group, (HR 0.94; 95% CI 0.77 to 1.16, p=0.019) per-protocol sample (HR 0.97; 95% CI 0.78 to 1.21; P=0.046) Death: 65 vs 73 pts (HR 0.88, P=0.44)   Hospitalizations: 151 vs 157 (HR 0.95, p=0.66)   Worsening of CHF requiring hospitalization: 63 vs 51 (HR 1.25, p = 0.23)

Figure 2 provides a comparative visual summary of key outcomes, including total mortality and the composite endpoint of death or hospitalization, across the CIBIS I, II, and III trials. It highlights the consistent trend of improved outcomes in the bisoprolol groups versus comparator arms, establishing the clinical benefits of bisoprolol in patients with HFrEF.

Figure 2. Key clinical outcomes from CIBIS trials comparing bisoprolol vs placebo (CIBIS, CIBIS II) and vs enalapril (CIBIS III) in HFrEF.

Figure 2

Reference numbers [37,38,41].

HFrEF: Heart failure with reduced ejection fraction.

Although BBs have comparable effectiveness in HFrEF management, pharmacokinetic differences, impact on glucose and lipid metabolism, and effects in the presence of comorbidities distinguish them from each other [26] (Table 1). The expert recommendations on the use of BBs in patients with HFrEF are given in Table 5. While bisoprolol remains a key component in the management of HFrEF, the role of fixed-dose combinations is increasingly recognized for their potential to enhance treatment adherence and simplify therapy. Looking ahead, combining bisoprolol with other pillars of GDMT, such as angiotensin receptor-neprilysin inhibitors (ARNI), sodium-glucose co-transporter-2 inhibitors (SGLT2i), and mineralocorticoid receptor antagonists (MRA)in a fixed-dose combination (FDC) format could represent a promising direction in HF management. Efforts are underway to explore the feasibility and potential clinical utility of some FDCs in HFrEF management.

Table 5. Table of recommendations.

Experts' Opinion on Use of beta-blockers (BBs) in in HFrEF
BB is an essential component of GDMT for HFrEF, and its benefits are seen regardless of the dosage and sequence in the GDMT. BBs, including bisoprolol, should be initiated in patients with HFrEF unless contraindicated, starting at low doses and titrating up to target dose or the maximally tolerated dose. Bisoprolol is preferred BB considering high cardioselectivity, low pharmacokinetic variability, neutral effect on lipid levels and glucose metabolism making it suitable for use in patients who have cardiovascular risk factors, diabetes, or prediabetes.  
Experts' Opinion on Use of BBs in Stable Angina Pectoris
BBs are to be used as the first-line therapy for the treatment of angina either as a single therapy or in conjunction with CCBs. Bisoprolol has shown long term effectiveness in angina patients. Bisoprolol is preferred for angina due to high cardioselectivity, low pharmacokinetic variability, neutral effect on lipid levels and glucose metabolism making it suitable for use in patients who have cardiovascular risk factors, diabetes, or prediabetes. Bisoprolol treatment is linked to better CV outcomes than other BBs and non-BB medications.
Experts' Opinion on Use of BBs in Patients with CVD having Renal Impairment
Compared to other BBs, bisoprolol is safer in patients with renal disease because of its favourable balance of pharmacokinetics, cardio selective action, minimal interactions, and tolerability. No adjustment of bisoprolol dosage required for patients with renal impairment. Cardioselective BBs like bisoprolol provide superior survival benefits than other BBs such as carvedilol among high-risk patients, like patients on maintenance haemodialysis.  
Experts' Opinion on Use of BBs in Patients with CVD having Hepatic Impairment
Bisoprolol is safe to use in patients with liver impairments, however for hepatic cirrhosis and portal hypertension in end-stage cardiac failure, carvedilol is preferred.  
Experts' Opinion on Use of BBs in Patients with CVD having Chronic Airway Diseases
Cardioselectivity of bisoprolol helps minimize respiratory side effects, making it safer for patients with chronic airway diseases like COPD. Bisoprolol is associated with a significantly better dose-dependent survival outcome in patients with HF and COPD compared to non-users. Bisoprolol is associated with better clinical outcomes in patients with COPD and CVD, including reduced all-cause mortality and lower rates of hospitalization for heart failure exacerbations compared to carvedilol. Bisoprolol leads to better lung function outcomes compared to other BBs such as carvedilol and metoprolol. Bisoprolol can be administered as an antianginal treatment in patients with moderate persistent bronchial asthma at a dose not exceeding 5 mg while avoiding bronchial obstruction.

Use of BBs in the management of stable angina pectoris

Various guidelines recommend the use of BBs as the first-line therapy for patients with angina (Table 5). The 2024 European Society of Cardiology (ESC) guidelines recommend BBs as the first-line treatment for angina either as a monotherapy or as a combination with calcium channel blockers (CCBs) (Class IB recommendation) [44]. The ESC guidelines appear to have a different stance on BB use compared to the American College of Cardiology/American Heart Association (ACC/AHA) guidelines (Table 5). This can be due to different approaches to guideline development and the interpretation of evidence.

Evidence from the literature also supports the effective use of bisoprolol in patients with stable angina (Table 6). Long-term (five years) data showed a significant reduction in mortality risk and improved cardiovascular outcomes in angina patients, supporting bisoprolol as a first-line therapy in real-world primary care over other BBs and non-BB agents [45]. Several studies have highlighted the efficacy of bisoprolol in reducing angina symptoms and improving exercise tolerance [46-48]. Studies demonstrating a comparison of the safety and efficacy of bisoprolol (10 mg once daily) with atenolol (100 mg once daily) reported a significant reduction in the weekly anginal attack rate, demonstrating equivalent effectiveness as an antianginal drug with no significant difference in nature and incidence of an adverse event [49-51]. Bisoprolol has also been compared with other anti-anginal medications, such as CCBs and nitrates. In a study by Van De Ven et al., bisoprolol was compared with isosorbide dinitrate and was found to be more effective at improving exercise tolerance during bicycle exercise testing and reducing the time to onset of angina, with fewer side effects [52]. In the Total Ischemic Burden Bisoprolol Study (TIBBS study) with 330 patients, bisoprolol showed significantly higher responder rates than nifedipine in reducing the number of ischemic episodes(100% reduction: 40.6% vs 14.8% at low dose; 52.5% vs 15.6% at high dose; p < 0.0001). Only bisoprolol showed a circadian effect by lowering the morning peak of ischemic episodes by 68% while with nifedipine, the circadian profile was unchanged [53]. Bisoprolol has also demonstrated long-term antianginal effects. Three-month therapy with bisoprolol in 6799 Stage 1 arterial hypertension patients showed a significant reduction in blood pressure (BP) and the number of angina attacks, with weekly episodes dropping from 2.03 to 0.91 [54]. In another study involving 222 hypertensive patients with stable angina, significant improvements were observed in flow-mediated vasodilatation (FMD) and myocardial survival after 12 months of treatment [55]. As reported by Terol et al., bisoprolol was well tolerated among 236 angina patients. Eighty-nine percent of patients reported a decrease in angina attacks; 56% were entirely free of attacks, and 5.1% showed beta blocker-specific side effects, illustrating the efficacy of bisoprolol and a favourable safety profile [56]. Expert recommendations on the use of BBs in patients with stable angina are presented in Table 7.

