All experts involved in the development of these guidelines have submitted declarations of interest. These have been compiled in a report and simultaneously published in a supplementary document to the guidelines. The report is also available on the ESC website www.escardio.org/Guidelines

graphic See the European Heart Journal online for supplementary data that include background information and detailed discussion of the data that have provided the basis of the guidelines.

graphic Click here to access the corresponding ESC CardioMed chapters.

Table of contents

  • 1. Preamble 3832

  • 2. Introduction 3834

  •  2.1. What is new 3834

  •  2.2. The magnitude of the problem 3839

  •  2.3. Change in demographics 3840

  •  2.4. Purpose 3840

  •  2.5. The outcomes we want to prevent 3841

  • 3. Clinical risk evaluation 3841

  •  3.1. Surgery-related risk 3841

  •   3.1.1. Timing of surgery 3842

  •  3.2. Type of surgical approach 3842

  •   3.2.1. Laparoscopy 3842

  •    3.2.1.1. Vascular and endovascular procedures 3843

  •    3.2.1.2. Video-assisted non-cardiac surgery 3843

  •  3.3. Patient-related risk 3843

  •   3.3.1. Initial assessment 3843

  •    3.3.1.1. Patients aged <65 years without a history of cardiovascular disease or cardiovascular risk factors 3843

  •    3.3.1.2. Patients aged ≥65 years or with cardiovascular risk factors 3843

  •    3.3.1.3. Patients with established cardiovascular disease 3844

  •   3.3.2. Patients with murmurs, chest pain, dyspnoea, or peripheral oedema 3845

  •    3.3.2.1. Murmurs 3845

  •    3.3.2.2. Chest pain 3845

  •    3.3.2.3. Dyspnoea 3845

  •    3.3.2.4. Peripheral oedema 3845

  •  3.4. Timing of adequate risk evaluation 3846

  •  3.5. Avoidance or allowance for surgery in the individual patient 3846

  •  3.6. The patient perspective 3846

  • 4. Pre-operative assessment tools 3847

  •  4.1. Risk scores 3847

  •   4.1.1. General risk calculators 3847

  •   4.1.2. Frailty 3849

  •  4.2. Functional capacity 3849

  •  4.3. Electrocardiography 3850

  •  4.4. Biomarkers 3850

  •  4.5. Non-invasive and invasive procedures 3851

  •   4.5.1. Resting transthoracic echocardiography 3851

  •   4.5.2. Stress tests 3852

  •    4.5.2.1. Exercise stress test 3852

  •    4.5.2.2. Stress imaging 3852

  •   4.5.3. Angiography 3853

  •    4.5.3.1. Coronary computed tomography angiography 3853

  •    4.5.3.2. Invasive coronary angiography 3853

  • 5. General risk-reduction strategies 3854

  •  5.1. Cardiovascular risk factors and lifestyle interventions 3854

  •  5.2. Pharmacological 3854

  •   5.2.1. Beta-blockers 3854

  •   5.2.2. Amiodarone 3855

  •   5.2.3. Statins 3855

  •   5.2.4. Renin–angiotensin–aldosterone system inhibitors 3855

  •   5.2.5. Calcium channel blockers 3855

  •   5.2.6. Alpha-2 receptor agonists 3856

  •   5.2.7. Diuretics 3856

  •   5.2.8. Ivabradine 3856

  •   5.2.9. Sodium–glucose co-transporter-2 inhibitors 3856

  •  5.3. Peri-operative handling of antithrombotic agents 3857

  •   5.3.1. Antiplatelets 3857

  •    5.3.1.1. Single antiplatelet therapy 3857

  •    5.3.1.2. Dual antiplatelet therapy 3860

  •    5.3.1.3. De-escalation of antiplatelet effect 3862

  •    5.3.1.4. Platelet function guided peri-operative management of antiplatelet therapy 3862

  •   5.3.2. Oral anticoagulants 3863

  •    5.3.2.1. Vitamin K antagonists 3863

  • 5.3.2.1.1. Vitamin K antagonists in patients with mechanical heart valves 3863

  • 5.3.2.1.2. Vitamin K antagonists for atrial fibrillation/venous thromboembolism 3864

  • 5.3.2.1.3. Restarting vitamin K antagonists after invasive procedures or surgery 3864

  • 5.3.2.1.4. Reversal of vitamin K antagonists 3864

  •    5.3.2.2. Non-vitamin K antagonist oral anticoagulants 3864

  • 5.3.2.2.1. Unplanned surgery in patients on non-vitamin K antagonist oral anticoagulants and reversal for emergency procedures 3864

  • 5.3.2.2.2. Planned interventions in patients on non-vitamin K oral anticoagulants 3866

  • 5.3.2.2.3. Bridging 3866

  • 5.3.2.2.4. Laboratory testing before surgery 3866

  • 5.3.2.2.5. Considerations for specific procedures 3867

  • 5.3.2.2.6. When to restart non-vitamin K antagonist oral anticoagulants after interventions 3868

  •    5.3.2.3. Combination therapy (antiplatelet and anticoagulant) 3868

  •  5.4. Peri-operative thromboprophylaxis 3869

  •  5.5. Patient blood management 3869

  •   5.5.1. Pre-operative anaemia—diagnosis and treatment 3870

  •   5.5.2. Bleeding and reduction of iatrogenic diagnostic/surgery-related blood loss 3870

  •   5.5.3. Optimal blood component use with patient-centred clinical decision support 3871

  • 6. Specific diseases 3871

  •  6.1. Coronary artery disease 3871

  •   6.1.1. Risk for patients with coronary artery disease 3871

  •   6.1.2. Pre-operative risk assessment and management 3872

  •   6.1.3. Revascularization strategies 3872

  •    6.1.3.1. Chronic coronary syndromes 3872

  •    6.1.3.2. Acute coronary syndromes 3872

  •  6.2. Chronic heart failure 3874

  •   6.2.1. Risk for patients with heart failure 3874

  •   6.2.2. Pre- and post-operative management strategies 3874

  •   6.2.3. Hypertrophic obstructive cardiomyopathy 3875

  •   6.2.4. Patients with ventricular assist devices undergoing non-cardiac surgery 3875

  •  6.3. Valvular heart disease 3875

  •   6.3.1. Risk for patients with valvular heart disease 3875

  •   6.3.2. Pre-operative management strategies and risk-reduction strategy 3876

  •    6.3.2.1. Aortic valve stenosis 3876

  •    6.3.2.2. Mitral valve stenosis 3877

  •    6.3.2.3. Aortic valve regurgitation 3878

  •    6.3.2.4. Mitral valve regurgitation 3878

  •    6.3.2.5. Patients with prosthetic valve(s) 3878

  •    6.3.2.6. Prophylaxis of infective endocarditis 3878

  •  6.4. Known or newly diagnosed arrhythmias 3879

  •   6.4.1. Peri-operative management—general measures 3879

  •   6.4.2. Supraventricular arrhythmias 3879

  •   6.4.3. Atrial fibrillation/flutter 3879

  •   6.4.4. Ventricular arrhythmias 3879

  •   6.4.5. Bradyarrhythmias 3881

  •   6.4.6. Management of patients with cardiac implantable electronic devices 3881

  •  6.5. Adult congenital heart disease 3882

  •  6.6. Pericardial diseases 3883

  •  6.7. Pulmonary disease and pulmonary arterial hypertension 3884

  •   6.7.1. Pulmonary disease 3884

  •   6.7.2. Pulmonary arterial hypertension 3884

  •  6.8. Arterial hypertension 3885

  •  6.9. Peripheral artery disease 3886

  •   6.9.1. Peripheral artery disease and non-vascular non-cardiac surgery 3886

  •   6.9.2. Peripheral artery disease and vascular non-cardiac surgery 3886

  •  6.10. Cerebrovascular disease 3887

  •  6.11. Renal disease 3887

  •  6.12. Obesity 3888

  •  6.13. Diabetes 3889

  •  6.14. Cancer 3889

  •  6.15. Coronavirus disease 2019 2019

  • 7. Peri-operative monitoring and anaesthesia 3890

  •  7.1. Peri-operative monitoring 3890

  •  7.2. Anaesthesia 3891

  •   7.2.1. Intra-operative haemodynamics 3891

  •   7.2.2. Choice of anaesthetic agent 3892

  •  7.3. Locoregional techniques 3892

  •  7.4. Peri-operative goal-directed haemodynamic therapy 3893

  •  7.5. Post-operative management 3893

  • 8. Peri-operative cardiovascular complications 3893

  •  8.1. Peri-operative myocardial infarction/injury 3894

  •  8.2. Spontaneous myocardial infarction (after day 2) 3897

  •  8.3. Takotsubo syndrome 3897

  •  8.4. Acute heart failure 3897

  •  8.5. Venous thromboembolism 3897

  •  8.6. Atrial fibrillation and other relevant arrhythmias 3897

  •   8.6.1. Prevention of post-operative atrial fibrillation 3897

  •   8.6.2. Management of post-operative atrial fibrillation 3898

  •    8.6.2.1. Rate and/or rhythm control 3898

  •    8.6.2.2. Prevention of atrial fibrillation-related thromboembolic complications 3899

  •  8.7. Peri-operative stroke 3899

  • 9. Key messages 3900

  • 10. Gaps in evidence 3900

  • 11. Sex differences 3901

  • 12. ‘What to do’ and ‘what not to do’ messages from the Guidelines 3901

  • 13. Quality indicators 3906

  • 14. Central illustration 3906

  • 15. Supplementary data 3907

  • 16. Data availability statement 3907

  • 17. Author information 3907

  • 18. Appendix 3907

  • 19. References 3908

Tables of Recommendations

  • Recommendation Table 1 — Recommendations for selection of surgical approach and impact on risk 3843

  • Recommendation Table 2 — Recommendations for all patients scheduled for non-cardiac surgery 3845

  • Recommendation Table 3 — Recommendations for patients aged <65 years without signs, symptoms, or history of cardiovascular disease 3845

  • Recommendation Table 4 — Recommendations for pre-operative assessment in patients with previously unknown murmur, angina, dyspnoea, or peripheral oedema 3845

  • Recommendation Table 5 — Recommendations for patient information 3847

  • Recommendation Table 6 — Recommendations for pre-operative assessment of frailty and functional capacity 3849

  • Recommendation Table 7 — Recommendations for pre-operative risk assessment—electrocardiography and biomarkers 3851

  • Recommendation Table 8 — Recommendations for transthoracic echocardiography 3852

  • Recommendation Table 9 — Recommendations for stress imaging  3853

  • Recommendation Table 10 — Recommendations for coronary angiography 3853

  • Recommendation Table 11 — Recommendations for lifestyle and cardiovascular risk factors 3854

  • Recommendation Table 12 — Recommendations for pharmacological treatment 3856

  • Recommendation Table 13 — Recommendations for use of antiplatelet therapy in patients undergoing non-cardiac surgery 3862

  • Recommendation Table 14 — Recommendations for interruption and resumption of anticoagulants in patients undergoing non-cardiac surgery 3868

  • Recommendation Table 15 — Recommendations for thromboprophylaxis 3869

  • Recommendation Table 16 — Recommendations for intra- and post-operative complications associated with anaemia 3870

  • Recommendation Table 17 — Recommendations for intra- and post-operative complications associated with blood loss 3871

  • Recommendation Table 18 — Recommendations for intra- and post-operative complications associated with allogeneic blood transfusion 3871

  • Recommendation Table 19 — Recommendations for the timing of non-cardiac surgery and revascularization in patients with known coronary artery disease 3874

  • Recommendation Table 20 — Recommendations for management of heart failure in patients undergoing non-cardiac surgery 3875

  • Recommendation Table 21 — Recommendations for management of valvular heart disease in patients undergoing non-cardiac surgery 3878

  • Recommendation Table 22 — Recommendations for management of known or newly diagnosed arrhythmias 3880

  • Recommendation Table 23 — Recommendations for management of bradyarrhythmia and patients carrying cardiac implantable devices 3882

  • Recommendation Table 24 — Recommendations for management of patients with adult congenital heart disease undergoing non-cardiac surgery 3883

  • Recommendation Table 25 — Recommendations for pericardial diseases 3884

  • Recommendation Table 26 — Recommendations for patients with pulmonary arterial hypertension undergoing non-cardiac surgery 3885

  • Recommendation Table 27 — Recommendations for pre-operative management of hypertension 3886

  • Recommendation Table 28 — Recommendations for management of patients with peripheral artery disease and/or abdominal aortic aneurysm undergoing non-cardiac surgery 3887

  • Recommendation Table 29 — Recommendations for management of patients with suspected or established carotid artery disease undergoing non-cardiac surgery 3887

  • Recommendation Table 30 — Recommendations for management of patients with renal disease undergoing non-cardiac surgery 3888

  • Recommendation Table 31 — Recommendations for management of patients with obesity undergoing non-cardiac surgery 3888

  • Recommendation Table 32 — Recommendations for management of patients with diabetes mellitus undergoing non-cardiac surgery 3889

  • Recommendation Table 33 — Recommendations for peri-operative monitoring and anaesthesia 3893

  • Recommendation Table 34 — Recommendations for peri-operative cardiovascular complications 3899

List of tables

  • Table 1 Classes of recommendations 3833

  • Table 2 Levels of evidence 3833

  • Table 3 New concepts and sections in the current guidelines 3834

  • Table 4 What is new 3834

  • Table 4A New recommendations 3834

  • Table 4B Revised recommendations 3838

  • Table 5 Surgical risk estimate according to type of surgery or intervention 3842

  • Table 6 Risk score calculators 3848

  • Table 7 Pharmacokinetic and pharmacodynamic characteristics of antiplatelets 3857

  • Table 8 Pharmacokinetic and pharmacodynamic characteristics of oral anticoagulants 3858

  • Table 9 Bleeding risk according to type of non-cardiac surgery 3858

  • Table 10 Laboratory parameters for the diagnosis of absolute iron-deficiency anaemia 3870

  • Table 11 Peri-operative approach to patients with ventricular assist devices undergoing non-cardiac surgery 3875

  • Table 12 Peri-operative management of patients with arrhythmias  3880

  • Table 13 Risk stratification for non-cardiac surgery in adults with congenital heart disease 3883

  • Table 14 Patient-related and surgery-related factors to be considered when assessing peri-operative risk in patients with pulmonary arterial hypertension 3885

  • Table 15 Factors that could influence peri-operative risk during cancer surgery and preventive strategies 3890

List of figures

  • Figure 1 Total risk is an interaction of patient-related and surgery-related risk 3841

  • Figure 2 Pre-operative assessment before non-cardiac surgery 3844

  • Figure 3 Examples of questions and concerns expressed by patients 3847

  • Figure 4 Recommended measurements to assess and detect the risk of post-operative cardiac complications 3850

  • Figure 5 Recommendations for management of antiplatelet therapy in patients undergoing non-cardiac surgery 3859

  • Figure 6 P2Y12 inhibitor interruption after percutaneous coronary intervention before elective non-cardiac surgery 3860

  • Figure 7 Bridging with intravenous antiplatelet agents. ASA, acetylsalicylic acid; FU, follow-up; LD, loading dose; NCS, non-cardiac surgery; o.d., once a day 3861

  • Figure 8 Recommendations for management of oral anticoagulation therapy in patients undergoing non-cardiac surgery 3863

  • Figure 9 Peri-operative management of non-vitamin K antagonist oral anticoagulant according to the periprocedural risk of bleeding  3865

  • Figure 10 Timing of last non-vitamin K antagonist oral anticoagulant dose before elective NCS according to renal function 3866

  • Figure 11 Suggested strategy for potential reversal of non-vitamin K oral anticoagulants effect 3867

  • Figure 12 Management of patients with acute or chronic coronary syndrome scheduled for non-cardiac surgery 3873

  • Figure 13 Management of patients with severe aortic valve stenosis scheduled for non-cardiac surgery 3876

  • Figure 14 Management of patients with secondary mitral valve regurgitation scheduled for non-cardiac surgery 3877

  • Figure 15 Optimal location of return electrode during unipolar electrosurgery in patients with cardiac implantable electronic devices, depending on the surgery site 3882

  • Figure 16 Pathophysiological approach to address intra-operative hypotension 3892

  • Figure 17 Factors associated with peri-operative cardiovascular complications. SNS, sympathetic nervous system 3894

  • Figure 18 Differential diagnosis of elevated post-operative cardiac troponin concentrations 3895

  • Figure 19 Systematic work-up (aetiology) and therapy of peri-operative myocardial infarction/injury 3896

  • Figure 20 Prevention and management of post-operative atrial fibrillation 3898

  • Figure 21 Central illustration: the complex interplay between the intrinsic risk of surgery and the patient risk of peri-operative cardiovascular complications 3906

Abbreviations and acronyms

     
  • AAA

    Abdominal aortic aneurysm

  •  
  • AAD

    Antiarrhythmic drug

  •  
  • ACEI

    Angiotensin-converting-enzyme inhibitor

  •  
  • ACHD

    Adults with congenital heart disease

  •  
  • ACS

    Acute coronary syndrome

  •  
  • ACS NSQIP

    American College of Surgery National Surgical Quality Improvement Program

  •  
  • AF

    Atrial fibrillation

  •  
  • AKI

    Acute kidney injury

  •  
  • aPTT

    Activated partial thromboplastin time

  •  
  • AR

    Aortic valve regurgitation

  •  
  • ARB

    Angiotensin receptor blocker

  •  
  • ARNI

    Angiotensin receptor neprilysin inhibitor

  •  
  • AS

    Aortic valve stenosis

  •  
  • ASA

    Acetylsalicylic acid

  •  
  • ASA–PS

    American Society of Anesthesiology Physical Status

  •  
  • ASCVD

    Atherosclerotic cardiovascular disease

  •  
  • AUB-HAS2

    American University of Beirut (AUB)-HAS2

  •  
  • AUC

    Area under curve

  •  
  • AVR

    Aortic valve replacement

  •  
  • BAV

    Balloon aortic valvuloplasty

  •  
  • BCSH

    British Committee for Standards in Haematology

  •  
  • b.i.d.