Table 6. Guideline recommendation for role of BBs in chronic stable angina pectoris.

COR: Class of Recommendation, LOE: Level of Evidence, Class I (strong recommendation), Class II (moderate recommendation), Level A: High-quality evidence from multiple randomized controlled trials (RCTs) or meta-analyses, Level B: Evidence from a single randomized controlled trial or large non-randomized studies, MI: Myocardial Infarction, LVEF: Left Ventricular Ejection Fraction.

Guideline Patient profile                                                                                               COR LOE
American College of Cardiology /American Heart Association (ACC/AHA) 2012 [8] Uncomplicated patient I B
Previous MI I B
Reduced LVEF (<40 %) I A
European Society of Cardiology (ESC) [57] 2024    Uncomplicated patient I B
Previous MI II B
Reduced LVEF (<40 %) II B
National Institute for Health and Care Excellence 2019 [58] Uncomplicated patient First line treatment (if symptoms are not controllable, switch to CCB or use both)
Previous MI
Reduced LVEF (<40 %)
Canadian Cardiovascular Society 2014 [59] Uncomplicated patient II B
Previous MI II B
Reduced LVEF (<40 %) I A

Table 7. Clinical evidence of bisoprolol instable angina pectoris.

BB: Beta blocker; HR: Heart rate; SBP: Systolic blood pressure; RPP: Rate-pressure product; GTN: Glyceryl trinitrate (nitroglycerin); CHD: Coronary heart disease; CAD: Coronary artery disease; TID: Three times a day; BID: Twice a day.

Study Population Sample size Dosage of bisoprolol/ comparator Findings
Sabido et al. 2019 [45]. Real-world study, cohort analysis 7607 angina patients Bisoprolol (N=987) Other BBs (N=1348) Other than BBs (N=5272) -- 55% less mortality risk and  42% less risk of angina attack with bisoprolol vs other BBs
De Divitiis et al. 1987 [46]. Double-blind parallel groups 26 angina patients     Bisoprolol (N=13) Verapamil (N=13)   Bisoprolol 10-20 mg/day. Verapamil: 80-120 mg TID. Both treatments significantly reduced angina episodes, nitroglycerin tablet consumption, ischemic episodes, and ST depression
Kato et al. 1986 [47]. Single blind study 36 stable angina patients N=36 Bisoprolol 5 to 10 mg/day Bisoprolol significantly reduced the HR frequency of anginal attacks, HR, SBP, and RPP at peak exercise and nitroglycerin consumption
Schnellbacher et al. 1986 [48]. Placebo-controlled double-blind crossover study 12 stable angina patients N=12 Bisoprolol 10 mg/day. Atenolol 100 mg/day   Bisoprolol reduced exercise-induced symptoms, HR, RPP, and ischemic ST depression
MIRSA study [49]. Double-blind, randomized, parallel-group study 147 stable angina patients Bisoprolol (N=76)   Atenolol (N=71)   Bisoprolol 10 mg/day. Atenolol 100 mg/day   Both drugs significantly reduced angina attacks and RPP; similar exercise capacity improvement
Maltz et al. 1987 [50]. Double-blind, randomized, three-way crossover study 19 CAD and angina patients N=19 Bisoprolol 5 mg/day, 10 mg/day. Atenolol 100 mg/day   All regimens reduced angina symptoms, GTN use, and HR (p<0.01); similar exercise time improvement
Kohli et al. 1986 [60]. Placebo-controlled double-blind RCT 20 stable angina patients N=20 Bisoprolol 10 mg/day. Atenolol 100 mg/day   Bisoprolol and Atenolol improved exercise time and prolonged time to ST  depression (p<0.001). A significant decrease in the peak exercise HR was seen with both drugs (p<0.001).
Van De Ven et al. 1995 [52]   Double-blind, randomized crossover study 27 CAD and stable angina patients (Class II-III) Treatment order group   Nitrate/ Bisoprolol (N=14) Bisoprolol/nitrate (N=71)   Bisoprolol 10 mg/day. Isosorbide dinitrate 20 mg TID    Bisoprolol improved cardiac workload and time to angina onset; fewer side effects vs nitrates
TIBBS study 1995 [53]. Randomized double-blind, parallel group study 330 stable angina patients with transient ischemic episodes Bisoprolol (N=161) Nifedipine SR (N=169)   Phase 1: bisoprolol (10 mg /day). Nifedipine SR (2 x 20 mg/day). Phase 2: bisoprolol (20 mg/day), nifedipine SR (2x 40 mg/day). Statistically significant reduction in the number of transient ischemic episodes and total duration of ischemia (p<0.0001) by both drugs. Bisoprolol demonstrated a distinctive circadian impact by reducing the morning peak of transient ischemic episodes by 68% compared to Nifedipine SR. Bisoprolol lowered the HR significantly (p < 0.001), while nifedipine had no effect on HR

Use of BBs in patients with CVD and renal impairment

The prevalence of concomitant HF and chronic kidney disease (CKD) is high, with 40% of patients affected by HF demonstrating renal dysfunction. As well as, the proportion of HF in CKD patients ranges between 17-50% depending on the stage of CKD and the patient’s age [61]. Studies have described the clinical advantages of BB therapy irrespective of CKD stage, and indicated significant reductions in all-cause mortality and cardiovascular outcomes in patients with HF [62,63]. Bisoprolol is mainly metabolized in the kidneys as well as the liver, since both organs contribute to half of the elimination of the drug each. This balanced excretion reduces the dependency on any single organ for drug elimination, minimizing the risk of drug accumulation and toxicity in patients with either renal or hepatic impairment. In addition, bisoprolol plasma level remains consistent, which reduces the need for frequent dose adjustment in patients with mild-to-moderate dysfunction. However, bisoprolol dosage should not exceed 10 mg daily in severe and end-stage renal failure [64]. In contrast, the BBs, such as atenolol and metoprolol, depend greatly on renal and hepatic elimination, respectively, which may cause adverse effects in patients with impairment of one of the elimination organs [64]. Unlike metoprolol and carvedilol, which are primarily metabolized by the liver through the cytochrome P450 system, especially CYP2D6, bisoprolol undergoes minimal CYP450 metabolism. This reduces its potential for drug-drug interactions, which is particularly important in patients with renal impairment on multiple medications [65].

Another factor contributing to the safety profile of BBs is dialyzability, as this significantly influences clinical decision-making in patients undergoing hemodialysis, affecting drug clearance, dosing strategies, and therapeutic consistency. Highly dialyzable BBs (e.g., metoprolol, atenolol) are substantially removed during dialysis, which may lead to subtherapeutic levels post-session, reducing cardiovascular protection and requiring additional dosing adjustments. Although bisoprolol is poorly dialyzable, it has been shown to be partially cleared during hemodialysis, but to a significantly lesser extent than atenolol or metoprolol. However, it maintains more stable plasma levels due to its high bioavailability and balanced renal and hepatic clearance, making it a safer and more consistent β-blocker option for hemodialysis patients [66].

In contrast, carvedilol, though poorly dialyzable, with additional α-1 blocking properties, is more likely to cause intradialytic hypotension in dialysis patients [67,68]. In patients with severe renal impairment (creatinine clearance <10 mL/min), bisoprolol exposure doubles, and its plasma elimination half-life extends to approximately 24.2 hours [28]. The subgroup analysis of the CIBIS II trial also demonstrated the efficacy of bisoprolol in HFrEF patients with renal impairment. Mortality risk was similarly reduced in patients with CrCl ≤ 60 mL/min (RR 0.66, 95% CI 0.50-0.88) and >60 mL/min (RR 0.69, 95% CI 0.54-0.89), with no significant interaction (p=0.69) [40]. In dialysis-dependent patients, cardioselective BBs are associated with 15% fewer CV events (HR =0.85; 95% CI =0.81, 0.89) and 17% lower all-cause mortality (HR =0.83; 95% CI =0.69, 0.99) than non-selective BBs [69].

Furthermore observational data suggest that, bisoprolol was associated with lower all-cause mortality (HR 0.66; 95% CI 0.60-0.73) and reduced risk of major adverse cardiovascular events (MACEs) (HR 0.85; 95% CI 0.80-0.91), HF (HR 0.83; 95% CI 0.77-0.92), and ischemic stroke (HR 0.84; 95% CI 0.72-0.97) compared to carvedilol in patients undergoing maintenance hemodialysis for more than 90 days [67,70]. Robust real-world observational data can help guide BB choice in patients undergoing dialysis, but should be interpreted cautiously due to potential confounding factors, bias, and patient variability.

Use of BB in patients with CVD and hepatic impairments

Hepatic dysfunction is a common comorbidity in patients with CHF and CVD, often complicating pharmacologic management due to altered drug metabolism and increased vulnerability to adverse effects. Liver impairment is prevalent in CHF [71], and choosing an appropriate BB can be challenging in this population. Among BBs, carvedilol, a non-selective BB, is the drug of choice for patients with cirrhosis. Carvedilol has been shown to offer a significant survival advantage, with a median survival of 7.8 years [72], and achieves a higher rate of hemodynamic response in reducing portal pressure, a major complication of cirrhosis, compared to propranolol, further supporting its potency in cirrhotic patients with portal hypertension [73].

However, when considering overall cardiovascular protection in patients with cirrhosis beyond portal hypertension, a 13-year nationwide population-based study in Asia found that compared to nonselective β1-blockers, β1-selective blockers (bisoprolol, metoprolol, atenolol) were associated with a 38% lower risks of major adverse cardiac and cerebrovascular events (HR=0.62; 95% CI =0.42-0.91; p=0.014, a 34% reduction in all-cause mortality (HR=0.66; 95% CI= 0.38-1.14; p=0.135, and a similar non-significant trend was observed for worsening liver outcomes in cirrhosis patients (HR=0.66; 95% CI=0.38-1.14; p=0.354) [74].

Use of BBs in patients with CVD with glucose and lipid abnormalities

Bisoprolol has been described to have a generally neutral effect on lipid metabolism, which makes it the preferred BB for patients with vulnerable lipid profiles. Unlike non-selective BBs, which can raise triglycerides and LDL cholesterol while lowering HDL cholesterol, bisoprolol is less likely to cause metabolic derangements that include lipid levels, insulin sensitivity, and alterations in blood sugar than non-selective and partially selective BBs [75]. Bisoprolol did not adversely affect lipid profiles even in the presence of cardiovascular risk factors and type 2 diabetes [76]. A comparison of bisoprolol’s effects on lipid profile and glucose metabolism versus other BBs is presented in Table 1.

In the TENACITY study, bisoprolol improved left ventricular ejection fraction (LVEF) and reduced HR in post-MI patients with left ventricular dysfunction, without adversely altering lipid or glucose metabolism [13]. In a meta-analysis involving bisoprolol versus other BBs, including metoprolol and atenolol, bisoprolol demonstrated its effectiveness in lowering BP and heart rate with significant improvement in HDL cholesterol at 52 weeks (p<0.01) and 104 weeks (p<0.01) [77].

Another positive aspect of bisoprolol is its neutrality or even the possibility of a slightly beneficial effect on glucose metabolism, while being used in patients with HF and angina having diabetes or prediabetes [78]. A double-blind, crossover study indicated that bisoprolol 10 mg OD did not affect blood glucose and HbA1C in patients with diabetes compared to placebo, confirming its safety profile in this population [79]. A similar neutral effect was observed in young individuals with hypertension and obesity [76].

Use of BBs in patients with CVD and chronic airway diseases

Chronic obstructive pulmonary disease (COPD) and asthma are often comorbid with CVDs such as ischemic heart disease, congestive heart failure (CHF), and cardiac arrhythmia. Similarly, 10-46% COPD patients show evidence of abnormal LVEF [80]. Despite their clinical benefits, BBs are often under-prescribed in this population because of concerns over bronchospasm. However, emerging evidence supports the safe and effective use of cardioselective BBs in patients with chronic airway diseases and coexisting CVD.

COPD

A meta-analysis evaluating the use of BBs in COPD patients with concomitant CVD has documented clinical benefit in improving survival and decreasing the risk of re-hospitalization [81,82]. Neef et al. [83] showed that cardioselective β1-blockers are well tolerated in COPD patients, even during hospitalization for exacerbation.

As being cardioselective BBs, bisoprolol selectively blocks β-1 adrenergic receptors primarily found in the heart, with less impact on β-2 receptors in the lungs. This selectivity is crucial for minimizing respiratory side effects in patients with chronic airway diseases [84]. It is associated with improved clinical outcomes in patients with COPD and CVD compared to non-selective BBs such as carvedilol and metoprolol. Its use in patients with COPD and HF has been linked to reduced all-cause mortality and lower rates of hospitalization due to HF exacerbations [85-87]. A study by Su et al. involving 11,558 patients found that bisoprolol use was associated with a significant dose-dependent mortality benefit, low-dose HR, 0.76 (95% CI, 0.59-0.97, p = 0.030), high-dose bisoprolol HR, 0.40 (95% CI, 0.26-0.63), p< 0.001) compared to non-users [85]. Similar improved clinical outcomes have been reported in other studies [86,87]. These outcomes include a lower incidence of CHF and/or COPD exacerbation (p=0.033) [87], reduced mortality, and decreased hospitalisation rate [71] in the bisoprolol arm compared to the carvedilol group.

A comparison of bisoprolol and carvedilol in 63 BB naive patients with CHF and moderate-to-severe COPD found a significant increase in forced expiratory volume in one second (FEV1) in the bisoprolol arm (1.56 ± 0.41 to 1.70 ± 0.52 L; p=0.046) but not in the carvedilol arm [88]. In a triple-crossover trial, bisoprolol led to the highest FEV1 among the beta blockers tested, followed by metoprolol and carvedilol [89].