    Bis in die (twice a day)

  •  
  • BTKi

    Bruton tyrosine kinase inhibitors

  •  
  • BMI

    Body mass index

  •  
  • BMS

    Bare metal stent

  •  
  • BNP

    B-type natriuretic peptide

  •  
  • BP

    Blood pressure

  •  
  • b.p.m.

    Beats per minute

  •  
  • BSA

    Body surface area

  •  
  • CABG

    Coronary artery bypass graft

  •  
  • CAD

    Coronary artery disease

  •  
  • CARP

    Coronary Artery Revascularization Prophylaxis (trial)

  •  
  • CAS

    Carotid artery stenting

  •  
  • CASS

    Coronary Artery Surgery Study

  •  
  • CCB

    Calcium channel blocker

  •  
  • CCS

    Chronic coronary syndrome

  •  
  • CCTA

    Coronary computed tomography angiography

  •  
  • CEA

    Carotid endarterectomy

  •  
  • CHA2DS2-VASc

    Congestive heart failure, hypertension, age ≥75 years, diabetes mellitus, stroke, vascular disease, age 65–74 years, sex category (female)

  •  
  • CI

    Confidence interval

  •  
  • CIED

    Cardiac implantable electronic device

  •  
  • CK

    Creatinine kinase

  •  
  • CKD

    Chronic kidney disease

  •  
  • CKD-EPI

    Chronic Kidney Disease Epidemiology Collaboration

  •  
  • Cmax

    Maximum serum concentration

  •  
  • CMR

    Cardiac magnetic resonance

  •  
  • COAPT

    Cardiovascular Outcomes Assessment of the MitraClip Percutaneous Therapy for Heart Failure Patients with Functional Mitral Regurgitation (trial)

  •  
  • COPD

    Chronic obstructive pulmonary disease

  •  
  • CORIDA

    Per-procedural Concentration of Direct Oral Anticoagulants (trial)

  •  
  • Coronary CTA VISION

    Coronary Computed Tomographic Angiography and Vascular Events in Noncardiac Surgery Patients Cohort Evaluation (trial)

  •  
  • COVID-19

    Coronavirus disease 2019

  •  
  • CPET

    Cardiopulmonary exercise testing

  •  
  • CRF

    Cardiorespiratory fitness

  •  
  • CRT

    Cardiac resynchronization therapy

  •  
  • CT

    Computed tomography

  •  
  • cTn T/I

    Cardiac troponin T/I

  •  
  • CTO

    Chronic total occlusion

  •  
  • CV

    Cardiovascular

  •  
  • CVD

    Cardiovascular disease

  •  
  • DAPT

    Dual antiplatelet therapy

  •  
  • DASI

    Duke Activity Status Index

  •  
  • DES

    Drug-eluting stent

  •  
  • DM

    Diabetes mellitus

  •  
  • DSE

    Dobutamine stress echocardiography

  •  
  • dTT

    Diluted thrombin time

  •  
  • EACTS

    European Association for Cardio-Thoracic Surgery

  •  
  • ECG

    Electrocardiographic/electrocardiogram

  •  
  • EDKA

    Euglycaemic diabetic ketoacidosis

  •  
  • eGFR

    Estimated glomerular filtration rate

  •  
  • EMI

    Electromagnetic interference

  •  
  • EORP

    EURObservational Research Programme

  •  
  • ESA

    European Society of Anaesthesiology

  •  
  • ESC

    European Society of Cardiology

  •  
  • ESH

    European Society of Hypertension

  •  
  • ESTS

    European Society of Thoracic Surgeons

  •  
  • ESVS

    European Society for Vascular Surgery

  •  
  • EuSOS

    European Surgical Outcomes Study

  •  
  • EVAR

    Endovascular abdominal aortic aneurysm repair

  •  
  • FDA

    US Food and Drug Administration

  •  
  • FFR

    Fractional flow reserve

  •  
  • FIIa

    Factor IIa

  •  
  • FOCUS

    Focused cardiac ultrasound

  •  
  • FXa

    Factor Xa

  •  
  • GDMT

    Guideline-directed medical therapy

  •  
  • GFR

    Glomerular filtration rate

  •  
  • HbA1c

    Glycated haemoglobin

  •  
  • HF

    Heart failure

  •  
  • HIP-ATTACK

    HIP Fracture Accelerated Surgical TreaTment And Care tracK (trial)

  •  
  • HR

    Hazard ratio

  •  
  • hs-cTn

    High-sensitivity cardiac troponin

  •  
  • i.v.

    Intravenous

  •  
  • ICA

    Invasive coronary angiography

  •  
  • ICD

    Implantable cardioverter–defibrillator

  •  
  • ICU

    Intensive care unit

  •  
  • ID

    Iron deficiency

  •  
  • IHD

    Ischaemic heart disease

  •  
  • INR

    International normalized ratio

  •  
  • ISCHEMIA

    International Study of Comparative Health Effectiveness with Medical and Invasive Approaches (trial)

  •  
  • iwFR

    Instantaneous wave-free ratio

  •  
  • KDIGO

    Kidney Disease: Improving Global Outcomes

  •  
  • LD

    Loading dose

  •  
  • LMWH

    Low molecular weight heparin

  •  
  • LOAD

    Lowering the Risk of Operative Complications Using Atorvastatin Loading Dose (trial)

  •  
  • LoE

    Level of evidence

  •  
  • LV

    Left ventricular

  •  
  • LVEF

    Left ventricular ejection fraction

  •  
  • LVESD

    Left ventricular end-systolic diameter

  •  
  • LVESDi

    Left ventricular end-systolic dimension index

  •  
  • MACE

    Major adverse cardiovascular event

  •  
  • MET

    Metabolic equivalent

  •  
  • METS

    Measurement of Exercise Tolerance before Surgery (trial)

  •  
  • MHV

    Mechanical heart valve

  •  
  • MI

    Myocardial infarction

  •  
  • MINS

    Myocardial injury following non-cardiac surgery

  •  
  • MR

    Mitral valve regurgitation

  •  
  • MS

    Mitral valve stenosis

  •  
  • NCS

    Non-cardiac surgery

  •  
  • NOAC

    Non-vitamin K antagonist oral anticoagulant

  •  
  • NSAID

    Non-steroidal anti-inflammatory drug

  •  
  • NSTE-ACS

    Non-ST-segment elevation acute coronary syndrome

  •  
  • NT-proBNP

    N-terminal pro-B-type natriuretic peptide

  •  
  • NYHA

    New York Heart Association

  •  
  • OAC

    Oral anticoagulant

  •  
  • o.d.

    Omnie die (once a day)

  •  
  • OR

    Odds ratio

  •  
  • OSA

    Obstructive sleep apnoea

  •  
  • PA

    Pulmonary artery

  •  
  • PAD

    Peripheral artery disease

  •  
  • PAH

    Pulmonary arterial hypertension

  •  
  • PAUSE

    Perioperative Anticoagulant Use for Surgery Evaluation (trial)

  •  
  • PBM

    Patient Blood Management

  •  
  • PCC

    Prothrombin complex concentrate

  •  
  • PCI

    Percutaneous coronary intervention

  •  
  • PE

    Pulmonary embolism

  •  
  • PMC

    Percutaneous mitral commissurotomy

  •  
  • PMI

    Peri-operative myocardial infarction/injury

  •  
  • POISE

    PeriOperative ISchemic Evaluation Trial

  •  
  • PPC

    Prothrombin complex concentrate

  •  
  • PT

    Prothrombin time

  •  
  • PVC

    Premature ventricular contractions

  •  
  • QI

    Quality indicator

  •  
  • RAAS

    Renin−angiotensin−aldosterone system

  •  
  • RBC

    Red blood cell

  •  
  • RCRI

    Revised Cardiac Risk Index

  •  
  • RCT

    Randomized controlled trial

  •  
  • RF

    Radiofrequency

  •  
  • rHuEPO

    Recombinant human erythropoietin

  •  
  • RR

    Relative risk

  •  
  • RV

    Right ventricular

  •  
  • SAPT

    Single antiplatelet therapy

  •  
  • SARS-CoV-2

    Severe acute respiratory syndrome coronavirus 2

  •  
  • SAVR

    Surgical aortic valve replacement

  •  
  • SCD

    Sudden cardiac death

  •  
  • SGLT-2

    Sodium–glucose co-transporter-2

  •  
  • SORT

    Surgical Outcome Risk Tool

  •  
  • SPAP

    Systolic pulmonary artery pressure

  •  
  • STEMI

    ST-segment elevation myocardial infarction

  •  
  • SVT

    Supraventricular tachycardia

  •  
  • TAVI

    Transcatheter aortic valve implantation

  •  
  • TEE

    Transoesophageal echocardiography

  •  
  • TEER

    Transcatheter edge-to-edge repair

  •  
  • TIA

    Transient ischaemic attack

  •  
  • TTE

    Transthoracic echocardiography

  •  
  • UFH

    Unfractionated heparin

  •  
  • ULN

    Upper limit of normal

  •  
  • VAD

    Ventricular assist device

  •  
  • VATS

    Video-assisted thoracic surgery

  •  
  • VEGFi

    Vascular endothelial grow factor inhibitor

  •  
  • VF

    Ventricular fibrillation

  •  
  • VHD

    Valvular heart disease

  •  
  • VISION

    Vascular Events in Noncardiac Surgery Patients Cohort Evaluation (trial)

  •  
  • VKA

    Vitamin K antagonist

  •  
  • VKORC1

    Vitamin K epoxide reductase complex 1

  •  
  • VO2

    Oxygen consumption

  •  
  • VT

    Ventricular tachycardia

  •  
  • VTE

    Venous thromboembolism

  •  
  • WHA

    World Health Assembly

  •  
  • WPW

    Wolff–Parkinson–White

1. Preamble

Guidelines summarize and evaluate available evidence, with the aim of assisting health professionals in proposing the best management strategies for an individual patient with a given condition. Guidelines and their recommendations should facilitate decision-making of health professionals in their daily practice. Guidelines, however, are not a substitute for the patient’s relationship with their practitioner. The final decisions concerning an individual patient must be made by the responsible health professional(s), based on what they consider to be the most appropriate in the circumstances. These decisions are made in consultation with the patient and caregiver as appropriate.

Guidelines are intended for use by health professionals. To ensure that all users have access to the most recent recommendations, the European Society of Cardiology (ESC) makes its guidelines freely available. The ESC warns readers that the technical language may be misinterpreted and declines any responsibility in this respect.

Many guidelines have been issued in recent years by the ESC. Because of their impact on clinical practice, quality criteria for the development of guidelines have been established in order to make all decisions transparent to the user. The recommendations for formulating and issuing ESC Guidelines can be found on the ESC website (https://www.escardio.org/Guidelines). The ESC Guidelines represent the official position of the ESC on a given topic and are regularly updated.

In addition to the publication of Clinical Practice Guidelines, the ESC carries out the EURObservational Research Programme of international registries of cardiovascular diseases and interventions, which are essential to assess diagnostic/therapeutic processes, use of resources, and adherence to guidelines. These registries aim to provide a better understanding of medical practice in Europe and around the world, and are based on high-quality data collected during routine clinical practice. Furthermore, the ESC develops sets of quality indicators (QIs)—which are tools to evaluate the level of implementation of the guidelines and may be used by the ESC, hospitals, healthcare providers, and professionals to measure clinical practice, and in educational programmes—alongside the key messages from the guidelines, to improve quality of care and clinical outcomes.

The members of this Task Force were selected by the ESC to represent professionals involved with the medical care of patients with this pathology. The selection procedure aimed to ensure that there is a representative mix of members, predominantly from across the whole of the ESC region and from relevant ESC Subspecialty Communities. Consideration was given to diversity and inclusion, notably with respect to gender and country of origin. A critical evaluation of diagnostic and therapeutic procedures was performed, including assessment of the risk–benefit ratio. The level of evidence and the strength of the recommendation of particular management options were weighed and scored according to pre-defined scales, as outlined below. The Task Force followed the ESC voting procedures. All recommendations subject to a vote achieved at least 75% among voting members.

The experts of the writing and reviewing panels provided declaration of interest forms for all relationships that might be perceived as real or potential sources of conflicts of interest. Their declarations of interest were reviewed according to the ESC declaration of interest rules and can be found on the ESC website (http://www.escardio.org/Guidelines) and have been compiled in a report and simultaneously published in a supplementary document to the guidelines. This process ensures transparency and prevents potential biases in the development and review processes. Any changes in declarations of interest that arose during the writing period were notified to the ESC and updated. The Task Force received its entire financial support from the ESC without any involvement from the healthcare industry.

The ESC CPG Committee supervises and coordinates the preparation of new guidelines. The Committee is also responsible for the approval process of these guidelines. The ESC Guidelines undergo extensive review by the CPG Committee and external experts, including a mix of members from across the whole of the ESC region and from relevant ESC Subspecialty Communities and National Cardiac Societies. After appropriate revisions, the guidelines are signed-off by all the experts involved in the Task Force. The finalized document is signed-off by the CPG Committee for publication in the European Heart Journal. The guidelines are developed after careful consideration of the scientific and medical knowledge and the evidence available at the time of their writing.

The task of developing the ESC Guidelines also includes creating educational tools and implementating programmes for the recommendations, including condensed pocket guidelines versions, summary slides, summary cards for non-specialists, and an electronic version for digital applications (smartphones, etc.). These versions are abridged and thus, for more detailed information, the user should always access the full text version of the guidelines, which is freely available via the ESC website and the European Heart Journal. The National Cardiac Societies of the ESC are encouraged to endorse, adopt, translate, and implement all ESC Guidelines. Implementation programmes are needed because it has been shown that the outcome of disease may be favourably influenced by the thorough application of clinical recommendations.

Health professionals are encouraged to take the ESC Guidelines fully into account when exercising their clinical judgment, and in determining and implementing preventive, diagnostic, or therapeutic medical strategies. However, the ESC Guidelines do not override, in any way whatsoever, the individual responsibility of health professionals to make appropriate and accurate decisions in considering each patient's health condition and in consulting with that patient or the patient’s caregiver where appropriate and/or necessary. It is also the health professional's responsibility to verify the rules and regulations applicable in each country to drugs and devices at the time of prescription and, where appropriate, to respect the ethical rules of their profession.

Off-label use of medication may be presented in these guidelines if a sufficient level of evidence shows that it can be considered medically appropriate to a given condition and if patients could benefit from the recommended therapy. However, the final decisions concerning an individual patient must be made by the responsible health professional, giving special consideration to:

  • the specific situation of the patient. In this respect, it is specified that, unless otherwise provided for by national regulations, off-label use of medication should be limited to situations where it is in the patient’s interest to do so, with regard to the quality, safety, and efficacy of care, and only after the patient has been informed and provided consent;

  • and country-specific health regulations, indications by governmental drug regulatory agencies, and the ethical rules to which health professionals are subject, where applicable.

2. Introduction

2.1. What is new

2.2. The magnitude of the problem

The annual volume of major surgery worldwide is estimated to be more than 300 million patients (about 5% of the world population), which is a 34% increase from 2004 to 2012.1,2 Nearly 74% of these operations are performed in countries spending substantial amounts on health care. When applied to European Union countries, which had an overall population of 448 million in 2020 (27 countries), this figure translates into a crude estimate of nearly 22 million major procedures annually.2

Nearly 85% of major operations are non-cardiac surgical procedures.3 In a recent report from the USA National Inpatient Sample database, nearly half of adults aged ≥45 years undergoing major non-cardiac surgery (NCS) presented with at least two cardiovascular (CV) risk factors, 18% had coronary artery disease (CAD), 4.7% had a history of stroke, and 7.7% had a modified Revised Cardiac Risk Index (RCRI) score ≥3 (range 0–6) in 2012–13. These prevalence rates show a substantial increase compared with the equivalent rates in 2008–09.4 In a large registry including 37 915 consecutive patients undergoing percutaneous coronary interventions (PCIs) with drug-eluting stent (DES), the rates of NCS after PCI were 11% and 24%, 1 and 3 years after PCI respectively. The cut-off ages at which NCS was more likely to occur within 1 and 3 years of PCI were 62 and 73 years respectively.5

The prevalence of comorbidities, the clinical condition of patients before surgery, and the urgency, magnitude, type, and duration of the surgical procedure determine the risk of peri-operative complications. In a recent cohort study of 40 000 patients aged ≥45 years undergoing inpatient NCS, one of seven experienced a major cardiac or cerebrovascular complication at 30 days.6 Cardiovascular complications can particularly occur in patients with documented or asymptomatic coronary heart disease, left ventricular (LV) dysfunction, valvular heart disease (VHD), and arrhythmias, who undergo surgical procedures that are associated with prolonged haemodynamic and cardiac stress. In the case of peri-operative myocardial ischaemia, three mechanisms are important: (i) oxygen supply–demand mismatch on the background of coronary artery stenosis that may become flow-limiting by peri-operative haemodynamic fluctuations; (ii) acute coronary syndrome (ACS) due to stress-induced erosion or rupture of a vulnerable atherosclerotic plaque in combination with pro-inflammatory and hypercoagulable states induced by surgery, and the haemodynamic distress resulting from fluid shifts and anaesthesia; and (iii) surgery-associated bleeding risk requiring interruption of antiplatelet therapies, which might lead to stent thrombosis among patients undergoing NCS after recent coronary stent placement. Left ventricular dysfunction and arrhythmias may occur for various reasons at all ages. Because the prevalence of CAD, VHD, heart failure, and arrhythmias increases with age, peri-operative CV mortality and morbidity are predominantly an issue in the adult population undergoing major NCS.