Additionally bisoprolol reduced mortality and hospitalization rate due to CHF exacerbations more effectively than carvedilol and metoprolol in patients with coexistent CHF and COPD (mortality: adjusted hazard ratio (aHR) =0.51, 95% CI, 0.29-0.89; hospitalization rate due to CHF exacerbation: aHR =0.48, 95% CI, 0.23-1.00) [86]. A systematic review and meta-analysis involving 3269 adults with COPD with or without HF who were treated with bisoprolol concluded that compared to the control group, bisoprolol improved lung function and exercise performance while reducing inflammatory markers without significant adverse effects. Bisoprolol showed improvements in FEV1 (mean difference (MD) 0.46, 95% CI 0.27 to 0.65, p=0.000), FEV1% (MD 0.64, 95% CI 0.42 to 0.86, p=0.000), and a modest increase in FVC (MD 0.20, 95% CI 0.05 to 0.34, P=0.008) [90]. In patients with non-asthmatic chronic obstructive lung disease and co-existing stable angina pectoris, bisoprolol was found to be effective in its therapeutic dose range (5-20 mg OD), with no increased wheeziness or shortness of breath, highlighting its safety and efficacy in such patients [91].

Asthma

The use of cardioselective BB in patients with asthma and CVD did not significantly increase the risk of moderate or severe asthma exacerbations. A meta-analysis of 19 trials evaluating the effects of cardioselective ß-blockers on FEV1 found that a single dose led to a 7.46% decrease in FEV1 [92]. In a network meta-analysis, assessing risk of exacerbation with various BB in asthma patients, oral bisoprolol had a risk ratio (RR) of 0.46 (95% CI 0.02-11.65), suggesting that bisoprolol does not significantly increase the risk of asthma attacks [93]. However, the extremely wide confidence interval reflects substantial statistical uncertainty, limiting the ability to draw definitive conclusions. It can be administered as an antianginal treatment in patients with moderate persistent bronchial asthma at a dose not exceeding 5 mg to prevent bronchial obstruction [94]. The expert recommendations on the use of BBs in patients with chronic airway diseases are presented in Table 7.

Contraindications for bisoprolol

Bisoprolol is contraindicated in patients with marked sinus bradycardia, cardiogenic shock, and complete heart block, and should be used with caution in those with second-degree AV block or recent fluid retention without a diuretic. They are generally safe in mild to moderate asthma or COPD, but should be avoided in severe cases. In diabetes, they may mask hypoglycemia, requiring close monitoring.

Study limitations

This expert opinion provides valuable, practice-oriented guidance on the use of bisoprolol in HFrEF and stable angina, with both clinical trial evidence and real-world experience. However, its generalizability is limited by the lack of direct head-to-head comparisons with other BBs, limited representation of specific patient populations, such as the elderly, those with CKD, and COPD, and reliance on expert consensus rather than prospective, large-scale studies.

Conclusions

Bisoprolol, as one of the selective β1-adrenergic antagonists, plays a crucial role in the treatment of HFrEF and stable angina pectoris. Its β1 selectivity and balanced hepato-renal metabolism provide a reliable and predictable therapeutic effect, which makes it suitable for patients with HFrEF and stable angina having renal or hepatic impairment, and COPD. While bisoprolol has shown favorable outcomes in patients with COPD, careful patient selection remains essential when used in those with chronic airway diseases, particularly in asthma or individuals with severe airway reactivity, where safety data are limited.

Literature evidence and opinions of experts confirm that bisoprolol is effective in lowering mortality, hospitalization, and major cardiovascular events risk. In addition, it has minimal intrinsic sympathomimetic activity and high cardio-selectivity, which affords it safety over the long-term in patients with CVDs. These characteristics make bisoprolol a preferred BB in both HFrEF and angina in routine clinical practice scenarios. While current evidence is encouraging, more large-scale, prospective comparative studies are warranted to further validate bisoprolol’s role across diverse patient subgroups. Moreover, while bisoprolol remains a key component in the management of HFrEF, its integration with other pillars of GDMT through FDC offers a promising strategy to improve treatment adherence and streamline therapy. Further exploration and clinical evaluation of such combinations may help optimize patient outcomes.

Acknowledgments

All authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work, and have given final approval for the version to be published. The authors thank Dr. Sushama Patil, Mr. Rahul Iyer, and Dr. Amarnath Sugumaran from the Department of Medical Affairs, Cipla Ltd, Mumbai, for organizing and conducting the expert forum meetings.

Funding Statement

The medical writing and editorial support have been provided by Dr. Punit Srivastava and Dr. Nidhi Gupta of Mediception Science Pvt. Ltd (www.mediception.com) and were paid for by Cipla Ltd., India. The journal's open-access charges have been paid for by Cipla Ltd., India.

Disclosures

Conflicts of interest: In compliance with the ICMJE uniform disclosure form, all authors declare the following:

Payment/services info: The medical writing and editorial support have been provided by Dr. Punit Srivastava and Dr. Nidhi Gupta of Mediception Science Pvt. Ltd (www.mediception.com) and were paid for by Cipla Ltd., India. The journal's open-access charges have been paid for by Cipla Ltd., India.

Financial relationships: All authors have declared that they have no financial relationships at present or within the previous three years with any organizations that might have an interest in the submitted work.

Other relationships: All authors have declared that there are no other relationships or activities that could appear to have influenced the submitted work.

Author Contributions

Concept and design:  Kamal Sharma, Sunil Sathe, Bhupen Desai, Subhash Manchanda, Jagdish Mohan, Manish Bansal, Nagamalesh U. M., Abraham Oomman, Arindam Pande , Jay Shah, Johann Christopher, Sachin Patil, Jabir Abdullakutty, Akshay Bafna, Sarita Rao

Acquisition, analysis, or interpretation of data:  Kamal Sharma, Sunil Sathe, Bhupen Desai, Subhash Manchanda, Jagdish Mohan, Manish Bansal, Nagamalesh U. M., Abraham Oomman, Arindam Pande , Jay Shah, Johann Christopher, Sachin Patil, Jabir Abdullakutty, Akshay Bafna, Sarita Rao

Drafting of the manuscript:  Kamal Sharma, Sunil Sathe, Bhupen Desai, Subhash Manchanda, Jagdish Mohan, Manish Bansal, Nagamalesh U. M., Abraham Oomman, Arindam Pande , Jay Shah, Johann Christopher, Sachin Patil, Jabir Abdullakutty, Akshay Bafna, Sarita Rao

Critical review of the manuscript for important intellectual content:  Kamal Sharma, Sunil Sathe, Bhupen Desai, Subhash Manchanda, Jagdish Mohan, Manish Bansal, Nagamalesh U. M., Abraham Oomman, Arindam Pande , Jay Shah, Johann Christopher, Sachin Patil, Jabir Abdullakutty, Akshay Bafna, Sarita Rao

Supervision:  Kamal Sharma, Sunil Sathe, Bhupen Desai, Subhash Manchanda, Jagdish Mohan, Manish Bansal, Nagamalesh U. M., Abraham Oomman, Arindam Pande , Jay Shah, Johann Christopher, Sachin Patil, Jabir Abdullakutty, Akshay Bafna, Sarita Rao

References

  • 1.The cardiovascular disease continuum validated: clinical evidence of improved patient outcomes: part II: Clinical trial evidence (acute coronary syndromes through renal disease) and future directions. Dzau VJ, Antman EM, Black HR, et al. Circulation. 2006;114:2871–2891. doi: 10.1161/CIRCULATIONAHA.106.655761. [DOI] [PubMed] [Google Scholar]
  • 2.A new paradigm in the treatment of the cardiovascular disease continuum: focus on prevention. Chrysant SG. https://pmc.ncbi.nlm.nih.gov/articles/PMC3093150/ Hippokratia. 2011;15:7–11. [PMC free article] [PubMed] [Google Scholar]
  • 3.Global public health burden of heart failure: an updated review. Shahim B, Kapelios CJ, Savarese G, Lund LH. Card Fail Rev. 2023;9:0. doi: 10.15420/cfr.2023.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Five-year mortality and readmission rates in patients with heart failure in India: Results from the Trivandrum heart failure registry. Harikrishnan S, Jeemon P, Ganapathi S, et al. Int J Cardiol. 2021;326:139–143. doi: 10.1016/j.ijcard.2020.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.The Cardiology Society of India-Kerala Acute Heart Failure Registry: poor adherence to guideline-directed medical therapy. Joseph S, Panniyammakal J, Abdullakutty J, et al. Eur Heart J. 2021 doi: 10.1093/eurheartj/ehab793. [DOI] [PubMed] [Google Scholar]
  • 6.The natriuretic peptides system in the pathophysiology of heart failure: from molecular basis to treatment. Volpe M, Carnovali M, Mastromarino V. Clin Sci (Lond) 2016;130:57–77. doi: 10.1042/CS20150469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Role of bisoprolol in heart failure management: a consensus statement from India. Chopra HK, Nair T, Wander GS, et al. https://europepmc.org/article/med/38736057. J Assoc Physicians India. 2023;71:77–88. doi: 10.59556/japi.71.0426. [DOI] [PubMed] [Google Scholar]
  • 8.ACCF/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease: executive summary: a report of the American College of Cardiology Foundation/American Heart Association task force on practice guidelines, and the American College of Physicians, American Association for Thoracic Surgery, Preventive Cardiovascular Nurses Association, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. Fihn SD, Gardin JM, Abrams J, et al. Circulation. 2012;126:3097–3137. doi: 10.1161/CIR.0b013e3182776f83. [DOI] [PubMed] [Google Scholar]
  • 9.Heart rate in the pathophysiology of coronary blood flow and myocardial ischaemia: benefit from selective bradycardic agents. Heusch G. Br J Pharmacol. 2008;153:1589–1601. doi: 10.1038/sj.bjp.0707673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.The Real Role of β-Blockers in Daily Cardiovascular Therapy. Dézsi CA, Szentes V. Am J Cardiovasc Drugs. 2017;17:361–373. doi: 10.1007/s40256-017-0221-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.β-blockers are not all the same: pharmacologic similarities and differences, potential combinations and clinical implications. Taddei S, Tsabedze N, Tan RS. Curr Med Res Opin. 2024;40:15–23. doi: 10.1080/03007995.2024.2318058. [DOI] [PubMed] [Google Scholar]
  • 12.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. Heidenreich PA, Bozkurt B, Aguilar D, et al. Circulation. 2022;145:895–1032. doi: 10.1161/CIR.0000000000001063. [DOI] [PubMed] [Google Scholar]
  • 13.To evaluate the 'real world' clinical performance of bisoprolol in post-myocardial infarction with left ventricular dysfunction: tenacity study. Sharma K, Desai H, Sharm N, Laddha M, Hansora K, Vazirani P. https://europepmc.org/article/med/35443400 J Assoc Physicians India. 2022;70:11–12. [Google Scholar]
  • 14.Variability of bioavailability and intestinal absorption characteristics of bisoprolol. Ishida K, Horie A, Nishimura M, et al. Drug Metab Pharmacokinet. 2013;28:491–496. doi: 10.2133/dmpk.dmpk-13-rg-017. [DOI] [PubMed] [Google Scholar]
  • 15.Bazroon AA, Alrashidi NF. Vol. 19. StatPearls. StatPearls Publishing; 2024. Bisoprolol. In; p. 2024. [PubMed] [Google Scholar]
  • 16.Balanced pharmacokinetics and metabolism of bisoprolol. Leopold G. J Cardiovasc Pharmacol. 19868;11:16–20. doi: 10.1097/00005344-198511001-00003. [DOI] [PubMed] [Google Scholar]
  • 17.High beta 1-selectivity and favourable pharmacokinetics as the outstanding properties of bisoprolol. Haeusler G, Schliep HJ, Schelling P, et al. J Cardiovasc Pharmacol. 19868;11:2–15. doi: 10.1097/00005344-198511001-00002. [DOI] [PubMed] [Google Scholar]
  • 18.Beta-blockers: historical perspective and mechanisms of action. Oliver E, Mayor F Jr, D'Ocon P. Rev Esp Cardiol (Engl Ed) 2019;72:853–862. doi: 10.1016/j.rec.2019.04.006. [DOI] [PubMed] [Google Scholar]
  • 19.New advances in beta-blocker therapy in heart failure. Barrese V, Taglialatela M. Front Physiol. 2013;4 doi: 10.3389/fphys.2013.00323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2D6, ADRB1, ADRB2, ADRA2C, GRK4, and GRK5 genotypes and beta-blocker therapy. Duarte JD, Thomas CD, Lee CR, et al. Clin Pharmacol Ther. 2024;116:939–947. doi: 10.1002/cpt.3351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Effect of Beta - Blockers on Insulin Resistance in Patients with Hypertension and Metabolic Syndrome after 6 Months of Treatment. In. Sirenko Y. Journal of Endocrinology and Diabetes. Vol 4. 2017:1–11. [Google Scholar]
  • 22.Effects of beta-blockers on glucose and lipid metabolism. Fonseca VA. Curr Med Res Opin. 2010;26:615–629. doi: 10.1185/03007990903533681. [DOI] [PubMed] [Google Scholar]
  • 23.Use of beta blockers in the patient with diabetes. Bell DSH. https://journals.lww.com/theendocrinologist/Abstract/2003/03000/Use_of_Beta_Blockers_in_the_Patient_With_Diabetes.10.aspx The Endocrinologist. 2003;13:116. [Google Scholar]
  • 24.Advantages of a third-generation beta-blocker in patients with diabetes mellitus. Bell DSH. Am J Cardiol. 2004;93:49–52. doi: 10.1016/j.amjcard.2004.01.026. [DOI] [PubMed] [Google Scholar]