In Europe, recent systematic data on the annual number and type of operations, and on patient outcomes are unfortunately lacking. Additionally, data definitions vary, as do data quantity and quality. Based on the estimates outlined above, nearly 6.6 million procedures are performed annually in European patients with CAD, peripheral artery disease (PAD), and cerebrovascular disease who are at high risk of CV complications. In a 7 day cohort study, the European Surgical Outcomes Study (EuSOS) group investigated the outcomes of NCS in 498 hospitals across 27 European nations and the UK; up to 8% of patients undergoing NCS required critical care admission, while in-hospital mortality ranged 1.4–21.5% (mean 4.0%), depending on safety precautions.7 In a recent prospective study of 2265 high-risk patients undergoing NCS in Switzerland, one out of five developed major adverse events within 365 days.8 When applied to the population in European Union countries, these figures translate into at least 660 000 major cardiac or cerebrovascular complications occurring annually due to NCS procedures.

The 2022 ESC Guidelines on cardiovascular assessment and management of patients undergoing NCS focus on the pre-operative CV risk assessment and peri-operative management of patients in whom cardiovascular disease (CVD) is a potential source of complications during NCS.

2.3. Change in demographics

Within the next 30 years, the ageing of the population will have a major impact on peri-operative patient management. Patients undergoing NCS are older than the rest of the population. Furthermore, it is estimated that by 2030, one-fifth of individuals aged >75 years will undergo surgery each year. In addition, between 2018 and 2050, the number of people in Europe aged 75–84 years is projected to increase by ∼60%. The total number of surgical procedures may increase even faster because of the greater need for interventions with increasing age. Demographics of patients undergoing surgery show trends towards increasing numbers of elderly patients and increasing numbers of patients with comorbidities, particularly CVDs. Thus, adults aged ≥75 years have a greater risk of peri-operative major adverse cardiovascular events (MACEs) (9.5% vs. 4.8% for younger adults [P < 0.001]).9 However, age per se seems to be responsible for a small increase in the risk of complications; greater risks are associated with urgency and significant CV, pulmonary, and renal disease.

2.4. Purpose

As many years have passed and new evidence has become available since the publication of the 2014 ESC/European Society of Anaethesiology (ESA) Guidelines on non-cardiac surgery: cardiovascular assessment and management,10 the ESC has decided to revise the guidelines on NCS. These new guidelines are based on the 2014 edition, but all sections have been revised or rewritten, and several new sections have been added. Some of the old recommendations are unchanged or have been revised, and new recommendations have been added.

These guidelines are intended for physicians, healthcare workers, and collaborators involved in the pre-operative, operative, and post-operative care of patients undergoing NCS. The objective is to endorse a standardized and evidence-based approach to peri-operative CV management. The guidelines recommend a stepwise evaluation of the patient that integrates clinical risk factors and test results with the estimated stress of the planned surgical procedure and the risks involved with the discontinuation of drugs. This results in an individualized risk assessment, with the opportunity of initiating medical therapy, coronary interventions, and specific surgical and anaesthetic techniques, or withholding medical therapy, in order to optimize the patient’s peri-operative condition. Further, it should be discussed in which institutions (specialized small hospital vs. tertiary care) the NCS will be performed. It is important that patients’ values and preferences with respect to the benefits and risks of surgery are taken into consideration, and that patients are involved in the decisions. This is particularly important when it comes to decisions about undergoing elective surgery or not, the timing of surgery, and choice of surgical and anaesthetic techniques.

Compared with non-surgical settings, randomized controlled trials (RCTs) are scarce in this field. However, since the publication of the 2014 ESC/ESA Guidelines on non-cardiac surgery: cardiovascular assessment and management there has been a significant increase in RCTs that are relevant in this setting. When no trials are available on a specific CV management regimen in the surgical setting, data from the non-surgical setting may be extrapolated and similar recommendations made, but with different levels of evidence.

These guidelines have the potential to improve peri- and post-operative outcomes and highlight the existence of a clear opportunity for improving the quality of care. Following the publication of these updated guidelines on NCS, their effects on outcomes should be monitored. The objective evaluations of the quality of the assessments and the outcomes are described in quality indicators (Section 13).

2.5. The outcomes we want to prevent

The recommendations in these guidelines are intended to prevent peri-operative CV morbidity and mortality, for example: peri-operative myocardial infarction/injury (PMI), stent thrombosis, acute heart failure (HF), haemodynamically relevant arrhythmias, pulmonary embolism (PE), ischaemic stroke, and death. It is also important to prevent bleeding complications, especially associated with antithrombotic treatment, since bleeding is associated with an increased risk of MI and death.6,11–13

3. Clinical risk evaluation

Cardiovascular morbidity and mortality in patients undergoing NCS are determined by two main factors: patient-related risk and type of surgery or procedure, including the circumstances under which it takes place (experience of institution, elective vs. emergency procedure).14 The risk may be reduced by an adequate pre-operative evaluation and proper selection of type and timing of the surgical procedure (Figure 1).

Total risk is an interaction of patient-related and surgery-related risk.
Figure 1

Total risk is an interaction of patient-related and surgery-related risk.

Ideally, the total risk should be as close as possible to the lower left corner, by choosing surgery/procedure/anaesthesia/institution with the lowest possible risk along with efforts to mitigate the patient’s CV risk.

3.1. Surgery-related risk

The surgery-related risk is determined by the type and duration of the surgery, and the urgency of the procedure or intervention. The type of anaesthesia and anaesthetic drugs may also influence the risk of complications in patients at intermediate to high cardiac risk undergoing NCS (see Section 7).15 The surgical risk estimate is a broad approximation of 30 day risk of CV death, MI, and stroke, which only takes into account the specific surgical intervention without considering the patient’s comorbidities (Table 5).10,16

Table 1

Classes of recommendations

graphic
graphic
Table 1

Classes of recommendations

graphic
graphic
Table 2

Levels of evidence

graphic
graphic
Table 2

Levels of evidence

graphic
graphic
Table 3

New concepts and sections in the current guidelines

A new flowchart for general assessment of patients before NCS.
A new section on pre-operative assessment of patients with newly detected murmurs, dyspnoea, oedema, or angina.
A new section on the patient perspective.
A new section on assessment of frailty.
A revised and expanded focus on use of biomarkers in NCS
A revised and expanded section on peri-operative management of antiplatelet therapy.
A revised and expanded section on peri-operative management of oral anticoagulants.
A new section on peri-operative thromboprophylaxis.
A dedicated section on patient blood management.
A new section on management of cardiovascular risk in patients with cancer undergoing NCS.
A small section on NCS in patients with recent COVID-19.
A new section on diagnosis and management of post-operative complications during NCS.
A new flowchart for general assessment of patients before NCS.
A new section on pre-operative assessment of patients with newly detected murmurs, dyspnoea, oedema, or angina.
A new section on the patient perspective.
A new section on assessment of frailty.
A revised and expanded focus on use of biomarkers in NCS
A revised and expanded section on peri-operative management of antiplatelet therapy.
A revised and expanded section on peri-operative management of oral anticoagulants.
A new section on peri-operative thromboprophylaxis.
A dedicated section on patient blood management.
A new section on management of cardiovascular risk in patients with cancer undergoing NCS.
A small section on NCS in patients with recent COVID-19.
A new section on diagnosis and management of post-operative complications during NCS.

COVID-19, coronavirus 2019; NCS, non-cardiac surgery

Table 3

New concepts and sections in the current guidelines

A new flowchart for general assessment of patients before NCS.
A new section on pre-operative assessment of patients with newly detected murmurs, dyspnoea, oedema, or angina.
A new section on the patient perspective.
A new section on assessment of frailty.
A revised and expanded focus on use of biomarkers in NCS
A revised and expanded section on peri-operative management of antiplatelet therapy.
A revised and expanded section on peri-operative management of oral anticoagulants.
A new section on peri-operative thromboprophylaxis.
A dedicated section on patient blood management.
A new section on management of cardiovascular risk in patients with cancer undergoing NCS.
A small section on NCS in patients with recent COVID-19.
A new section on diagnosis and management of post-operative complications during NCS.
A new flowchart for general assessment of patients before NCS.
A new section on pre-operative assessment of patients with newly detected murmurs, dyspnoea, oedema, or angina.
A new section on the patient perspective.
A new section on assessment of frailty.
A revised and expanded focus on use of biomarkers in NCS
A revised and expanded section on peri-operative management of antiplatelet therapy.
A revised and expanded section on peri-operative management of oral anticoagulants.
A new section on peri-operative thromboprophylaxis.
A dedicated section on patient blood management.
A new section on management of cardiovascular risk in patients with cancer undergoing NCS.
A small section on NCS in patients with recent COVID-19.
A new section on diagnosis and management of post-operative complications during NCS.

COVID-19, coronavirus 2019; NCS, non-cardiac surgery

Table 4

What is new

Table 4A New recommendations

graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
Table 4

What is new

Table 4A New recommendations

graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
graphic
Table 4B

Revised recommendations

graphic
graphic
graphic
graphic
Table 4B

Revised recommendations

graphic
graphic
graphic
graphic
Table 5

Surgical risk estimate according to type of surgery or intervention

Low surgical risk (<1%)Intermediate surgical risk (1–5%)High surgical risk (>5%)
  • Breast

  • Dental

  • Endocrine: thyroid

  • Eye

  • Gynaecological: minor

  • Orthopaedic minor (meniscectomy)

  • Reconstructive

  • Superficial surgery

  • Urological minor: (transurethral resection of the prostate)

  • VATS minor lung resection

  • Carotid asymptomatic (CEA or CAS)

  • Carotid symptomatic (CEA)

  • Endovascular aortic aneurysm repair

  • Head or neck surgery

  • Intraperitoneal: splenectomy, hiatal hernia repair, cholecystectomy

  • Intrathoracic: non-major

  • Neurological or orthopaedic: major (hip and spine surgery)

  • Peripheral arterial angioplasty

  • Renal transplants

  • Urological or gynaecological: major

  • Adrenal resection

  • Aortic and major vascular surgery

  • Carotid symptomatic (CAS)

  • Duodenal-pancreatic surgery

  • Liver resection, bile duct surgery

  • Oesophagectomy

  • Open lower limb revascularization for acute limb ischaemia or amputation

  • Pneumonectomy (VATS or open surgery)

  • Pulmonary or liver transplant

  • Repair of perforated bowel

  • Total cystectomy

Low surgical risk (<1%)Intermediate surgical risk (1–5%)High surgical risk (>5%)
  • Breast

  • Dental

  • Endocrine: thyroid

  • Eye

  • Gynaecological: minor

  • Orthopaedic minor (meniscectomy)

  • Reconstructive

  • Superficial surgery

  • Urological minor: (transurethral resection of the prostate)

  • VATS minor lung resection

  • Carotid asymptomatic (CEA or CAS)

  • Carotid symptomatic (CEA)

  • Endovascular aortic aneurysm repair

  • Head or neck surgery

  • Intraperitoneal: splenectomy, hiatal hernia repair, cholecystectomy

  • Intrathoracic: non-major

  • Neurological or orthopaedic: major (hip and spine surgery)

  • Peripheral arterial angioplasty

  • Renal transplants

  • Urological or gynaecological: major

  • Adrenal resection

  • Aortic and major vascular surgery

  • Carotid symptomatic (CAS)

  • Duodenal-pancreatic surgery

  • Liver resection, bile duct surgery

  • Oesophagectomy

  • Open lower limb revascularization for acute limb ischaemia or amputation

  • Pneumonectomy (VATS or open surgery)

  • Pulmonary or liver transplant

  • Repair of perforated bowel

  • Total cystectomy

CAS, carotid artery stenting; CEA, carotid endarterectomy; CV, cardiovascular; MI, myocardial infarction; VATS, video-assisted thoracic surgery.

Surgical risk estimate is a broad approximation of 30 day risk of CV death, MI, and stroke that takes into account only the specific surgical intervention, without considering the patient’s comorbidities.

Adapted from data in Glance et al., Muller et al., Bendixen et al., and Falcoz et al.18–23

Table 5

Surgical risk estimate according to type of surgery or intervention

Low surgical risk (<1%)Intermediate surgical risk (1–5%)High surgical risk (>5%)
  • Breast

  • Dental

  • Endocrine: thyroid

  • Eye

  • Gynaecological: minor

  • Orthopaedic minor (meniscectomy)

  • Reconstructive

  • Superficial surgery

  • Urological minor: (transurethral resection of the prostate)

  • VATS minor lung resection

  • Carotid asymptomatic (CEA or CAS)

  • Carotid symptomatic (CEA)

  • Endovascular aortic aneurysm repair

  • Head or neck surgery

  • Intraperitoneal: splenectomy, hiatal hernia repair, cholecystectomy

  • Intrathoracic: non-major

  • Neurological or orthopaedic: major (hip and spine surgery)

  • Peripheral arterial angioplasty

  • Renal transplants

  • Urological or gynaecological: major

  • Adrenal resection

  • Aortic and major vascular surgery

  • Carotid symptomatic (CAS)

  • Duodenal-pancreatic surgery

  • Liver resection, bile duct surgery

  • Oesophagectomy

  • Open lower limb revascularization for acute limb ischaemia or amputation

  • Pneumonectomy (VATS or open surgery)

  • Pulmonary or liver transplant

  • Repair of perforated bowel

  • Total cystectomy

Low surgical risk (<1%)Intermediate surgical risk (1–5%)High surgical risk (>5%)
  • Breast

  • Dental

  • Endocrine: thyroid

  • Eye

  • Gynaecological: minor

  • Orthopaedic minor (meniscectomy)

  • Reconstructive

  • Superficial surgery

  • Urological minor: (transurethral resection of the prostate)

  • VATS minor lung resection

  • Carotid asymptomatic (CEA or CAS)

  • Carotid symptomatic (CEA)

  • Endovascular aortic aneurysm repair

  • Head or neck surgery

  • Intraperitoneal: splenectomy, hiatal hernia repair, cholecystectomy

  • Intrathoracic: non-major

  • Neurological or orthopaedic: major (hip and spine surgery)

  • Peripheral arterial angioplasty

  • Renal transplants

  • Urological or gynaecological: major

  • Adrenal resection

  • Aortic and major vascular surgery

  • Carotid symptomatic (CAS)

  • Duodenal-pancreatic surgery

  • Liver resection, bile duct surgery

  • Oesophagectomy

  • Open lower limb revascularization for acute limb ischaemia or amputation

  • Pneumonectomy (VATS or open surgery)

  • Pulmonary or liver transplant

  • Repair of perforated bowel

  • Total cystectomy

CAS, carotid artery stenting; CEA, carotid endarterectomy; CV, cardiovascular; MI, myocardial infarction; VATS, video-assisted thoracic surgery.

Surgical risk estimate is a broad approximation of 30 day risk of CV death, MI, and stroke that takes into account only the specific surgical intervention, without considering the patient’s comorbidities.

Adapted from data in Glance et al., Muller et al., Bendixen et al., and Falcoz et al.18–23

Any surgical procedure may increase the level of cortisol and catecholamines as stress responses due to tissue injury and inflammation, and neuro–endocrine and sympathovagal imbalance. Changes in body core temperature, blood loss, and fluid shifts may cause a rise in vascular resistance as well as hypotension,17 leading to imbalance between myocardial oxygen demand and delivery. Bleeding, transfusion of blood products, tissue injury, and inflammatory response may affect the coagulation system, inducing a prothrombotic state.

3.1.1. Timing of surgery

In general, acute procedures carry a higher risk of complications than elective procedures. Uniform timing definitions are unfeasible, as the time spans may vary between diseases. These guidelines use the timing definitions below.

Immediate: surgery/intervention should be performed without any delay to save life or organ function.

Urgent: surgery/intervention should be performed without unnecessary delay to save life, limb, or organ function.

Time-sensitive: surgery/intervention should be performed as soon as possible as there is a time-dependent risk of losing limb or organ function, or increased risk of complications. Cancer surgery is typically time-sensitive, as is carotid surgery to prevent stroke in a symptomatic case. The time window for time-sensitive surgery will vary depending on the underlying disease.

Elective: surgery/intervention can be performed electively (not further defined) without significant risk of losing limb, or organ function, or increased risks of complications.

Many factors affect outcomes when comparing acute or time-sensitive vs. elective surgery: the general condition of the patient vs. the stage of the acute illness, and how far it has progressed. The best interests of the patient should be considered before deciding on treatment, informed consent to management should be obtained, if at all possible, and decisions should be clearly recorded.24

The degree of urgency should also be considered (i.e. does the procedure need to be performed outside working hours or can it wait until the next day?). In general, competences and supportive functions are not always present in the evenings or during the night; thus, an overall evaluation of what best serves the patient is necessary. The optimal timing of NCS should be discussed within the multidisciplinary team, including an anaesthesiologist, in order to achieve optimized anaesthesia for each patient (see Section 7).

3.2. Type of surgical approach

New surgical techniques have been introduced to replace open surgery and to reduce the overall risk for the patient.

3.2.1. Laparoscopy

Laparoscopic procedures, compared with open surgical procedures, have the advantage of causing less tissue trauma and intestinal paralysis, resulting in less incisional pain, better post-operative pulmonary function, significantly fewer wall complications, and diminished post-operative fluid shifts related to bowel paralysis.25 However, the pneumoperitoneum required for these procedures results in elevated intra-abdominal pressure and a reduction in venous return. Typical physiological sequelae are secondary to increased intra-abdominal pressure and absorption of the gaseous medium used for insufflation.

While healthy individuals on controlled ventilation typically tolerate pneumoperitoneum, patients with CVD, some types of adults with congenital heart disease (ACHD), and obese patients may experience adverse consequences.26 Pneumoperitoneum and Trendelenburg position result in increased mean arterial pressure, central venous pressure, mean pulmonary artery pressure, pulmonary capillary wedge pressure, and systemic vascular resistance impairing cardiac function.27,28 Therefore, compared with open surgery, the CV risk in patients with CVD is not necessarily reduced in patients undergoing laparoscopy, and both should be evaluated in the same way. This is especially true in patients undergoing interventions for morbid obesity, but also in other types of surgery, considering the risk of conversion to an open procedure.29,30 Superior short-term outcomes of laparoscopic vs. open procedures have been reported, depending on type of surgery, operator experience, and hospital volume; however, few studies provide direct measures of cardiac complications.31–33 The benefit of laparoscopic procedures is probably greater in elderly patients, with reduced length of hospital stay, intra-operative blood loss, incidence of post-operative pneumonia, time to return of normal bowel function, incidence of post-operative cardiac complications, and wound infections.34

3.2.1.1. Vascular and endovascular procedures

Endovascular abdominal aortic aneurysm repair (EVAR) is a procedure using femoral artery access only, and is therefore associated with lower operative mortality and morbidity than open repair. It minimizes the surgical risk in simultaneous surgery for the treatment of abdominal aortic aneurysm (AAA) and a non-cardiac disorder, and shortens the time delay from the treatment of AAA and the non-cardiac disorder in patients undergoing two-phase surgery.35–37 The early gain in mortality from EVAR procedures is lost after 3–4 years, compared with open surgical treatment, due to general morbidity (especially CV mortality) of AAA patients.