  • 25.β-Blockers, heart disease and COPD: current controversies and uncertainties. Baker JG, Wilcox RG. Thorax. 2017;72:271–276. doi: 10.1136/thoraxjnl-2016-208412. [DOI] [PubMed] [Google Scholar]
  • 26.Pharmacokinetic variability of beta-adrenergic blocking agents used in cardiology. Ågesen FN, Weeke PE, Tfelt-Hansen P, et al. Pharmacol Res Perspect. 2019;7:496. doi: 10.1002/prp2.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Beta blockers in older persons with heart failure: tolerability and impact on quality of life. Baxter AJ, Spensley A, Hildreth A, et al. Heart Br Card Soc. 2002;88:611–614. doi: 10.1136/heart.88.6.611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pharmacokinetics of bisoprolol during repeated oral administration to healthy volunteers and patients with kidney or liver disease. Kirch W, Rose I, Demers HG, Leopold G, Pabst J, Ohnhaus EE. Clin Pharmacokinet. 1987;13:110–117. doi: 10.2165/00003088-198713020-00003. [DOI] [PubMed] [Google Scholar]
  • 29.Role of β-blockers in the cardiovascular disease continuum: a collaborative Delphi survey-based consensus from Asia-Pacific. Ray S, Nair T, Sawhney J, et al. Curr Med Res Opin. 2023;39:1671–1683. doi: 10.1080/03007995.2023.2256218. [DOI] [PubMed] [Google Scholar]
  • 30.2023 focused update of the 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. McDonagh TA, Metra M, Adamo M, et al. Eur Heart J. 2023;44:3627–3639. doi: 10.1093/eurheartj/ehad195. [DOI] [PubMed] [Google Scholar]
  • 31.ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. McDonagh TA, Metra M, Adamo M, et al. Eur Heart J. 2021;42:3599–3726. doi: 10.1093/eurheartj/ehab853. [DOI] [PubMed] [Google Scholar]
  • 32.Effect of metoprolol CR/XL in chronic heart failure: metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF) https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(99)04440-2/abstract. Lancet. 1999;353:2001–2007. [PubMed] [Google Scholar]
  • 33.The Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) trial. Eichhorn EJ, Bristow MR. Curr Control Trials Cardiovasc Med. 2001;2:20–23. doi: 10.1186/cvm-2-1-020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, Shusterman NH. N Engl J Med. 1996;334:1349–1355. doi: 10.1056/NEJM199605233342101. [DOI] [PubMed] [Google Scholar]
  • 35.2021 update to the 2017 ACC expert consensus decision pathway for optimization of heart failure treatment: answers to 10 pivotal issues about heart failure with reduced ejection fraction: a report of the American College of Cardiology Solution Set Oversight Committee. Maddox TM, Januzzi JL Jr, Allen LA, et al. J Am Coll Cardiol. 2021;77:772–810. doi: 10.1016/j.jacc.2020.11.022. [DOI] [PubMed] [Google Scholar]
  • 36.Heart Failure Guidelines for India: update 2017. https://europepmc.org/article/med/38736057 2017;3:133–138. [Google Scholar]
  • 37.A randomized trial of beta-blockade in heart failure. The Cardiac Insufficiency Bisoprolol Study (CIBIS). CIBIS Investigators and Committees. Circulation. 1994;90:1765–1773. doi: 10.1161/01.cir.90.4.1765. [DOI] [PubMed] [Google Scholar]
  • 38.The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial: The. Lancet. 1999;353:9–13. [PubMed] [Google Scholar]
  • 39.Bisoprolol dose-response relationship in patients with congestive heart failure: a subgroup analysis in the cardiac insufficiency bisoprolol study(CIBIS II) Simon T, Mary-Krause M, Funck-Brentano C, et al. Eur Heart J. 2003;24:552–559. doi: 10.1016/s0195-668x(02)00743-1. [DOI] [PubMed] [Google Scholar]
  • 40.Results from post‐hoc analyses of the CIBIS II trial: effect of bisoprolol in high‐risk patient groups with chronic heart failure. Erland E, Philippe L, Patricia V, et al. Eur J Heart Fail. 2001;3:469–479. doi: 10.1016/s1388-9842(01)00174-x. [DOI] [PubMed] [Google Scholar]
  • 41.Effect on survival and hospitalization of initiating treatment for chronic heart failure with bisoprolol followed by enalapril, as compared with the opposite sequence: results of the randomized Cardiac Insufficiency Bisoprolol Study (CIBIS) III. Willenheimer R, van Veldhuisen DJ, Silke B, et al. Circulation. 2005;112:2426–2435. doi: 10.1161/CIRCULATIONAHA.105.582320. [DOI] [PubMed] [Google Scholar]
  • 42.Bisoprolol for the treatment of chronic heart failure: a meta-analysis on individual data of two placebo-controlled studies--CIBIS and CIBIS II. Cardiac Insufficiency Bisoprolol Study. Leizorovicz A, Lechat P, Cucherat M, Bugnard F. Am Heart J. 2002;143:301–307. doi: 10.1067/mhj.2002.120768. [DOI] [PubMed] [Google Scholar]
  • 43.Consensus and development of document for management of stabilized acute decompensated heart failure with reduced ejection fraction in India. Kaul U, Das MK, Agarwal R, et al. Indian Heart J. 2020;72:477–481. doi: 10.1016/j.ihj.2020.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.2013 ESC guidelines on the management of stable coronary artery disease: the Task Force on the management of stable coronary artery disease of the European Society of Cardiology. Montalescot G, Sechtem U, Achenbach S, et al. Eur Heart J. 2013;34:2949–3003. doi: 10.1093/eurheartj/eht296. [DOI] [PubMed] [Google Scholar]
  • 45.Long-term effectiveness of bisoprolol in patients with angina: a real-world evidence study. Sabidó M, Thilo H, Guido G. Pharmacol Res. 2019;139:106–112. doi: 10.1016/j.phrs.2018.10.031. [DOI] [PubMed] [Google Scholar]
  • 46.Bisoprolol in the treatment of angina pectoris: a double blind comparison with verapamil. De Divitiis O, Liguori V, Di Somma S, et al. Eur Heart J. 1987;8 Suppl M:43–54. doi: 10.1093/eurheartj/8.suppl_m.43. [DOI] [PubMed] [Google Scholar]
  • 47.Clinical evaluation of bisoprolol in patients with stable angina pectoris: a preliminary report. Kato K, Niitani H, Kanazawa T, Shimomura K, Toyama S. J Cardiovasc Pharmacol. 1986;8 Suppl 11:0–9. doi: 10.1097/00005344-198511001-00028. [DOI] [PubMed] [Google Scholar]
  • 48.Effect of bisoprolol on exercise tolerance in patients with coronary heart disease: placebo-controlled double-blind crossover study. Schnellbacher K, Marsovszky E, Roskamm H. J Cardiovasc Pharmacol. 1986;8 Suppl 11:0–7. doi: 10.1097/00005344-198511001-00026. [DOI] [PubMed] [Google Scholar]