Various vascular and non-vascular NCS procedures bear different operative risks. While aortic and infra-inguinal vascular surgical procedures are both regarded as high-risk procedures, their risk can be modified by adequate peri-operative measures.38 For patients undergoing treatment of femoropopliteal artery disease, an endovascular-first approach may be advisable in case of additional significant comorbidity. A meta-analysis of studies comparing open surgery with PCI for the treatment of femoropopliteal arterial disease showed that femoral bypass surgery was associated with higher morbidity (odds ratio [OR] 2.93; 95% confidence interval [CI], 1.34–6.41) but similar mortality at 30 days compared with endovascular treatment.39

3.2.1.2. Video-assisted non-cardiac surgery

Video-assisted thoracic surgery (VATS) is supported by a trial showing fewer peri-operative complications and a better quality of life in the first year following surgery for stage 1 lung cancer compared with anterolateral thoracotomy.20 Also, a large propensity matched study conducted by the European Society of Thoracic Surgeons (ESTS) showed fewer post-operative complications following VATS compared with open thoracotomy.21 Overall, the benefits seem greatest in patients with reduced functional lung capacity.

Recommendation Table 1

Recommendations for selection of surgical approach and impact on risk

graphic
graphic
Recommendation Table 1

Recommendations for selection of surgical approach and impact on risk

graphic
graphic

3.3. Patient-related risk

3.3.1. Initial assessment

Patient-related risk is determined by patient’s age, the presence or absence of CV risk factors (e.g. smoking, hypertension, diabetes, dyslipidaemia, family disposition)40 or established CV disease, and comorbidities.41

Identification of patients at risk of CV complications is of paramount importance to choice of therapy when non-surgical options are available, or when the type of surgery or anaesthesia impacts the risk of complications. When emergency surgery is needed, the evaluation must necessarily be limited; however, most clinical circumstances allow a systematic approach.

As an initial assessment, it is recommended that all patients scheduled for NCS are evaluated by accurate history and physical examination, with special emphasis on CV risk factors, established CV disease, and comorbidities.40 It is also recommended to measure standard laboratory tests (e.g. haemoglobin and renal function) in all patients undergoing intermediate- to high-risk surgery. Based on this information, further assessment of patient-related risk can proceed depending on the surgery-related risk, as shown in Figure 2. It is recommended to perform an electrocardiogram (ECG), assess the functional capacity, and/or measure biomarkers (cardiac troponins and/or N-terminal pro-B-type natriuretic peptide [NT-proBNP]/B-type natriuretic peptide [BNP]) depending on the patient-related and surgery-related risk (Figure 2). Detailed information on available tools for risk assessment, their prognostic ability, and indications to perform them is given in Section 4. More details on pre-operative management of patients with specific CV diseases are given in Section 6.

Pre-operative assessment before non-cardiac surgery.
Figure 2

Pre-operative assessment before non-cardiac surgery.

CV, cardiovascular; CVD, cardiovascular disease; ECG, electrocardiogram; N, no; NCS, non-cardiac surgery. Y, yes; aCV risk factors: hypertension, smoking, dyslipidaemia, diabetes, family history of CVD. bBiomarkers: hs-cTn T/I (Class I) and/or BNP/NT-proBNP (Class IIa). If pathological, consult a cardiologist. cFunctional capacity based on Duke Activity Status Index (DASI) or the ability to climb two flights of stairs. dFor diagnostic and therapeutic efforts to be considered, see Section 6. eClose follow-up after intervention and subsequent management of heart disease are advised.

3.3.1.1. Patients aged <65 years without a history of cardiovascular disease or cardiovascular risk factors

Patients aged <65 years without signs, symptoms, or history of CVD or CV risk factors are considered to be of low risk, and can proceed to low- and moderate-risk surgery without additional pre-operative risk assessment.41 Before high-risk surgery, ECG and biomarkers should be considered (see Sections 4.3 and 4.4).42

Patients without signs or symptoms of CVD, but with a family history of genetic cardiomyopathy (i.e. dilatated, hypertrophic, arrhythmic, or restrictive cardiomyopathy, or LV non-compaction) should be evaluated with an ECG and an echocardiographic examination to rule out the presence of the disease, irrespective of the age.43 No specific data are available in the literature regarding risk of family members without the phenotype; however, they are at risk of developing the disease, which may be subclinical at the time of the NCS.43

3.3.1.2. Patients aged ≥65 years or with cardiovascular risk factors

Patients who are aged ≥65 years and patients with risk factors for CVD—such as hypertension, dyslipidaemia, or smoking—have an increased risk of having undetected CVD. The SCORE2 risk-prediction tool can be used to estimate their 10 year CVD risk outside the setting of NCS.40 Patients who are aged ≥65 years and patients with risk factors for CVD also have an increased risk of peri-operative complications during NCS.41,44 These patients need additional assessment before intermediate- and high-risk surgery (Figure 2) and optimal treatment of risk factors. This is also the case for patients with other diseases known to be associated with a high risk of concomitant undetected or known CVD (Sections 6.8 and 6.116.14).

3.3.1.3. Patients with established cardiovascular disease

The surgical procedure has the potential to aggravate the disease and increase morbidity and mortality in patients with established CVD. This may be preventable by implementing appropriate CV risk stratification prior to NCS and individually tailoring peri-operative therapy to reduce the risk.45 If time allows, it is also recommended to optimize guideline-recommended treatment of the disease before NCS. See Section 6 for a detailed discussion of risk assessment and management of patients with known CVD.

Recommendation Table 2

Recommendations for all patients scheduled for non-cardiac surgery

graphic
graphic
Recommendation Table 2

Recommendations for all patients scheduled for non-cardiac surgery

graphic
graphic
Recommendation Table 3

Recommendations for patients aged <65 years without signs, symptoms, or history of cardiovascular disease

graphic
graphic
Recommendation Table 3

Recommendations for patients aged <65 years without signs, symptoms, or history of cardiovascular disease

graphic
graphic

3.3.2. Patients with murmurs, chest pain, dyspnoea, or peripheral oedema

Patients without known CVD and scheduled for elective or acute NCS are often referred to a cardiologist because of symptoms or signs that may be caused by CVD. Murmurs, chest pain, dyspnoea, and oedema may suggest severe CVD, but may also be caused by non-cardiac disease. Thus, the medical history, family history, and risk factors have to be obtained and considered. The patient’s physical capacity should be assessed. The need for further evaluation of the patient should be decided according to the risk of the planned procedure or surgery.

3.3.2.1. Murmurs

In a patient with a heart murmur, but without any symptoms of CVD, the value of performing an echocardiogram is not well-established and consensus is missing.54–56 However, if a heart murmur suggesting clinically significant pathology is present before high-risk NCS, it is recommended to perform an echocardiogram, even in patients without any symptoms of CVD. Old age or increased NT-proBNP may increase the pre-test probability of haemodynamically significant but asymptomatic valvular disease. If the patient with the murmur also has symptoms of CVD, an echocardiogram is indicated before all NCS. The pre-operative setting is challenging, as the need for NCS and the risk of CVD have to be considered as independent factors. Thus, an echocardiogram may be useful in risk stratification for some patients, but whether it would improve outcome is uncertain. It is important to bear in mind that the time delay when performing additional but unnecessary examinations may worsen the patient’s prognosis.57 It has also been discussed that a focused cardiac ultrasound (FOCUS) could replace auscultation in general in the pre-operative evaluation of patients.58 While cardiac auscultation has severe limitations,59,60 the value of performing a FOCUS as a standard pre-operative evaluation remains uncertain. Cardiac auscultation should not be replaced by FOCUS.

3.3.2.2. Chest pain

Patients scheduled for NCS may also present with previously unrecognized symptoms suggestive of CAD. The disease leading to the need for NCS may aggravate a subclinical CAD, or the patient may have a concomitant undetected CAD. In an elective setting, if the symptoms are suggestive of CAD, the guidelines for CAD patients in the non-surgical setting should be followed (see Sections 4.5.3 and 6.1.2). If immediate, urgent, or time-sensitive NCS is needed, the time for and access to adequate diagnostic tools may be limited. However, ECG and troponins can be used to detect or exclude ACS (see Sections 4.3 and 4.4).

3.3.2.3. Dyspnoea

Dyspnoea is a symptom of a wide range of diseases and conditions. In a large series of patients, self-reported dyspnoea identified a subgroup of otherwise asymptomatic patients at increased risk of death from CVD and any cause.61 In the diagnostic work-up to find the reason for dyspnoea, spirometry, D-dimer, NT-proBNP/BNP, arterial blood gases, and transthoracic echocardiography (TTE) have diagnostic utility61 but limited specificity. If NT-proBNP/BNP is elevated, an echocardiogram should be performed. If NT-proBNP/BNP is not elevated, other reasons for dyspnoea should be explored.

3.3.2.4. Peripheral oedema

Increased hydrostatic pressure leading to oedema is a feature of a wide range of CV diseases, but an upright position is also a common cause of oedema. There is a spectrum of other diseases that can result in peripheral oedema not listed here.

Recommendation Table 4

Recommendations for pre-operative assessment in patients with previously unknown murmur, angina, dyspnoea, or peripheral oedema

graphic
graphic
graphic
graphic
Recommendation Table 4

Recommendations for pre-operative assessment in patients with previously unknown murmur, angina, dyspnoea, or peripheral oedema

graphic
graphic
graphic
graphic

3.4. Timing of adequate risk evaluation

Pre-operative CV assessment should be performed prior to surgery, ideally at the time when the decision for NCS has been made. Accurate estimates of the risks and benefits of surgery is a prerequisite for informed decision-making by both physicians and patients about the appropriateness of surgery. These estimates should also help in guiding surgical (endovascular/endoscopic vs. open approach) and monitoring (intermediate care, screening for CV complications) approaches, and help to detect an unexpectedly high CV risk.47 Therefore, the prognostic value of pre-operative CV risk assessment is much higher in elective vs. immediate or urgent surgery. Explicit communication of peri-operative CV risk, on the basis of the expected event rates,47 and risk communication tools such as the A to Z Inventory of Decision Aids (https://decisionaid.ohri.ca/AZinvent.php) are recommended.

3.5. Avoidance or allowance for surgery in the individual patient

In the clinical setting it can be difficult to decide whether CVD represents a contraindication to NCS. In general, the risk for the patient if not operated on must be considerably higher than the risk of the treatment. Ideally, an unstable cardiac patient should be stabilized before NCS, but waiting can be detrimental for acute surgical disease. No definite list can be made for which cardiac disease is a clear contraindication to NCS, but in patients with severe HF (New York Heart Association [NYHA] class IV), cardiogenic shock, severe pulmonary hypertension, or patients with severe frailty (see Section 4.1.2 for frailty assessment), high-risk NCS should probably be avoided. Life expectancy and quality of life should also be taken into consideration. However, the decision should be made after discussions between the surgeon, anaesthesiologist, cardiologist, and also a geriatrician for elderly patients, along with the patient and relatives.

3.6. The patient perspective

Patients with established CVD may face concerns about their underlying disease and current CV medication, co-ordination between the surgical team and their cardiologist (examples provided in Figure 3), and the potential excessive risk compared with the expected outcome of the surgery. Time should be allowed to address concerns and to provide evidence-based information on the risk–benefit trade-offs and the surgical treatment options (including non-surgical or ‘do nothing’ alternatives) to ensure informed consent, and to allow patients to engage in shared decision-making with the aim of supporting the best decision. The team needs to understand the patient’s concerns and expectations about the treatment and short- and long-term goals, as the risk-benefits of the intervention may not be aligned with patient preferences and wishes. Communicating in plain language (oral and written) and targeting communication to fit the individual level of health literacy is pivotal. Several studies have indicated a relatively high prevalence of limited health literacy in patients with CVD (e.g. with HF),62 and limited health literacy is associated with adverse outcomes.63 An example of a patient information sheet to be used in the communication with patients is given in the Supplementary data, Table S1.

Examples of questions and concerns expressed by patients.
Figure 3

Examples of questions and concerns expressed by patients.

Recent systematic reviews and meta-analyses have focused on shared decision-making in the field of surgery across disease areas.64–67 In general, shared decision-making positively impacts decisional conflicts, knowledge gained, satisfaction, and decisional anxiety (although cultural variations may exist).67 In the breast cancer/endocrine and urology specialties, decision-making and communication aids appear to be effective methods for supporting patients’ involvement in decision-making when undergoing elective surgery. Moreover, educational information, provided through interactive multimedia, computer, or on DVD, used prior to the surgical consultation could enhance the decision-making process in addition to face-to-face communication.66

In Europe, the prevalence of pre-operative anxiety among patients undergoing surgical procedures varies from 27–80%.68 Although a certain level of anxiety in patients must be expected, peri-operative anxiety is associated with worse surgical outcomes and longer recovery,69–72 which highlights the importance of pre-surgical assessment and, in some patients, treatment of anxiety. Factors associated with pre-operative anxiety are complex and include, among others, age, sex, educational level, type of surgery, and fear of post-operative complications or the outcome.68 Psychological reactions in patients undergoing high- or medium–high-risk procedures and/or patients with previous negative experiences of NCS may warrant particular attention. Concerns and fears expressed by patients and relatives should be taken seriously. A number of reviews and meta-analyses have summarized the effects of interventions on surgical outcomes in abdominal, cardiac, and orthopaedic surgery, which may also be applicable to patients with CV conditions in these settings.73–75

Recommendation Table 5

Recommendations for patient information

graphic
graphic
Recommendation Table 5

Recommendations for patient information

graphic
graphic

4. Pre-operative assessment tools

4.1. Risk scores

4.1.1. General risk calculators

Several risk indices have been developed based on multivariable analyses of observational data and have been validated during the last decade (Table 6).47,49,76 Most risk calculators integrate both patient-related and surgery-related risk factors, but none of them include biomarkers among their variables. Calculators for most of the commonly used risk indices are available online (Table 6). The risk calculators can be used in addition, or as an alternative, to the assessment of surgery-related and patient-related risk factors described in Section 3.3. The Task Force decided against recommending one specific risk score. The Task Force also decided that the selection criteria for further pre-operative testing should be clinical criteria, and not based on a specific score.

Table 6

Risk score calculators

Revised Cardiac Risk Index (RCRI) (1999)aSurgical Risk Calculator (2011)The American College of Surgery National Surgical Quality Improvement Program (ACS NSQIP) (2013)Surgical Outcome Risk Tool (SORT) (2014)The American University of Beirut (AUB)-HAS2 Cardiovascular Risk Index (2019)b
VariablesIschaemic heart disease
Cerebrovascular disease
History of congestive heart failure
Insulin therapy for diabetes
Serum creatinine level ≥2 mg/dL
High-risk surgery
(each assigned 1 point)
Age
ASA–PS grade
Pre-operative dependent functional status
Creatinine >1.5 mg/dL
Type of surgery
Age
Sex
Functional status
Emergency case
ASA class
Current steroid use
Ascites within 30 days
Systemic sepsis within 48 h
Ventilator dependence
Disseminated cancer
Diabetes
Hypertension on treatment
Congestive HF
Dyspnoea
Current smoker
History of severe COPD
Dialysis
Acute renal failure
Body mass index
Surgery code
ASA–PS grade
Urgency of surgery
High-risk surgical specialty
Surgical severity (from minor to complex major)
Cancer
Age ≥65 years or over
History of Heart disease
Symptoms of Heart disease (angina or dyspnoea)
Age ≥75 years
Anaemia (haemoglobin <12 g/dL)
Vascular Surgery
Emergency Surgery
(2 H, 2 A and 2 S)
(each assigned 1 point)
Score rangeScore 1; risk 6.0% (4.9–7.4)
Score 2; risk 10.1% (8.1–10.6)
Score ≥3; risk 15% (11.1–20.0)
Absolute risk: 0–100%Absolute risk: 0–100%Absolute risk: 0–100%Low risk (score 0–1); (0.3 and 1.6%)c
Intermediate risk (score 2–3); (7.1 and 17%)c
High risk (score >3); (>17%)c
Outcome30 day MI, cardiac arrest, deathIntra-operative and 30 day MI or cardiac arrestSerious complications and any complications at 30 days30 day mortality30 day death, MI, or stroke
Derivation population1422211 4101 414 00611 2193284
Validation populationExternally validated in various surgical populations257 385Externally validated in various surgical populations22 6311 167 414
Model performance (AUC)0.68–0.760.81–0.850.730.81–0.920.82
Interactive calculatorhttps://www.mdcalc.com/revised-cardiac-risk-index-pre-operative-riskhttp://www.surgicalriskcalculator.com/miorcardiacarresthttps://riskcalculator.facs.orghttp://www.sortsurgery.com
Revised Cardiac Risk Index (RCRI) (1999)aSurgical Risk Calculator (2011)The American College of Surgery National Surgical Quality Improvement Program (ACS NSQIP) (2013)Surgical Outcome Risk Tool (SORT) (2014)The American University of Beirut (AUB)-HAS2 Cardiovascular Risk Index (2019)b
VariablesIschaemic heart disease
Cerebrovascular disease
History of congestive heart failure
Insulin therapy for diabetes
Serum creatinine level ≥2 mg/dL
High-risk surgery
(each assigned 1 point)
Age
ASA–PS grade
Pre-operative dependent functional status
Creatinine >1.5 mg/dL
Type of surgery
Age
Sex
Functional status
Emergency case
ASA class
Current steroid use
Ascites within 30 days
Systemic sepsis within 48 h
Ventilator dependence
Disseminated cancer
Diabetes
Hypertension on treatment
Congestive HF
Dyspnoea
Current smoker
History of severe COPD
Dialysis
Acute renal failure
Body mass index
Surgery code
ASA–PS grade
Urgency of surgery
High-risk surgical specialty
Surgical severity (from minor to complex major)
Cancer
Age ≥65 years or over
History of Heart disease
Symptoms of Heart disease (angina or dyspnoea)
Age ≥75 years
Anaemia (haemoglobin <12 g/dL)
Vascular Surgery
Emergency Surgery
(2 H, 2 A and 2 S)
(each assigned 1 point)
Score rangeScore 1; risk 6.0% (4.9–7.4)
Score 2; risk 10.1% (8.1–10.6)
Score ≥3; risk 15% (11.1–20.0)
Absolute risk: 0–100%Absolute risk: 0–100%Absolute risk: 0–100%Low risk (score 0–1); (0.3 and 1.6%)c
Intermediate risk (score 2–3); (7.1 and 17%)c
High risk (score >3); (>17%)c
Outcome30 day MI, cardiac arrest, deathIntra-operative and 30 day MI or cardiac arrestSerious complications and any complications at 30 days30 day mortality30 day death, MI, or stroke
Derivation population1422211 4101 414 00611 2193284
Validation populationExternally validated in various surgical populations257 385Externally validated in various surgical populations22 6311 167 414
Model performance (AUC)0.68–0.760.81–0.850.730.81–0.920.82
Interactive calculatorhttps://www.mdcalc.com/revised-cardiac-risk-index-pre-operative-riskhttp://www.surgicalriskcalculator.com/miorcardiacarresthttps://riskcalculator.facs.orghttp://www.sortsurgery.com