  • 49.Comparison of the safety and efficacy of bisoprolol versus atenolol in stable exercise-induced angina pectoris: a Multicenter International Randomized Study of Angina Pectoris (MIRSA) de Muinck ED, Buchner-Moell D, van de Ven LL, Lie KI. J Cardiovasc Pharmacol. 1992;19:870–875. doi: 10.1097/00005344-199206000-00005. [DOI] [PubMed] [Google Scholar]
  • 50.A comparison of once daily bisoprolol, 5 and 10 mg, and atenolol 100 mg in the treatment of angina pectoris. Maltz MB, Dymond DS, Nathan AW, Camm AJ. Eur Heart J. 1987;8 Suppl M:37–42. doi: 10.1093/eurheartj/8.suppl_m.37. [DOI] [PubMed] [Google Scholar]
  • 51.Efficacy of once daily bisoprolol in stable angina pectoris: an objective comparison with atenolol and long term follow-up. Kohli RS, Khurmi NS, Kardash MM, Hughes LO, Lahiri A, Raftery EB. Eur Heart J. 1985;6:845–850. doi: 10.1093/oxfordjournals.eurheartj.a061771. [DOI] [PubMed] [Google Scholar]
  • 52.Which drug to choose for stable angina pectoris: a comparative study between bisoprolol and nitrates. Van De Ven LL, Vermeulen A, Tans JG, Tans AC, Liem KL, Lageweg NC, Lie KI. Int J Cardiol. 1995;47:217–223. doi: 10.1016/0167-5273(94)02194-n. [DOI] [PubMed] [Google Scholar]
  • 53.Medical treatment to reduce total ischemic burden: total ischemic burden bisoprolol study (TIBBS), a multicenter trial comparing bisoprolol and nifedipine. Von Arnim T. J Am Coll Cardiol. 1995;25:231–238. doi: 10.1016/0735-1097(94)00345-q. [DOI] [PubMed] [Google Scholar]
  • 54.Bisoprolol therapy reduces the number of angina attacks. Stojanov V, Marjanovic M, Radivojevic N, et al. J Hypertens. 2016;34:195. [Google Scholar]
  • 55.Bisoprolol improved endothelial function and myocardium survival of hypertension with stable angina: a randomized double-blinded trial. Lin ZP, Dong M, Liu J. http://chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/https://www.europeanreview.org/wp/wp-content/uploads/794-801.pdf. Eur Rev Med Pharmacol Sci. 2013;17:794–801. [PubMed] [Google Scholar]
  • 56.Terol I, Plaza L, Rodrigo F: Bisoprolol in the Treatment of Chronic Stable Angina Pectoris. https://pubmed.ncbi.nlm.nih.gov/11527131/ J Cardiovasc Pharmacol. 1990;16:208. [PubMed] [Google Scholar]
  • 57.ESC Guidelines for the management of chronic coronary syndromes. Vrints C, Andreotti F, Koskinas KC, et al. Eur Heart J. 2024;45:3415–3537. doi: 10.1093/eurheartj/ehae177. [DOI] [PubMed] [Google Scholar]
  • 58.Stable angina medical therapy management guidelines: a critical review of guidelines from the European Society of Cardiology and National Institute for Health and Care Excellence. Rousan TA, Thadani U. Eur Cardiol. 2019;14:18–22. doi: 10.15420/ecr.2018.26.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Canadian Cardiovascular Society guidelines for the diagnosis and management of stable ischemic heart disease. Mancini GB, Gosselin G, Chow B, et al. Can J Cardiol. 2014;30:837–849. doi: 10.1016/j.cjca.2014.05.013. [DOI] [PubMed] [Google Scholar]
  • 60.Management of chronic stable angina with once-daily bisoprolol or atenolol and long-term efficacy of bisoprolol. Kohli RS, Lahiri A, Raftery EB. J Cardiovasc Pharmacol. 1986;8 Suppl 11:0–53. doi: 10.1097/00005344-198511001-00027. [DOI] [PubMed] [Google Scholar]
  • 61.Heart Failure in CKD Population. Madarasu RC, Marri HP. Indian J Clin Cardiol. 2024;5:84–94. [Google Scholar]
  • 62.Effects of Beta-Adrenergic Antagonists in Patients With Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Badve SV, Roberts MA, Hawley CM, et al. J Am Coll Cardiol. 2011;58:1152–1161. doi: 10.1016/j.jacc.2011.04.041. [DOI] [PubMed] [Google Scholar]
  • 63.Impact of Renal Impairment on Beta-Blocker Efficacy in Patients With Heart Failure. Kotecha D, Gill SK, Flather MD, et al. J Am Coll Cardiol. 2019;74:2893–2904. doi: 10.1016/j.jacc.2019.09.059. [DOI] [PubMed] [Google Scholar]
  • 64.The single dose pharmacokinetics of bisoprolol (10 mg) in renal insufficiency: the clinical significance of balanced clearance. Payton CD, Fox JG, Pauleau NF, Boulton-Jones JM, Ioannides C, Johnston A, Thomas P. Eur Heart J. 1987;8 Suppl M:15–22. doi: 10.1093/eurheartj/8.suppl_m.15. [DOI] [PubMed] [Google Scholar]
  • 65.Drug-drug interactions of beta-adrenoceptor blockers. Brodde OE, Kroemer HK. Arzneimittelforschung. 2003;53:814–822. doi: 10.1055/s-0031-1299835. [DOI] [PubMed] [Google Scholar]
  • 66.β-Blocker Dialyzability in Maintenance Hemodialysis Patients. Tieu A, Velenosi TJ, Kucey AS, et al. Clin J Am Soc Nephrol CJASN. 2018;13:604–611. doi: 10.2215/CJN.07470717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.β-blockers in hemodialysis: simple questions, complicated answers. Hundemer GL, Sood MM, Canney M. Clin Kidney J. 2021;14:731–734. doi: 10.1093/ckj/sfaa249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.β-Blocker Dialyzability and Mortality in Older Patients Receiving Hemodialysis. Weir MA, Dixon SN, Fleet JL, et al. J Am Soc Nephrol. 2015;26:987–996. doi: 10.1681/ASN.2014040324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cardio-selective versus non-selective β-blockers for cardiovascular events and mortality in long-term dialysis patients: A systematic review and meta-analysis. PLOS ONE. Tao S, Huang J, Xiao J, et al. 2022;17:279171. doi: 10.1371/journal.pone.0279171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Comparative effectiveness of bisoprolol and carvedilol among patients receiving maintenance hemodialysis. Wu PH, Lin YT, Liu JS, et al. Clin Kidney J. 2021;14:983–990. doi: 10.1093/ckj/sfaa248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liver abnormalities in cardiac diseases and heart failure. Alvarez AM, Mukherjee D. Int J Angiol. 2011;20:135–142. doi: 10.1055/s-0031-1284434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Carvedilol is associated with improved survival in patients with cirrhosis: a long-term follow-up study. McDowall HR, Chuah CS, Tripathi D, et al. Aliment Pharmacol Ther. 2021;53:531–539. doi: 10.1111/apt.16189. [DOI] [PubMed] [Google Scholar]