AUC, area under curve; ASA–PS, American Society of Anesthesiology Physical Status; COPD, chronic obstructive pulmonary disease; HF, heart failure; MI, myocardial infarction; RCRI, Revised Cardiac Risk Index.

a

The RCRI was updated January 2019.

b

Dakik et al. 2019, 2020, and 2022, and Msheik et al.49–51,81

c

The percentages relate to general surgeries.50

Table 6

Risk score calculators

Revised Cardiac Risk Index (RCRI) (1999)aSurgical Risk Calculator (2011)The American College of Surgery National Surgical Quality Improvement Program (ACS NSQIP) (2013)Surgical Outcome Risk Tool (SORT) (2014)The American University of Beirut (AUB)-HAS2 Cardiovascular Risk Index (2019)b
VariablesIschaemic heart disease
Cerebrovascular disease
History of congestive heart failure
Insulin therapy for diabetes
Serum creatinine level ≥2 mg/dL
High-risk surgery
(each assigned 1 point)
Age
ASA–PS grade
Pre-operative dependent functional status
Creatinine >1.5 mg/dL
Type of surgery
Age
Sex
Functional status
Emergency case
ASA class
Current steroid use
Ascites within 30 days
Systemic sepsis within 48 h
Ventilator dependence
Disseminated cancer
Diabetes
Hypertension on treatment
Congestive HF
Dyspnoea
Current smoker
History of severe COPD
Dialysis
Acute renal failure
Body mass index
Surgery code
ASA–PS grade
Urgency of surgery
High-risk surgical specialty
Surgical severity (from minor to complex major)
Cancer
Age ≥65 years or over
History of Heart disease
Symptoms of Heart disease (angina or dyspnoea)
Age ≥75 years
Anaemia (haemoglobin <12 g/dL)
Vascular Surgery
Emergency Surgery
(2 H, 2 A and 2 S)
(each assigned 1 point)
Score rangeScore 1; risk 6.0% (4.9–7.4)
Score 2; risk 10.1% (8.1–10.6)
Score ≥3; risk 15% (11.1–20.0)
Absolute risk: 0–100%Absolute risk: 0–100%Absolute risk: 0–100%Low risk (score 0–1); (0.3 and 1.6%)c
Intermediate risk (score 2–3); (7.1 and 17%)c
High risk (score >3); (>17%)c
Outcome30 day MI, cardiac arrest, deathIntra-operative and 30 day MI or cardiac arrestSerious complications and any complications at 30 days30 day mortality30 day death, MI, or stroke
Derivation population1422211 4101 414 00611 2193284
Validation populationExternally validated in various surgical populations257 385Externally validated in various surgical populations22 6311 167 414
Model performance (AUC)0.68–0.760.81–0.850.730.81–0.920.82
Interactive calculatorhttps://www.mdcalc.com/revised-cardiac-risk-index-pre-operative-riskhttp://www.surgicalriskcalculator.com/miorcardiacarresthttps://riskcalculator.facs.orghttp://www.sortsurgery.com
Revised Cardiac Risk Index (RCRI) (1999)aSurgical Risk Calculator (2011)The American College of Surgery National Surgical Quality Improvement Program (ACS NSQIP) (2013)Surgical Outcome Risk Tool (SORT) (2014)The American University of Beirut (AUB)-HAS2 Cardiovascular Risk Index (2019)b
VariablesIschaemic heart disease
Cerebrovascular disease
History of congestive heart failure
Insulin therapy for diabetes
Serum creatinine level ≥2 mg/dL
High-risk surgery
(each assigned 1 point)
Age
ASA–PS grade
Pre-operative dependent functional status
Creatinine >1.5 mg/dL
Type of surgery
Age
Sex
Functional status
Emergency case
ASA class
Current steroid use
Ascites within 30 days
Systemic sepsis within 48 h
Ventilator dependence
Disseminated cancer
Diabetes
Hypertension on treatment
Congestive HF
Dyspnoea
Current smoker
History of severe COPD
Dialysis
Acute renal failure
Body mass index
Surgery code
ASA–PS grade
Urgency of surgery
High-risk surgical specialty
Surgical severity (from minor to complex major)
Cancer
Age ≥65 years or over
History of Heart disease
Symptoms of Heart disease (angina or dyspnoea)
Age ≥75 years
Anaemia (haemoglobin <12 g/dL)
Vascular Surgery
Emergency Surgery
(2 H, 2 A and 2 S)
(each assigned 1 point)
Score rangeScore 1; risk 6.0% (4.9–7.4)
Score 2; risk 10.1% (8.1–10.6)
Score ≥3; risk 15% (11.1–20.0)
Absolute risk: 0–100%Absolute risk: 0–100%Absolute risk: 0–100%Low risk (score 0–1); (0.3 and 1.6%)c
Intermediate risk (score 2–3); (7.1 and 17%)c
High risk (score >3); (>17%)c
Outcome30 day MI, cardiac arrest, deathIntra-operative and 30 day MI or cardiac arrestSerious complications and any complications at 30 days30 day mortality30 day death, MI, or stroke
Derivation population1422211 4101 414 00611 2193284
Validation populationExternally validated in various surgical populations257 385Externally validated in various surgical populations22 6311 167 414
Model performance (AUC)0.68–0.760.81–0.850.730.81–0.920.82
Interactive calculatorhttps://www.mdcalc.com/revised-cardiac-risk-index-pre-operative-riskhttp://www.surgicalriskcalculator.com/miorcardiacarresthttps://riskcalculator.facs.orghttp://www.sortsurgery.com

AUC, area under curve; ASA–PS, American Society of Anesthesiology Physical Status; COPD, chronic obstructive pulmonary disease; HF, heart failure; MI, myocardial infarction; RCRI, Revised Cardiac Risk Index.

a

The RCRI was updated January 2019.

b

Dakik et al. 2019, 2020, and 2022, and Msheik et al.49–51,81

c

The percentages relate to general surgeries.50

The RCRI estimates the risk of 30 day mortality, MI, or cardiac arrest, and is based on six variables.46,47 It has been validated in several countries and is easy to use.47 A score of 0 indicates a 4% risk of 30 day mortality, MI, or cardiac arrest; a score of 1 indicates a 6% risk; a score of 2, 10%; and a score of ≥3, 15%.47

The American College of Surgery National Surgical Quality Improvement Program (ACS NSQIP) developed an interactive risk calculator providing an estimate of the absolute 30 day probability of serious complications and any complications compared with the average patient.76 Evaluated in the US surgical database, the ACS NSQIP model performed better than the RCRI, but an external validation in the Philippines found both to have excellent discriminative abilities for predicting any MACE.48 The RCRI can be used without a web connection, whereas the ACS NSQIP is procedure-specific and is only available on the web. For clinical use, the RCRI is more accessible, but the ACS NSQIP offers procedure-specific absolute risk estimates, which are valuable in patient-guided decision-making. In vascular surgery, both risk calculators have shown moderate accuracy with an area under curve (AUC) of 0.64 (95% CI, 0.57–0.70) for ACS NSQIP and 0.60 (95% CI, 0.54–0.65) for RCRI, due to underestimation of the risk of MI.76a Attempts to generate procedure-specific vascular calculators have not given better predictions in validation cohorts.77

The Surgical Outcome Risk Tool (SORT) estimates 30 day mortality after NCS based on the American Society of Anesthesiologists Physical Status (ASA–PS) grade, urgency of surgery, surgical speciality and severity, cancer, and age ≥65 years. In the validation study, combining subjective assessment with the SORT was significantly better than using either alone.78,79 The Surgical Risk Calculator is another tool that predicts intra-operative and 30 day risk of MI or cardiac arrest based on age, ASA–PS grade, pre-operative dependent functional status, creatinine, and type of surgery.80

The American University of Beirut (AUB)-HAS2 Cardiovascular Risk Index is the most recently developed index to assess 30 day event risk (death, MI, or stroke), and stratifies patients undergoing NCS into low (score 0–1), intermediate (score 2–3), and high risk (score >3) based on six data elements (see Table 6); scores >3 denote a post-operative event rate of >10%.49 The AUB-HAS2 Index has been tested in a broad spectrum of surgical subpopulations and demonstrated superior discriminatory power compared with the commonly utilized RCRI (Table 6).50,51,81

There is significant variability in the predicted risk of cardiac complications using different risk-prediction tools; none can be disqualified with current evidence.82

4.1.2. Frailty

Frailty is an age-related, multidimensional state of decreased physiological reserve that results in diminished resiliency, loss of adaptive capacity, and increased vulnerability to stressors.83,84 The peri-operative evaluation of elderly patients (>70 years) who require elective intermediate- or high-risk NCS should include frailty screening, which has proven to be an excellent predictor of unfavourable health outcomes in the older surgical population.

Frailty has a relevant impact on mortality and MI risk but does not add to risk estimation derived from the ACS NSQIP calculator, as frailty is associated with variables already in the main model. By adding six variables, the ACS NSQIP predicts the risk of post-operative delirium, functional decline, need of a new mobility aid, or pressure ulcer.85 The use of this broader-inclusive score identifies cases that profit the most by involving a geriatrician in the pre- and post-operative team.86,87 A measure of frailty informs the patient and surgeon about further life expectancy and the chance of post-operative delirium, dependency of mobility support, and need of nursing home or other care support after planned surgery.

Of the available screening tools for frailty, the Frailty Index and the Frail Phenotype are the most commonly recommended.88,89 Of note, the Frailty Index includes cognitive testing, while both scores assess physical function.90,91 A simpler approach is offered by the Clinical Frailty Scale, which relies on information from the history taking. The Clinical Frailty Scale has been validated against the Frailty Index.88 For cognitive screening to incorporate with the Frailty Index, Mini-Cog© is a simple and fast screening tool validated for pre-operative screening92 (Supplementary data, Figure S1, and Tables S2 and S3).

Once a diagnosis of frailty is confirmed, the prognosis of a frail patient can be improved by shared decision-making between at least a treating physician (e.g. surgeon), anaesthesiologist, geriatrician, the patient, and the patient’s relatives. During the shared decision-making process, a careful discussion with a frail patient about goals of care could help them to have realistic expectations and make better informed decisions before surgery. After a shared decision to go ahead with a planned NCS, multimodal pre-habilitation programmes—including exercise, nutrition, and psychological interventions—could potentially improve the peri-operative prognosis of frail patients by an individualized approach tailored to the patient’s baseline functional status, comorbidities, and cognitive/psychological function.90

4.2. Functional capacity

Quantifying functional capacity has been a pivotal step in pre-operative cardiac risk assessment.10 Although the validity of interview-based assessment of functional capacity has been questioned,93 a recent large prospective cohort study of high-risk patients undergoing NCS found self-reported inability to climb two flights of stairs added incremental value to the 30 day cardiac event rate when added to the RCRI.94

Metabolic equivalents (METs) <4 have long been considered to indicate poor functional capacity; however, studies using METs have been based on subjective interviews and not shown proven value. In the Measurement of Exercise Tolerance before Surgery (METS) study, the Duke Activity Status Index (DASI) (https://www.mdcalc.com/duke-activity-status-index-dasi#evidence) had a more precise estimation of cardiac risk than subjectively assessed functional capacity, improving risk estimation using RCRI.95 A DASI score <34 was associated with increased odds of 30 day death or MI.96 From the DASI score, METs can be calculated as VO2 max (maximal oxygen consumption)/3.5; where VO2 max (mL/kg/min) = 0.43 × DASI + 9.6. Furthermore, cardiopulmonary exercise testing (CPET) did not predict 30 day mortality, post-operative MI, or cardiac arrest.94,95 It should be noted that a relatively low number of primary outcome events limited the statistical power of the analysis.

Recommendation Table 6

Recommendations for pre-operative assessment of frailty and functional capacity

graphic
graphic
graphic
graphic
Recommendation Table 6

Recommendations for pre-operative assessment of frailty and functional capacity

graphic
graphic
graphic
graphic

4.3. Electrocardiography

The 12-lead ECG is a widely available, simple, and inexpensive tool that is able to semi-quantitatively assess cardiac risk (e.g. Q waves indicative of previous MI), and detect unknown CV conditions requiring therapy (e.g. atrial fibrillation [AF] or AV-block).97–99 It is recommended to obtain a pre-operative 12-lead ECG in patients who are aged ≥65 years or have known CVD, CV risk factors, or symptoms suggestive of cardiac disorders, and scheduled to undergo intermediate- or high-risk surgery. It is not recommended to routinely obtain a pre-operative ECG in low-risk patients undergoing low-risk NCS.100

Comparison with previous ECG recordings is helpful whenever relevant abnormalities are identified. Pre-operative recording of ECG also enables identification of intra- and post-operative ECG changes.

4.4. Biomarkers

As the peri-operative risk for cardiac complications depends on the presence and extent of cardiac disease, widely available and simple biomarkers that detect and quantify essential prognostic aspects of cardiac involvement may aid in the evaluation. High-sensitivity cardiac troponin T/I (Hs-cTn T/I) quantifies myocardial injury, and BNP and NT-proBNP quantify haemodynamic cardiac wall stress (Figure 4). Both Hs-cTn T/I and BNP/NT-proBNP complement clinical assessment and ECG in risk prediction.9,52,53,101–103 Hs-cTn T/I and, to a lesser extent, BNP/NT-proBNP concentrations are higher in patients with stress-induced myocardial ischaemia vs. those without, and very low hs-cTn T/I concentrations achieve a very high negative predictive value to rule out myocardial ischaemia.104–107

Recommended measurements to assess and detect the risk of post-operative cardiac complications.
Figure 4

Recommended measurements to assess and detect the risk of post-operative cardiac complications.

ECG, electrocardiogram; hs-cTn, high-sensitivity cardiac troponin; PMI, peri-operative myocardial infarction/injury; ULN, upper limit of normal. In patients scheduled to undergo intermediate- or high-risk surgery, pre-operative risk assessment is complemented by ECG, hs-cTn, and BNP/NT-proBNP. An absolute increase in hs-cTn concentration of more than the ULN on days 1 or 2 after surgery compared to the pre-operative level is defined as PMI.109–111 In the absence of a pre-operative hs-cTn T/I concentration, a very high hs-cTn T/I concentration on day 1 (e.g. more than five-times the ULN) or a relevant change from day 1 to day 2 (absolute increase or decrease more than the ULN vs. day 1) would also achieve a reliable diagnosis of PMI. Detection of PMI should trigger ECG recording and detailed clinical evaluation for PMI work-up and therapy. The differential diagnosis of PMI according to the fourth universal definition of MI is discussed in Section 8. The ESC 0/1/2 h algorithm has not been validated for the peri-operative setting and cannot be used here.

Several large prospective studies have shown that both hs-cTn T/I and BNP/NT-proBNP have high and incremental prognostic value for peri-operative cardiac complications, including CV death, cardiac arrest, acute HF, and tachyarrhythmias. In a cohort of nearly 1000 subjects undergoing major elective NCS, individuals with pre-operative hs-cTn T concentrations of >14 ng/L had an in-hospital mortality of 6.9% vs. 1.2% in patients with hs-cTn T concentrations ≤14 ng/L (P < 0.001; AUC 0.81).53 In a large prospective cohort study including 10 402 patients from 16 centres, NT-proBNP improved risk predication beyond the RCRI.52 Among 1923 patients undergoing NCS, NT-proBNP outperformed both RCRI and echocardiographic parameters in the prediction of peri-operative CV events.103 Overall, hs-cTn T/I and BNP/NT-proBNP seem to have comparable accuracy in the prediction of cardiac complications.52,53,98–103,108 However, Hs-cTn T/I has four advantages over BNP/NT-proBNP: (i) it is more widely available; (ii) it is less expensive; (iii) if normal, it enables acute MI to be ruled out in the preceding days; and (iv) availability of pre-operative hs-cTn T/I concentration enables accurate diagnosis of PMI on Day 1 after surgery.109–111 See Section 8 for more details on diagnosis and treatment of PMI.