  • 73.Carvedilol as the new non-selective beta-blocker of choice in patients with cirrhosis and portal hypertension. Turco L, Reiberger T, Vitale G, et al. Liver Int Off J Int Assoc Study Liver. 2023;43:1183–1194. doi: 10.1111/liv.15559. [DOI] [PubMed] [Google Scholar]
  • 74.Selection of β‐Blocker in Patients With Cirrhosis and Acute Myocardial Infarction: A 13‐Year Nationwide Population‐Based Study in Asia. Wu VC, Chen S, Ting P, et al. J Am Heart Assoc. 2018;7:8982. doi: 10.1161/JAHA.118.008982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cardio-Selective Beta-Blocker: Pharmacological Evidence and Their Influence on Exercise Capacity. Ladage D, Schwinger RHG, Brixius K. Cardiovasc Ther. 2013;31:76–83. doi: 10.1111/j.1755-5922.2011.00306.x. [DOI] [PubMed] [Google Scholar]
  • 76.Efficacy and safety of bisoprolol in young men with arterial hypertension and obesity. Avtandilov AG, Esenova II, Petrosov SL. Ration Pharmacother Cardiol. 2016;7:193–198. [Google Scholar]
  • 77.Efficacy and safety of bisoprolol compared to other selective beta-1 blockers in the treatment of hypertension: a systematic review and meta-analysis of randomized parallel clinical trials. Jadhav U, Nair T, Bansal S, et al. Eur Heart J. 2021;42:724–2363. [Google Scholar]
  • 78.Beta-blockers used in cardiac failure and blood glucose level impairment - a literature review. Fularska K, Oleszko M, Wąsiewicz E, et al. J Educ Health Sport. 2023;23:40–51. [Google Scholar]
  • 79.Influence of bisoprolol on blood glucose, glucosuria, and haemoglobin A1 in noninsulin-dependent diabetics. Janka HU, Ziegler AG, Disselhoff G, et al. J Cardiovasc Pharmacol. 19868;11:96–99. doi: 10.1097/00005344-198511001-00018. [DOI] [PubMed] [Google Scholar]
  • 80.The role of beta-blockers in the management of chronic obstructive pulmonary disease. Leitao Filho FS, Alotaibi NM, Yamasaki K, et al. Expert Rev Respir Med. 2018;12:125–135. doi: 10.1080/17476348.2018.1419869. [DOI] [PubMed] [Google Scholar]
  • 81.Beta-blocker therapy in patients with COPD: a systematic literature review and meta-analysis with multiple treatment comparison. Gulea C, Zakeri R, Alderman V, et al. Respir Res. 2021;22:64. doi: 10.1186/s12931-021-01661-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Association of β-blocker use with survival and pulmonary function in patients with chronic obstructive pulmonary and cardiovascular disease: a systematic review and meta-analysis. Yang YL, Xiang ZJ, Yang JH, et al. Eur Heart J. 2020;41:4415–4422. doi: 10.1093/eurheartj/ehaa793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Commencement of cardioselective beta‐blockers during hospitalisation for acute exacerbations of chronic obstructive pulmonary disease. Neef PA, Burrell LM, McDonald CF, et al. Intern Med J. 2017;47:1043–1050. doi: 10.1111/imj.13518. [DOI] [PubMed] [Google Scholar]
  • 84.Beta-blockers use in patients with chronic obstructive pulmonary disease and concomitant cardiovascular conditions. Albouaini K, Andron M, Alahmar A, et al. https://www.tandfonline.com/doi/full/10.2147/copd.s12159987. Int J Chron Obstruct Pulmon Dis. 2007;2:535–540. [PMC free article] [PubMed] [Google Scholar]
  • 85.Carvedilol, bisoprolol, and metoprolol use in patients with coexistent heart failure and chronic obstructive pulmonary disease. Su VY, Chang YS, Hu YW, et al. Medicine (Baltimore) 2016;95:0. doi: 10.1097/MD.0000000000002427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.The evaluation of β-adrenoceptor blocking agents in patients with COPD and congestive heart failure: a nationwide study. Liao KM, Lin TY, Huang YB, Kuo CC, Chen CY. Int J Chron Obstruct Pulmon Dis. 2017;12:2573–2581. doi: 10.2147/COPD.S141694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Impact of β-blocker selectivity on long-term outcomes in congestive heart failure patients with chronic obstructive pulmonary disease. Kubota Y, Asai K, Furuse E, Nakamura S, Murai K, Tsukada YT, Shimizu W. Int J Chron Obstruct Pulmon Dis. 2015;10:515–523. doi: 10.2147/COPD.S79942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Differences between bisoprolol and carvedilol in patients with chronic heart failure and chronic obstructive pulmonary disease: a randomized trial. Lainscak M, Podbregar M, Kovacic D, Rozman J, von Haehling S. Respir Med. 2011;105 Suppl 1:0–9. doi: 10.1016/S0954-6111(11)70010-5. [DOI] [PubMed] [Google Scholar]
  • 89.Differences between beta-blockers in patients with chronic heart failure and chronic obstructive pulmonary disease: a randomized crossover trial. Jabbour A, Macdonald PS, Keogh AM, et al. J Am Coll Cardiol. 2010;55:1780–1787. doi: 10.1016/j.jacc.2010.01.024. [DOI] [PubMed] [Google Scholar]
  • 90.Efficacy and safety of bisoprolol in patients with chronic obstructive pulmonary disease: a systematic review and meta-analysis. Feng Z, Zhang L, Wang Y, Guo H, Liu J. Int J Chron Obstruct Pulmon Dis. 2023;18:3067–3083. doi: 10.2147/COPD.S438930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Effects of single oral doses of bisoprolol and atenolol on airway function in nonasthmatic chronic obstructive lung disease and angina pectoris. Dorow P, Bethge H, Tönnesmann U. Eur J Clin Pharmacol. 1986;31:143–147. doi: 10.1007/BF00606650. [DOI] [PubMed] [Google Scholar]
  • 92.Cardioselective beta-blockers in patients with reactive airway disease: a meta-analysis. Salpeter SR, Ormiston TM, Salpeter EE. Ann Intern Med. 2002;137:715–725. doi: 10.7326/0003-4819-137-9-200211050-00035. [DOI] [PubMed] [Google Scholar]
  • 93.Do beta-adrenergic blocking agents increase asthma exacerbation? A network meta-analysis of randomized controlled trials. Huang KY, Tseng PT, Wu YC, et al. Sci Rep. 2021;11:452. doi: 10.1038/s41598-020-79837-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.The possibilities of using beta-blocker bisoprolol in patients with stable angina with concomitant bronchial asthma. Grigorieva NY, Ilushina TP, Kolosova KS. https://cardio.elpub.ru/jour/article/view/1714/0?locale=en_US. Kardiologiia. 2022;62:32–39. doi: 10.18087/cardio.2022.1.n1714. [DOI] [PubMed] [Google Scholar]

Articles from Cureus are provided here courtesy of Cureus Inc.

RESOURCES