B-type natriuretic peptide/NT-proBNP has two advantages. First, if elevated, evidence from randomized controlled screening studies performed outside the peri-operative setting has supported the concept that BNP/NT-proBNP-triggered cardiac work-up and intensification of therapy improve outcomes.112,113 Second, HF is a frequently undiagnosed condition in the elderly population most often undergoing NCS.47,114 Interpreting BNP/NT-proBNP concentrations as quantitative markers of HF with evolving rule-in cut-offs may facilitate detection of HF, optimal intra-operative monitoring, and initiation or optimization of HF therapy after surgery.114

To date, there is insufficient evidence in support of other CV biomarkers for this specific indication.115,116

Recommendation Table 7

Recommendations for pre-operative risk assessment—electrocardiography and biomarkers

graphic
graphic
graphic
graphic
Recommendation Table 7

Recommendations for pre-operative risk assessment—electrocardiography and biomarkers

graphic
graphic
graphic
graphic

4.5. Non-invasive and invasive procedures

4.5.1. Resting transthoracic echocardiography

In large retrospective cohorts, routine pre-operative TTE before high-risk NCS did not reduce the risk of post-operative MACE or provide more information than clinical risk models.120–122 Poor exercise tolerance, abnormal ECG, suspected new or significant CVDs without follow-up within the last 90 days, unexplained dyspnoea, or coexisting clinical risk factors are appropriate indications for TTE.123,124 Pre-operative TTE provides information on three main risk markers for post-operative cardiac events: LV dysfunction, VHDs, and cardiomyopathies. Left ventricular systolic dysfunction is an important predictor of post-operative HF.125 However, low ventricular ejection fraction is a borderline independent predictor of major post-operative CV complications.126–128

Pre-operative FOCUS examination—with a hand-held ultrasound device for the assessment of murmurs, haemodynamic instability, ventricular function, and dyspnea—may impact patient management by improving the diagnostic accuracy of clinical assessment, and help to triage candidates for standard TTE, plan surgery and anaesthesia technique, and with post-operative monitoring.129–131 However, current evidence remains mostly confined to uncontrolled or retrospective observational studies with no clear benefits on the outcome, despite a favourable impact on peri-operative management.130,132 In a multicentre randomized trial, preliminary results showed that pre-operative FOCUS significantly reduced all-cause mortality.133 Notably, because of the lack of spectral Doppler capabilities, the FOCUS examination is only accurate for assessing main structural and functional abnormalities.

Patients with diastolic dysfunction are usually old, more hypertensive, obese, diabetic, and likely to have AF or chronic renal disease. Several studies with different clinical end-points have underlined the association of diastolic dysfunction with post-operative adverse events, including pulmonary oedema, AF, and mortality.134–138 A meta-analysis including 3876 patients undergoing NCS found pre-operative diastolic dysfunction to be an independent risk factor for pulmonary oedema, congestive HF, and MI after surgery.139 However, a recent retrospective study, including 7312 patients, showed no association between the degree of diastolic dysfunction and in-hospital mortality or hospital length of stay in NCS patients.140 Awareness of diastolic dysfunction or high LV filling pressure (e.g. pulmonary hypertension, left atrial volume, E/e′ ratio) seems necessary to optimize peri-operative patient management; however, evidence does not support screening for diastolic dysfunction.

Recommendation Table 8

Recommendations for transthoracic echocardiography

graphic
graphic
Recommendation Table 8

Recommendations for transthoracic echocardiography

graphic
graphic

4.5.2. Stress tests

4.5.2.1. Exercise stress test

Physical exercise, using a treadmill or bicycle ergometer, provides an estimate of functional capacity, evaluates blood pressure (BP) and heart rate response, and detects myocardial ischaemia through pathological ST-segment changes with poor sensitivity (61–73%) and specificity (60–80%).146 An exercise stress test alone should only be considered a valuable alternative to diagnose obstructive CAD if non-invasive imaging tests are unavailable.146 An exercise stress test is of no diagnostic value in patients with pre-existing ST-segment abnormalities (i.e. left bundle branch block, paced rhythm, Wolff–Parkinson–White [WPW] syndrome, ≥0.1 mV ST-segment depression on resting ECG, or taking digitalis). In addition, an exercise test is unsuitable for patients with limited exercise capacity, owing to their inability to reach their target heart rate. Therefore, an exercise stress test alone should only be considered a valuable alternative to diagnose obstructive CAD if non-invasive imaging tests are unavailable, or for assessing functional capacity when clinical history is ambiguous.146

4.5.2.2. Stress imaging

The use of stress imaging is appropriate for risk assessment in patients with clinical risk factors and poor functional capacity.147,148 The choice of the test is driven by local expertise. Selection, optimal, and safe performance of stress imaging should comply with related guidelines and recommendations.146,148,149 Stress imaging is not recommended in patients undergoing urgent surgery or with an unstable clinical condition. Evidence on the role of stress imaging for peri-operative risk prediction and patient management is largely based on inducible ischaemia by pharmacological stress testing, although no evidence indicates the superiority of pharmacological stress to exercise stress imaging in patients who are able to perform an adequate level of physical exercise. Several studies and meta-analyses have consistently defined clinical utility of pharmacological stress imaging for peri-operative risk assessment in patients undergoing NCS.150–154 Although RCTs related to post-operative outcome are lacking, there are large-scale prospective studies showing a risk-adjusted association of stress imaging results with peri-operative cardiac complications.155–157

Studies and meta-analyses have demonstrated similar prognostic value of stress echocardiography and myocardial perfusion imaging for peri-operative risk assessment with slightly higher negative predictive value of stress echocardiography,152 but the overall accuracy varies with ischaemic heart disease (IHD) prevalence.151 A moderate-to-large perfusion defect on either test is highly sensitive for post-operative cardiac events.152,158 Normal stress imaging exams without resting abnormalities have high negative predictive value.159,160 However, positive predictive value of stress imaging for peri-operative cardiac events is relatively low and requires confirmation by other tests.150,152,161

In a recent retrospective study including 4494 patients, dobutamine stress echocardiography (DSE) provided modest incremental predictive value for peri-operative CV complications over clinical variables and was found to be useful as part of a stepwise approach in the risk stratification of patients undergoing intermediate- to high-risk NCS.157 The strongest predictors of post-operative adverse events determined so far are significant ischaemia (more than four ventricular segments) during DSE, ischaemic threshold (60% of age-predicted maximal heart rate), and a history of congestive HF.162,163

A negative DSE without resting wall motion abnormality has excellent negative predictive value, even when target heart rate cannot be achieved despite an aggressive DSE regimen.164 In asymptomatic patients if functional capacity is unknown, stress echocardiography also enables dynamic evaluation of LV systolic and diastolic function; valvular diseases such as aortic valve stenosis (AS), mitral valve stenosis (MS), and hypertrophic obstructive cardiomyopathy; and pulmonary hypertension 165 However, the role of DSE for risk estimation in non-ischaemic heart diseases before NCS has yet to be studied.

Myocardial perfusion imaging is particularly suitable if patients have poor acoustic windows for DSE. Meta-analyses of patients undergoing major NCS have demonstrated that, compared with fixed defects, reversible perfusion defects were associated with higher risk of cardiac death or non-fatal MI. The risk of cardiac events correlates with the extent of reversible perfusion abnormalities (severe: >20% of myocardium). Normal myocardial perfusion imaging in high-risk patients identifies a low-risk subgroup comparable with those without clinical risk factors for adverse cardiac outcomes.154,159,160

Stress cardiac magnetic resonance (CMR) imaging and late gadolinium enhancement are also accurate tools for detection of IHD and prognostication.166

Recommendation Table 9

Recommendations for stress imaging

graphic
graphic
Recommendation Table 9

Recommendations for stress imaging

graphic
graphic

4.5.3. Angiography

4.5.3.1. Coronary computed tomography angiography

Coronary computed tomography angiography (CCTA) is recommended as an initial test for diagnosing CAD in stable patients with a low clinical likelihood or no previous diagnosis of CAD, and characteristics associated with a high likelihood of good image quality.146 In addition, CCTA is recommended as an alternative to invasive coronary angiography (ICA) for excluding non-ST-segment elevation acute coronary syndrome (NSTE-ACS) when there is low-to-intermediate likelihood of CAD, and when cardiac troponin and/or ECG are normal or inconclusive.98 The practical utility of CCTA is reduced when a high coronary calcium score is present.167

In patients undergoing NCS, the role of pre-operative CCTA to rule out CAD has been investigated in small- to medium-sized observational studies. The Coronary Computed Tomographic Angiography and Vascular Events in Noncardiac Surgery Patients Cohort Evaluation (Coronary CTA VISION) trial prospectively investigated the incremental predictive value of CCTA over RCRI in 955 patients with a history of or risk factors for CAD, or a history of congestive HF undergoing NCS.168 Coronary computed tomography angiography improved the risk estimation for the primary outcome of post-operative CV death and non-fatal MI within 30 days, although CCTA was associated with more than five times inappropriate risk overestimations among patients not experiencing the primary outcome. The predictive value of CCTA further improved when associated with non-invasive functional testing, such as myocardial perfusion imaging, with a positive and negative predictive value of 50% (95% CI, 21–79) and 100% (95% CI, 79–100), respectively.161

Coronary computed tomography angiography associated with additional functional assessment of coronary stenosis with fractional flow reserve (FFR) with computed tomography (CT) was able to identify functionally severe coronary stenosis in 57% of the asymptomatic patients with no history of cardiac disease undergoing carotid endarterectomy (CEA).169 In 135 asymptomatic patients with no history of cardiac disease undergoing peripheral vascular surgery, pre-operative FFR with CT facilitated the identification of functionally severe coronary stenosis in 53% of the patients. These patients benefited from further revascularization, with a 1 year lower rate of CV death and MI.170

4.5.3.2. Invasive coronary angiography

There is a lack of information from RCTs relating to the usefulness of ICA in patients scheduled for NCS. Adopting an ICA assessment may also cause an unnecessary and unpredictable delay in an already planned surgical intervention, and adding an independent procedural risk to the overall risk. Despite the fact that CAD may be present in a significant number of patients requiring NCS, indications for pre-operative coronary angiography and revascularization are similar to angiography indications in the non-surgical setting.98,146,171 Pre-operative treatment of patients with myocardial ischaemia, either medically or with intervention, is recommended.

Recommendation Table 10

Recommendations for coronary angiography

graphic
graphic
graphic
graphic
Recommendation Table 10

Recommendations for coronary angiography

graphic
graphic
graphic
graphic

5. General risk-reduction strategies

5.1. Cardiovascular risk factors and lifestyle interventions

Control of CV risk factors—including BP, dyslipidaemia, and diabetes—is important before NCS. For pre-operative management of BP and diabetes, see Sections 6.8 and 6.13, respectively.

While lifestyle modifications before intervention reduce the risk of several peri-operative complications, the impact on CV complications has not been adequately explored. Of the lifestyle changes recommended before surgery, smoking cessation is the best documented in RCTs. Smoking has been associated with a higher rate of post-operative complications at 30 days.173,174 Reviews of RCTs have shown an effect of smoking cessation up to 6 months post-operatively, with a clear reduction in any post-operative complications by hazard ratio (HR) 0.42 (95% CI, 0.27–0.65), particularly wound infections (HR, 0.43; 95% CI, 0.21–0.85).173,175 Regarding timing of cessation, reviews of observational studies have shown consistent associations with better surgical outcome for cessation >4 weeks before surgery, with each additional week resulting in a further improvement of 19%.176–178

Pre-operative exercise programmes have only been tested in small RCTs, and recent reviews have shown a relative risk (RR) reduction in post-operative complications of 67% (RR, 0.33; 95% CI, 0.17–0.61).179 Referral to a pre-operative exercise programme may be considered for patients scheduled for major or complex elective surgery.176,179,180 Weight reduction of obese patients immediately prior to surgery is not recommended.

Recommendation Table 11

Recommendations for lifestyle and cardiovascular risk factors

graphic
graphic
Recommendation Table 11

Recommendations for lifestyle and cardiovascular risk factors

graphic
graphic

5.2. Pharmacological

5.2.1. Beta-blockers

Beta-blockers reduce myocardial oxygen consumption by reducing contractile force and heart rate. Beta-blockers are also effective antiarrhythmic agents. In addition, some beta-blockers such as metoprolol have an effect on acute inflammatory responses by inhibiting neutrophil hyperactivation in acute settings.184 These properties mean that beta-blockers have been some of the most frequently tested cardioprotective agents in patients undergoing NCS. Several RCTs have evaluated the effects of peri-operative beta-blockade on clinical end-points in patients with different risk profiles (see Supplementary data, Section 3.1.1.). Type, dosing and titration, timing of initiation, duration of beta-blocker therapy, type of surgery, and risk profile of subjects significantly differ between studies, making comparisons complex.

The question about pre-surgery initiation of beta-blockers has been a matter of intense controversy (see Supplementary data, Section 3.1.1.1). The largest and latest trial on the topic, the Perioperative Ischemic Evaluation (POISE-1) trial, enrolled 8351 patients with or at risk of atherosclerotic disease, and not on beta-blockers before NCS. Patients were randomized to extended-release metoprolol succinate 200 mg daily or placebo 185 Treatment was initiated 2–4 h before surgery and maintained for 30 days. The primary outcome (composite of CV death, non-fatal MI, and non-fatal cardiac arrest) was significantly lower in the metoprolol arm (5.8% vs. 6.9% [P = 0.04]). Metoprolol was associated with significant reductions in MIs, coronary revascularizations, and AF. However, the incidences of all-cause death, stroke, and clinically significant hypotension or bradycardia were significantly higher in the metoprolol arm. Post hoc analysis showed that hypotension carried the greatest attributable risk of death and stroke 186 The high dose of extended metoprolol might have played a role in the adverse events seen at follow-up.

Several meta-analyses, systematic reviews, and observational studies have also been reported (see Supplementary data, Table S4).187–189 Overall, initiation of beta-blockers before NCS was not associated with a net clinical benefit in most analyses, but they might be beneficial in patients with high CV risk profiles or who are undergoing high-risk surgical interventions (including vascular interventions).188,190–192 When oral beta-blockade is initiated in CAD patients who undergo NCS, the use of atenolol or bisoprolol as a first choice may be considered.190,193–195

In patients who are on chronic beta-blocker therapy before surgery, it is recommended to maintain these in the peri-operative period. Increased mortality following pre-operative beta-blocker withdrawal has been reported in five observational studies.190,196–199 Interruption of this therapy for >2 days post-operatively may double the risk of AF.200

Post-operative tachycardia should initially lead to treatment of the underlying cause— such as hypovolaemia, pain, blood loss, or infection—rather than simply increasing the beta-blocker dose. When beta-blockers are indicated, the optimal duration of the peri-operative beta-blockade cannot be derived from randomized trials.

According to a meta-analysis of RCTs including 14 967 patients, beta-blockers can reduce the risk of post-operative AF after NCS;201 however, this comes at the cost of an increased risk of bradycardia, hypotension, and stroke.187

The ultra-short-acting beta-blockers esmolol and landiolol have the theoretical advantages of very fast onset of effects and short half-lives. Notably, landiolol may lower BP to a lower extent than esmolol. Evidence of prevention of AF with landiolol after NCS is less robust and inconsistent than in the context of cardiac surgery.202–205 The timing of the initiation of beta-blockers to prevent AF remains unclear, with most prophylactic regimens using short-acting agents being started intra-operatively.187

5.2.2. Amiodarone

Amiodarone is the most frequently used agent for prevention of post-operative AF,206 with a risk reduction of 58% in NCS in a meta-analysis evaluating different antiarrhythmic drugs (AADs), but may induce relevant non-cardiac side effects.201 In another meta-analysis, amiodarone (oral or intravenous [i.v.]) and beta-blockers were equally effective in reducing post-operative AF.207 In another prospective RCT, a combination of beta-blocker plus amiodarone outperformed beta-blockers alone in reducing post-operative AF.208 It should be noted that the two latter studies were undertaken in patients undergoing cardiac surgery.

Overall, while preventive amiodarone seems to reduce the incidence of AF, the clinical benefits associated with its routine use are unclear.

5.2.3. Statins

Despite the wide-ranging use of statins in patients undergoing surgery, RCTs assessing the effects of initiating statin therapy during the peri-operative period are scarce. This should be viewed separately from patients already on statin therapy. The long-term use of statins in patients with CVD or high risk of CVD is well-established.40

Observational data suggest a potential benefit of statins in the peri-operative phase. In a large, retrospective, and observational cohort, which included 180 000 veterans undergoing NCS, the use of statins on the day of or the day after surgery was associated with a reduction in mortality (RR, 0.82; 95% CI, 0.75–0.89).209 Similar results were seen in a retrospective cohort study using hospital discharge and pharmacy records.210 Although both studies used propensity matching to reduce bias, these analyses are prone to confounding, especially when discharge and pharmacy records are used. As such, RCTs give a more reliable effect estimation, for example: the Lowering the Risk of Operative Complications Using Atorvastatin Loading Dose (LOAD) trial studied 648 statin-naïve patients, of whom 24% had a history of CVD and 49% had diabetes.211 In this randomized, placebo-controlled trial, patients received a loading dose of atorvastatin 80 mg within 18 h before surgery followed by 40 mg daily for 7 days. Use of atorvastatin did not reduce the risk of major events (all-cause mortality, non-fatal MI, or stroke at 30 days [HR, 0.87; 95% CI, 0.60–1.26; P = 0.46]). However, the trial was underpowered to draw definite conclusions. In addition, several meta-analyses have shown ambiguous results and most studies are of a limited size with less than 100 patients.212,213

Therefore, routine peri-operative initiation of statin therapy is not recommended. However, in patients in whom statin use is already indicated, treatment should be considered peri-operatively, particularly in patients scheduled for high-risk surgery (e.g. vascular surgery).

5.2.4. Renin–angiotensin–aldosterone system inhibitors

Data on peri-operative use of renin–angiotensin–aldosterone system (RAAS) inhibitors are inconclusive. The majority of studies suggest that continued use of RAAS inhibitors is associated with a higher risk of peri-operative hypotension and, as a consequence, higher use of vasopressors and inotropes. Furthermore, intra-operative hypotension and its duration is associated with end-organ damage, including kidney injury, myocardial damage, and stroke.214 In a small trial of 275 subjects, randomized to either continuation of their angiotensin-converting-enzyme inhibitors (ACEIs) or omission of the final pre-operative ACEI dose, patients randomized to omission of the last dose before surgery experienced intra-operative hypotension less frequently (76/137 [55%] vs. 95/138 [69%]) and vasopressor use was less likely.215 On the other hand, post-operative hypertension was more frequent in the omission group. Furthermore, in an observational cohort study consisting of 4802 patients undergoing NCS and using an ACEI or angiotensin receptor blocker (ARB), discontinuation of these drugs in the 24 h before surgery was associated with a lower risk of intra-operative hypotension (adjusted RR, 0.80; 95% CI, 0.72–0.93; P < 0.001), and associated with a reduction in the composite end-point consisting of all-cause mortality, stroke, and MI (adjusted RR, 0.82; 95% CI, 0.70–0.96; P = 0.01);216 8% of the patients in this cohort were diagnosed with HF, in whom RAAS inhibitors are the cornerstone of medical therapy. A systematic review, including nine studies (five RCTs and four cohort studies), revealed that withholding ACEI/ARB on the morning of surgery was not associated with mortality or MACE;217 however, it did confirm that withholding therapy was associated with less intra-operative hypotension (OR, 0.63; 95% CI, 0.47–0.85). If an ACEI/ARB is withheld prior to NCS, it should be restarted as soon as possible in order to prevent unintended long-term omission. No data on peri-operative effects of angiotensin receptor neprilysin inhibitors (ARNI) exist, but hypotension is more common compared with patients on ACEI.218

Some important RCTs in this field are ongoing: the impact of renin–angiotensin system inhibitors continuation vs. discontinuation on outcome after major surgery trial STOPorNOT219 (NCT03374449), and the POISE-3 trial (NCT03505723) are both assessing a hypotension-avoidance strategy vs. a hypertension-avoidance strategy on the risk of vascular death and major vascular events in patients who are followed for 30 days after NCS.

5.2.5. Calcium channel blockers

The effects of calcium channel blockers (CCBs) on the balance between myocardial oxygen supply and demand makes them theoretically suitable for risk-reduction strategies. The relevance of randomized trials assessing the peri-operative effects of CCBs is limited by their small size, lack of risk stratification, and the absence of systematic reporting of cardiac death and MI. A meta-analysis pooled 11 randomized trials totalling 1007 patients.220 Treatment with CCBs significantly reduced the number of episodes of myocardial ischaemia and supraventricular tachycardia (SVT) in the pooled analyses. However, the decrease in mortality and MI reached statistical significance only when both end-points were combined (RR, 0.35; 95% CI, 0.08–0.83; P = 0.02). In contrast, a matched case-control study of 1000 patients undergoing acute or elective aortic aneurysm surgery suggested that dihydropyridine use was independently associated with an increased incidence of peri-operative mortality.221 These observational data may be biased by the indications for the use of CCBs. In patients already on CCBs, particularly in those with vasospastic angina, it is recommended to continue CCBs during the peri-operative period, but withholding the dose on the day of surgery in order to avoid post-operative hypotension.

5.2.6. Alpha-2 receptor agonists

Alpha-2 receptor agonists reduce post-ganglionic noradrenaline output and might therefore reduce catecholamine surge during surgery. The European Mivazerol trial randomized 1897 patients with IHD who underwent intermediate- or high-risk NCS.222 Mivazerol did not decrease the incidence of death or MI in the whole population. However, it did decrease the incidence of death in a subpopulation of 904 patients undergoing vascular surgery.222 The international Peri-Operative ISchemic Evaluation 2 (POISE-2) trial randomized 10 010 patients undergoing NCS to clonidine or placebo.223 Clonidine did not reduce the rate of death or non-fatal MI in general or in patients undergoing vascular surgery (RR, 1.08; 95% Cl, 0.93–1.26; P = 0.29), but it did increase the risk of clinically important hypotension (RR, 1.32; 95% Cl, 1.24–1.40; P < 0.001) and non-fatal cardiac arrest (RR, 3.20; 95% Cl, 1.17–8.73; P = 0.02).

5.2.7. Diuretics

Diuretics are frequently used in patients with hypertension or HF. In general, therapy for treatment of hypertension should be continued to the day of surgery and resumed orally when possible. However, the benefit for continuing diuretics as antihypertensive therapy is unclear, and alternative antihypertensive agents may be considered. In HF, the dosage of diuretics should be adjusted well in advance for an optimal fluid balance before surgery, and to avoid fluid retention or dehydration.

The possibility of electrolyte disturbance should be considered in any patient receiving diuretics. Hypokalaemia is reported to occur in up to 36% of patients undergoing surgery (mostly NCS).224,225 Special attention should be given to patients prone to developing arrhythmias. Any electrolyte disturbance, especially hypokalemia and hypomagnesaemia, should be corrected in due time before surgery. Acute pre-operative repletion in asymptomatic patients may be associated with more risks than benefits; thus, minor asymptomatic electrolyte disturbances should not delay acute surgery.

In the peri-operative period, volume status in patients with HF should be carefully monitored and optimized by loop diuretics or fluids. However, retrospective data suggest that intra-operative prescription of diuretics may increase the risk of acute kidney injury (AKI) after NCS.226

5.2.8. Ivabradine

Heart rate is an independent and modifiable risk factor for periprocedural MI (and maybe death) after NCS. Ivabradine is a negative chronotropic agent without associated hypotensive effect, and is therefore a possible alternative to beta-blockers. However, there are few studies about the value of ivabradine for high-risk patients undergoing NCS.227 The small (78 patients) PeRi-OperaTivE CardioproTection With Ivabradine in Non-cardiac Surgery (PROTECTIN) (NCT04436016) trial is ongoing.

5.2.9. Sodium–glucose co-transporter-2 inhibitors

The use of sodium–glucose co-transporter-2 (SGLT-2) inhibitors is increasing, due to proven CV benefits for patients with type-2 diabetes mellitus (DM) and a beneficial effect on outcomes for patients with HF and renal insufficiency. Euglycaemic diabetic ketoacidosis (EDKA) is a rare but serious complication. While the incidence was not significantly increased with SGLT-2 inhibitors in RCTs, several case reports indicate that EDKA may occasionally occur after (non-cardiac) surgery in patients on SGLT-2 inhibitors.228 A systematic review indicated that precipitating factors include diabetes medication changes, diet modifications, and intercurrent illnesses.229 The US Food and Drug Administration (FDA) recommends interrupting SGLT-2 inhibitor therapy for at least 3–4 days before scheduled surgery and to be vigilant for symptoms related to EDKA, prompting measurement of ketones.

Recommendation Table 12

Recommendations for pharmacological treatment

graphic
graphic
graphic
graphic
Recommendation Table 12

Recommendations for pharmacological treatment

graphic
graphic
graphic
graphic

5.3. Peri-operative handling of antithrombotic agents

Management of patients taking antithrombotic agents and needing surgery or an invasive procedure should consider patient- and procedure-related risk of bleeding and thrombosis. Furthermore, the pharmacokinetic and pharmacodynamic characteristics of the antithrombotic drugs in use must be considered (Tables 7 and 8). The risk of bleeding associated with different types of interventions is shown in Table 9. Risk estimation and decision-making in patients requiring long-term antithrombotic therapy is challenging, since relevant associations exist between peri-operative antithrombotic management, bleeding, thrombotic events (MI and stroke), and mortality.6,11–13 Thus, interdisciplinary risk assessment ahead of the intervention is crucial, in order to classify the patient-related ischaemic and bleeding risks (e.g. cardiologist, neurologist, vascular specialist, and haematologist), and the surgical risk (surgeon and anaesthesiologist). Information on timing of intervention by indicated duration of antithrombotic therapy should be communicated with the patient and treating general physician.

Table 7

Pharmacokinetic and pharmacodynamic characteristics of antiplatelets

ASAClopidogrelPrasugrelTicagrelorCangrelorEptifibatideTirofiban
Target
(type of blockade)
COX-1
(irreversible)
P2Y12
(irreversible)
P2Y12
(irreversible)
P2Y12
(reversible)
P2Y12
(reversible)
GPIIB/IIIa
(reversible)
GPIIB/IIIa
(reversible)
ApplicationOralOralOralOrali.v.i.v.i.v.
Time to Cmax0.5–1.0 h2 h
(after 600 mg LD)a
0.5 h
(after 60 mg LD)a
0.5 h
(after 180 mg LD)a
2 min5 min5 min
ProdrugNoYesYesNoNoNoNo
Bioavailability (%)∼50∼508036100100100
Drug interactionsNSAIDs
(in particular ibuprofen + naproxen)
CYP3A4, CYP3A5, or CYP2C19 inhibitors or inducersCYP3A4/A5 and CYP2B6 inhibitorCYP3A4 inducers or inhibitorsNoneNoneNone
Plasma half-life20 min0.5–1 h (active metabolite)0.5–1 h (active metabolite)6–12 h3–6 min2.5–2.8 h1.2–2 h
Duration of action after last dose7–10 days3–10 daysb7–10 daysb3–5 days1–2 h4 h8 h
Renal clearance of the active metabolite (%)NRNRNRNR58∼5065
Dose regimeno.d.o.d.o.d.b.i.d.Bolus, infusionBolus, infusionBolus, infusion
ASAClopidogrelPrasugrelTicagrelorCangrelorEptifibatideTirofiban
Target
(type of blockade)
COX-1
(irreversible)
P2Y12
(irreversible)
P2Y12
(irreversible)
P2Y12
(reversible)
P2Y12
(reversible)
GPIIB/IIIa
(reversible)
GPIIB/IIIa
(reversible)
ApplicationOralOralOralOrali.v.i.v.i.v.
Time to Cmax0.5–1.0 h2 h
(after 600 mg LD)a
0.5 h
(after 60 mg LD)a
0.5 h
(after 180 mg LD)a
2 min5 min5 min
ProdrugNoYesYesNoNoNoNo
Bioavailability (%)∼50∼508036100100100
Drug interactionsNSAIDs
(in particular ibuprofen + naproxen)
CYP3A4, CYP3A5, or CYP2C19 inhibitors or inducersCYP3A4/A5 and CYP2B6 inhibitorCYP3A4 inducers or inhibitorsNoneNoneNone
Plasma half-life20 min0.5–1 h (active metabolite)0.5–1 h (active metabolite)6–12 h3–6 min2.5–2.8 h1.2–2 h
Duration of action after last dose7–10 days3–10 daysb7–10 daysb3–5 days1–2 h4 h8 h
Renal clearance of the active metabolite (%)NRNRNRNR58∼5065
Dose regimeno.d.o.d.o.d.b.i.d.Bolus, infusionBolus, infusionBolus, infusion

ASA, acetylsalicylic acid; b.i.d., twice a day; Cmax, maximum serum concentration; i.v., intravenous; LD, loading dose; NR, non-relevant; o.d., once a day.

a

Time to Cmax for may be delayed by 8 h or more following a dose of opiate.

b

Depending on response status.

Table 7

Pharmacokinetic and pharmacodynamic characteristics of antiplatelets

ASAClopidogrelPrasugrelTicagrelorCangrelorEptifibatideTirofiban
Target
(type of blockade)
COX-1
(irreversible)
P2Y12
(irreversible)
P2Y12
(irreversible)
P2Y12
(reversible)
P2Y12
(reversible)
GPIIB/IIIa
(reversible)
GPIIB/IIIa
(reversible)
ApplicationOralOralOralOrali.v.i.v.i.v.
Time to Cmax0.5–1.0 h2 h
(after 600 mg LD)a
0.5 h
(after 60 mg LD)a
0.5 h
(after 180 mg LD)a
2 min5 min5 min
ProdrugNoYesYesNoNoNoNo
Bioavailability (%)∼50∼508036100100100
Drug interactionsNSAIDs
(in particular ibuprofen + naproxen)
CYP3A4, CYP3A5, or CYP2C19 inhibitors or inducersCYP3A4/A5 and CYP2B6 inhibitorCYP3A4 inducers or inhibitorsNoneNoneNone
Plasma half-life20 min0.5–1 h (active metabolite)0.5–1 h (active metabolite)6–12 h3–6 min2.5–2.8 h1.2–2 h
Duration of action after last dose7–10 days3–10 daysb7–10 daysb3–5 days1–2 h4 h8 h
Renal clearance of the active metabolite (%)NRNRNRNR58∼5065
Dose regimeno.d.o.d.o.d.b.i.d.Bolus, infusionBolus, infusionBolus, infusion
ASAClopidogrelPrasugrelTicagrelorCangrelorEptifibatideTirofiban
Target
(type of blockade)
COX-1
(irreversible)
P2Y12
(irreversible)
P2Y12
(irreversible)
P2Y12
(reversible)
P2Y12
(reversible)
GPIIB/IIIa
(reversible)
GPIIB/IIIa
(reversible)
ApplicationOralOralOralOrali.v.i.v.i.v.
Time to Cmax0.5–1.0 h2 h
(after 600 mg LD)a
0.5 h
(after 60 mg LD)a
0.5 h
(after 180 mg LD)a
2 min5 min5 min
ProdrugNoYesYesNoNoNoNo
Bioavailability (%)∼50∼508036100100100
Drug interactionsNSAIDs
(in particular ibuprofen + naproxen)
CYP3A4, CYP3A5, or CYP2C19 inhibitors or inducersCYP3A4/A5 and CYP2B6 inhibitorCYP3A4 inducers or inhibitorsNoneNoneNone
Plasma half-life20 min0.5–1 h (active metabolite)0.5–1 h (active metabolite)6–12 h3–6 min2.5–2.8 h1.2–2 h
Duration of action after last dose7–10 days3–10 daysb7–10 daysb3–5 days1–2 h4 h8 h
Renal clearance of the active metabolite (%)NRNRNRNR58∼5065
Dose regimeno.d.o.d.o.d.b.i.d.Bolus, infusionBolus, infusionBolus, infusion

ASA, acetylsalicylic acid; b.i.d., twice a day; Cmax, maximum serum concentration; i.v., intravenous; LD, loading dose; NR, non-relevant; o.d., once a day.

a

Time to Cmax for may be delayed by 8 h or more following a dose of opiate.

b

Depending on response status.

Table 8

Pharmacokinetic and pharmacodynamic characteristics of oral anticoagulants

WarfarinPhenprocoumonApixabanDabigatranEdoxabanRivaroxaban
Target
(type of blockade)
VKORC1VKORC1FXaFIIaFXaFXa
ApplicationOralOralOralOralOralOral
Time to Cmax2–6 h1.52 h ± 1.523–4 h1.25–3 h1–2 h2–4 h
ProdrugNoNoNoYesNoNo
Bioavailability (%)>95100506.56280–100
Drug interactionsCYP2C9, CYP2C19, CYP2C8, CYP2C18, CYP1A2, CYP3A4, vitamin KCYP2C9, CYP2C8, vitamin KCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductorsP-glycoprotein inhibitors or inductorsP-glycoprotein inhibitorsCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductors
Plasma half-life36–48 h∼100 h12 h12–14 h6–11 h7–11 h (11–13 h in the elderly)
Duration of action after last dose∼5 days∼7 days24 h24 h24 h24 h
Renal clearance of the active metabolite (%)Non-renalNon-renal278537–5033
Dose regimenAdjusted according to INRAdjusted according to INRb.i.d.b.i.d.o.d.o.d./b.i.d.
WarfarinPhenprocoumonApixabanDabigatranEdoxabanRivaroxaban
Target
(type of blockade)
VKORC1VKORC1FXaFIIaFXaFXa
ApplicationOralOralOralOralOralOral
Time to Cmax2–6 h1.52 h ± 1.523–4 h1.25–3 h1–2 h2–4 h
ProdrugNoNoNoYesNoNo
Bioavailability (%)>95100506.56280–100
Drug interactionsCYP2C9, CYP2C19, CYP2C8, CYP2C18, CYP1A2, CYP3A4, vitamin KCYP2C9, CYP2C8, vitamin KCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductorsP-glycoprotein inhibitors or inductorsP-glycoprotein inhibitorsCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductors
Plasma half-life36–48 h∼100 h12 h12–14 h6–11 h7–11 h (11–13 h in the elderly)
Duration of action after last dose∼5 days∼7 days24 h24 h24 h24 h
Renal clearance of the active metabolite (%)Non-renalNon-renal278537–5033
Dose regimenAdjusted according to INRAdjusted according to INRb.i.d.b.i.d.o.d.o.d./b.i.d.

b.i.d., twice a day; Cmax, maximum serum concentration; FIIa, factor IIa; FXa, factor Xa; INR, International normalized ratio; LD, loading dose; NOAC, non-vitamin K antagonist oral anticoagulant; o.d., once a day; VKORC1, vitamin K epoxide reductase complex 1.

Table 8

Pharmacokinetic and pharmacodynamic characteristics of oral anticoagulants

WarfarinPhenprocoumonApixabanDabigatranEdoxabanRivaroxaban
Target
(type of blockade)
VKORC1VKORC1FXaFIIaFXaFXa
ApplicationOralOralOralOralOralOral
Time to Cmax2–6 h1.52 h ± 1.523–4 h1.25–3 h1–2 h2–4 h
ProdrugNoNoNoYesNoNo
Bioavailability (%)>95100506.56280–100
Drug interactionsCYP2C9, CYP2C19, CYP2C8, CYP2C18, CYP1A2, CYP3A4, vitamin KCYP2C9, CYP2C8, vitamin KCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductorsP-glycoprotein inhibitors or inductorsP-glycoprotein inhibitorsCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductors
Plasma half-life36–48 h∼100 h12 h12–14 h6–11 h7–11 h (11–13 h in the elderly)
Duration of action after last dose∼5 days∼7 days24 h24 h24 h24 h
Renal clearance of the active metabolite (%)Non-renalNon-renal278537–5033
Dose regimenAdjusted according to INRAdjusted according to INRb.i.d.b.i.d.o.d.o.d./b.i.d.
WarfarinPhenprocoumonApixabanDabigatranEdoxabanRivaroxaban
Target
(type of blockade)
VKORC1VKORC1FXaFIIaFXaFXa
ApplicationOralOralOralOralOralOral
Time to Cmax2–6 h1.52 h ± 1.523–4 h1.25–3 h1–2 h2–4 h
ProdrugNoNoNoYesNoNo
Bioavailability (%)>95100506.56280–100
Drug interactionsCYP2C9, CYP2C19, CYP2C8, CYP2C18, CYP1A2, CYP3A4, vitamin KCYP2C9, CYP2C8, vitamin KCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductorsP-glycoprotein inhibitors or inductorsP-glycoprotein inhibitorsCYP3A4 inhibitors or inductors, P-glycoprotein inhibitors or inductors
Plasma half-life36–48 h∼100 h12 h12–14 h6–11 h7–11 h (11–13 h in the elderly)
Duration of action after last dose∼5 days∼7 days24 h24 h24 h24 h
Renal clearance of the active metabolite (%)Non-renalNon-renal278537–5033
Dose regimenAdjusted according to INRAdjusted according to INRb.i.d.b.i.d.o.d.o.d./b.i.d.

b.i.d., twice a day; Cmax, maximum serum concentration; FIIa, factor IIa; FXa, factor Xa; INR, International normalized ratio; LD, loading dose; NOAC, non-vitamin K antagonist oral anticoagulant; o.d., once a day; VKORC1, vitamin K epoxide reductase complex 1.

Table 9

Bleeding risk according to type of non-cardiac surgery

Surgery with minor bleeding riskSurgery with low bleeding risk (infrequent or with low clinical impact)Surgery with high bleeding risk (frequent or with significant clinical impact)
  • Cataract or glaucoma procedure

  • Dental procedures: extractions (1–3 teeth), periodontal surgery, implant positioning, endodontic (root canal) procedures, subgingival scaling/cleaning

  • Endoscopy without biopsy or resection

  • Superficial surgery (e.g. abscess incision, small skin excisions/biopsy)

  • Abdominal surgery: cholecystectomy, hernia repair, colon resection

  • Breast surgery

  • Complex dental procedures (multiple tooth extractions)

  • Endoscopy with simple biopsy

  • Gastroscopy or colonoscopy with simple biopsy

  • Large-bore needles procedures (e.g. bone marrow or lymph node biopsy)

  • Non-cataract ophthalmic surgery

  • Small orthopaedic surgery (foot, hand arthroscopy)

  • Abdominal surgery with liver biopsy, extracorporeal shockwave lithotripsy

  • Extensive cancer surgery (e.g. pancreas, liver)

  • Neuraxial (spinal or epidural) anaesthesia

  • Neurosurgery (intracranial, spinal)

  • Major orthopaedic surgery

  • Procedures with vascular organ biopsy (kidney or prostate)

  • Reconstructive plastic surgery

  • Specific interventions (colon polypectomy, lumbar puncture, endovascular aneurysm repair)

  • Thoracic surgery, lung resection surgery

  • Urological surgery (prostatectomy, bladder tumour resection)

  • Vascular surgery (e.g. AAA repair, vascular bypass)

Surgery with minor bleeding riskSurgery with low bleeding risk (infrequent or with low clinical impact)Surgery with high bleeding risk (frequent or with significant clinical impact)
  • Cataract or glaucoma procedure

  • Dental procedures: extractions (1–3 teeth), periodontal surgery, implant positioning, endodontic (root canal) procedures, subgingival scaling/cleaning

  • Endoscopy without biopsy or resection

  • Superficial surgery (e.g. abscess incision, small skin excisions/biopsy)

  • Abdominal surgery: cholecystectomy, hernia repair, colon resection

  • Breast surgery

  • Complex dental procedures (multiple tooth extractions)

  • Endoscopy with simple biopsy

  • Gastroscopy or colonoscopy with simple biopsy

  • Large-bore needles procedures (e.g. bone marrow or lymph node biopsy)

  • Non-cataract ophthalmic surgery

  • Small orthopaedic surgery (foot, hand arthroscopy)

  • Abdominal surgery with liver biopsy, extracorporeal shockwave lithotripsy

  • Extensive cancer surgery (e.g. pancreas, liver)

  • Neuraxial (spinal or epidural) anaesthesia

  • Neurosurgery (intracranial, spinal)

  • Major orthopaedic surgery

  • Procedures with vascular organ biopsy (kidney or prostate)

  • Reconstructive plastic surgery

  • Specific interventions (colon polypectomy, lumbar puncture, endovascular aneurysm repair)

  • Thoracic surgery, lung resection surgery

  • Urological surgery (prostatectomy, bladder tumour resection)

  • Vascular surgery (e.g. AAA repair, vascular bypass)

AAA, abdominal aortic aneurysm.

Adapted from Steffel et al.240

Table 9

Bleeding risk according to type of non-cardiac surgery

Surgery with minor bleeding riskSurgery with low bleeding risk (infrequent or with low clinical impact)Surgery with high bleeding risk (frequent or with significant clinical impact)
  • Cataract or glaucoma procedure

  • Dental procedures: extractions (1–3 teeth), periodontal surgery, implant positioning, endodontic (root canal) procedures, subgingival scaling/cleaning

  • Endoscopy without biopsy or resection

  • Superficial surgery (e.g. abscess incision, small skin excisions/biopsy)

  • Abdominal surgery: cholecystectomy, hernia repair, colon resection

  • Breast surgery

  • Complex dental procedures (multiple tooth extractions)

  • Endoscopy with simple biopsy

  • Gastroscopy or colonoscopy with simple biopsy

  • Large-bore needles procedures (e.g. bone marrow or lymph node biopsy)

  • Non-cataract ophthalmic surgery

  • Small orthopaedic surgery (foot, hand arthroscopy)

  • Abdominal surgery with liver biopsy, extracorporeal shockwave lithotripsy

  • Extensive cancer surgery (e.g. pancreas, liver)

  • Neuraxial (spinal or epidural) anaesthesia

  • Neurosurgery (intracranial, spinal)

  • Major orthopaedic surgery

  • Procedures with vascular organ biopsy (kidney or prostate)

  • Reconstructive plastic surgery

  • Specific interventions (colon polypectomy, lumbar puncture, endovascular aneurysm repair)

  • Thoracic surgery, lung resection surgery

  • Urological surgery (prostatectomy, bladder tumour resection)

  • Vascular surgery (e.g. AAA repair, vascular bypass)

Surgery with minor bleeding riskSurgery with low bleeding risk (infrequent or with low clinical impact)Surgery with high bleeding risk (frequent or with significant clinical impact)
  • Cataract or glaucoma procedure

  • Dental procedures: extractions (1–3 teeth), periodontal surgery, implant positioning, endodontic (root canal) procedures, subgingival scaling/cleaning

  • Endoscopy without biopsy or resection

  • Superficial surgery (e.g. abscess incision, small skin excisions/biopsy)

  • Abdominal surgery: cholecystectomy, hernia repair, colon resection

  • Breast surgery

  • Complex dental procedures (multiple tooth extractions)

  • Endoscopy with simple biopsy

  • Gastroscopy or colonoscopy with simple biopsy

  • Large-bore needles procedures (e.g. bone marrow or lymph node biopsy)

  • Non-cataract ophthalmic surgery

  • Small orthopaedic surgery (foot, hand arthroscopy)

  • Abdominal surgery with liver biopsy, extracorporeal shockwave lithotripsy

  • Extensive cancer surgery (e.g. pancreas, liver)

  • Neuraxial (spinal or epidural) anaesthesia

  • Neurosurgery (intracranial, spinal)

  • Major orthopaedic surgery

  • Procedures with vascular organ biopsy (kidney or prostate)

  • Reconstructive plastic surgery

  • Specific interventions (colon polypectomy, lumbar puncture, endovascular aneurysm repair)

  • Thoracic surgery, lung resection surgery

  • Urological surgery (prostatectomy, bladder tumour resection)

  • Vascular surgery (e.g. AAA repair, vascular bypass)

AAA, abdominal aortic aneurysm.

Adapted from Steffel et al.240

5.3.1. Antiplatelets

5.3.1.1. Single antiplatelet therapy

In patients taking aspirin for primary prevention, the risk of ischaemic events is low and aspirin can be withdrawn prior to NCS. Permanent discontinuation should be considered post-operatively in low- and moderate-risk atherosclerotic cardiovascular disease (ASCVD) risk patients and/or in patients with high bleeding risk based on negative/neutral trials and the recommendations for primary prevention of CVD in the 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice.40,241

Due to the better risk–benefit ratio, aspirin has an established role for the long-term prevention of new cardiovascular events in patients with established CVD.242 The POISE-2 trial is the largest, randomized, placebo-controlled trial of peri-operative aspirin in patients undergoing NCS.243 The trial randomized 10 010 patients undergoing NCS with established CVD, or who were at increased CV risk, to aspirin or placebo. Patients were stratified according to whether they had not been taking aspirin before the study or were already on aspirin; 33% of the patients had known vascular disease (23% CAD, 9% PAD, and 5% stroke). Aspirin did not reduce the rates of death or non-fatal MI at 30 days (7.0% vs. 7.1% in the placebo group [HR, 0.99; 95% CI, 0.86–1.15; P = 0.92]). Major bleeding was more common in the aspirin group than in the placebo group (4.6% vs. 3.8% [HR, 1.23; 95% CI, 1.01–1.49; P = 0.04]). The primary outcome results were similar, irrespective of whether or not patients had been taking aspirin before the study, and were also similar in patients with and without vascular disease.

In a post hoc analysis of 470 patients (<5%) who had undergone previous PCI, aspirin use was associated with a significant reduction in death or MI (HR, 0.50; 95% CI, 0.26–0.95; P = 0.036) and MI alone (HR, 0.44; 95% CI, 0.22–0.87; P = 0.021), while the risk of major or life-threatening bleeding was not significantly increased in this setting.244 Although the analysis carries several limitations, it supports the perception that the ischaemic benefit of peri-operative aspirin use outweighs the bleeding risk in patients with previous PCI. Thus, among patients with previous PCI, in the absence of a very high bleeding risk, low-dose aspirin should be continued during the peri-operative period.

In patients undergoing transcatheter aortic valve implantation (TAVI) who have no other indication for oral anticoagulant (OAC) therapy, low-dose aspirin has been recommended as standard therapy by recent guidelines based on an RCT.245,246 There are no randomized data available assessing the withdrawal vs. continuation of aspirin in patients after TAVI on aspirin alone undergoing NCS.

If the bleeding risk outweighs the potential CV benefit, aspirin should be discontinued. For patients with high peri-operative bleeding risk (e.g. undergoing spinal surgery or certain neurosurgical or ophthalmological operations) aspirin should be discontinued for at least 7 days.

On rare occasions, chronic coronary syndrome (CCS) patients might be on clopidogrel monotherapy due to the results of recent trials247 and the recommendations of the 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation;98 therefore, periprocedural management of clopidogrel-based single antiplatelet therapy (SAPT) is required. Consensus has been reached that a short interruption of P2Y12 inhibitor monotherapy is recommended in patients at high risk of bleeding.

Patients treated with P2Y12 inhibitor monotherapy as part of a de-escalation strategy after PCI/ACS, or due to a recent stroke, PAD, or aspirin intolerance, might require peri-operative management of this monotherapy.248–250 A careful interdisciplinary evaluation of peri-operative bleeding vs. ischaemic risk is warranted in these situations, and individual decisions based on the peri-operative bleeding and ischaemic risk (e.g. surgery under P2Y12 monotherapy, switching to aspirin, short interruption, or bridging in the peri-operative phase) may be applicable, although evidence for these different regimens is missing. It should be recognized that the effects of ticagrelor or clopidogrel monotherapy on haemostasis are considerably less than when they are combined with aspirin.

5.3.1.2. Dual antiplatelet therapy

P2Y12 inhibitors in addition to aspirin are recommended for patients after PCI.98,146 The frequency of major NCS in the first year after PCI is ∼4%; most frequently orthopaedic, abdominal, and vascular surgery.251 Other observational data report cumulative incidences of NCS after PCI for 30 days, 6 months, and 1 year of 1%, 5%, and 9%, respectively.252

Observational studies have reported a substantial rate of MACE—including cardiac death, MI, and stent thrombosis—ranging between 2–8%251,253,254 in PCI patients undergoing NCS, with a more than two-fold increased risk compared with non-stented patients.255,256 The proportion of risk attributable to underlying CVD or stent implantation remains uncertain.254 Risk factors for MACE after NCS are: time from PCI to surgery, with the highest risk in the first month; primary PCI for ST-segment elevation myocardial infarction (STEMI); dual antiplatelet therapy (DAPT) interruptions/discontinuation; and lesion characteristics, including ostial and distal lesions.252,257–259 Urgency of surgery is a further risk factor. The ESC/ESA classification of NCS is a validated tool with which to predict the impact of the type of surgery on MACE.16

A meta-analysis of observational data indicated that discontinuation of clopidogrel for at least 5 days reduced the risk of re-operation for major bleeding by 50%, without increasing the risk of MACE or death.260 Other observational data indicate an increase in MACE with brief DAPT interruptions.261 However, these non-randomized data may have been biased by the type and urgency of surgery.260 Of note, the prognosis of stent thrombosis appears to be worse than for de novo coronary occlusion (and depends on the site of stent deployment), and premature interruption of DAPT in patients with recent coronary stent implantation is the strongest predictor for stent thrombosis.

The preferred management of patients on DAPT due to PCI is to delay elective NCS until completion of the full course of DAPT (6 months after elective PCI and 12 months after ACS).98,146 However, several recent trials have indicated that shortening DAPT duration to 1–3 months after implantation of modern DES is associated with acceptable rates of MACE and stent thrombosis in low- and moderate-risk patients. Based on these newer data, it is recommended to delay time-sensitive NCS until a minimum of 1 month of DAPT treatment has been effectuated. In high-risk CV patients, for example due to an ACS, a DAPT duration of at least 3 months should be considered before time-sensitive NCS. See Figure 5 for the recommended duration of DAPT before time-sensitive NCS. Once the P2Y12 inhibitor has been discontinued, surgery should be performed while the patient is still on aspirin.

Recommendations for management of antiplatelet therapy in patients undergoing non-cardiac surgery.
Figure 5

Recommendations for management of antiplatelet therapy in patients undergoing non-cardiac surgery.

ACS, acute coronary syndrome; DAPT, dual antiplatelet therapy; GPI, glycoprotein IIb/IIIa inhibitors; PCI, percutaneous coronary intervention; N, no; NCS, non-cardiac surgery. Y, yes; aHigh risk of peri-operative stent thrombosis defined by at least one of the following: history of stent thrombosis under antiplatelet therapy, reduced left ventricular ejection fraction (<40%), poorly controlled diabetes, severely impaired renal function/haemodialysis, recent complex PCI (i.e. severely calcified lesion, left main PCI, chronic total occlusion, bifurcational/crush technique, bypass graft PCI), or stent malapposition/residual dissection. bTiming of resumption after interdisciplinary risk assessment as soon as possible (within 48 h) after surgery. cFor dosing, see Figure 7.

Indications for long-term DAPT have recently emerged. Long-term DAPT (beyond 1 year) with clopidogrel, prasugrel, or ticagrelor in addition to aspirin should be considered in patients with high ischaemic risk, and may be considered in patients with moderate ischaemic risk, both in the absence of increased risk of major or life-threatening bleeding.98 When NCS is required, discontinuation of P2Y12 inhibitors is recommended for 3–7 days (depending on the P2Y12 inhibitor) for these additional indications for DAPT.

5.3.1.3. De-escalation of antiplatelet effect

The management of antiplatelet therapy in patients who have undergone recent PCI and are scheduled for NCS should be discussed by the surgeon and cardiologist, so that the balance between the risk of life-threatening surgical bleeding on antiplatelet therapy—best understood by the surgeon—and the risk of life-threatening stent thrombosis due to premature DAPT discontinuation—best understood by the cardiologist—can be considered (Figure 5 and Figure 6). An increased risk of MACE as a consequence of (major) bleeding also needs to be taken into consideration when balancing risk.

P2Y12 inhibitor interruption after percutaneous coronary intervention before elective non-cardiac surgery.
Figure 6

P2Y12 inhibitor interruption after percutaneous coronary intervention before elective non-cardiac surgery.

LoE, level of evidence; MI, myocardial infarction; N, no; PCI, percutaneous coronary intervention. Y, yes; aAvailability of 24 h cath-lab service is suggested in case of major surgery within 6 months in non-ACS/non-high-risk patients and within 12 months in ACS/high-risk patients. bHigh risk of peri-operative stent thrombosis defined by at least one of the following: history of recurrent MI, history of stent thrombosis under antiplatelet therapy, reduced left ventricular ejection fraction (<40%), poorly controlled diabetes, severely impaired renal function/haemodialysis, recent complex PCI (i.e. severely calcified lesion, left main PCI, chronic total occlusion, bifurcational/crush technique, bypass graft PCI), stent malapposition/residual dissection. cClass III LoE C. dClass IIa LoE B 250,265, 266,267. eClass I LoE A 268,146. fClass III LoE B 269. gClass IIb LoE B 270, 271. hClass IIa LoE B 272,273,274, 275, 276,277,278. iClass I LoE A 279, 280, 281, 98.

When time-sensitive surgery cannot be postponed and be performed with the recommended DAPT on board, de-escalation or shortening of DAPT is recommended. This may encompass either a switch from the more potent P2Y12 inhibitors prasugrel or ticagrelor to clopidogrel, or cessation of aspirin and use of prasugrel or ticagrelor monotherapy. If neither of these options is deemed to be sufficient, premature discontinuation of the P2Y12 inhibitor may be considered. If discontinuation is required, ticagrelor needs to be withheld for 3–5 days, clopidogrel for 5 days, and prasugrel for 7 days prior to surgery.262–264

Whenever possible, in patients with an indication for DAPT, surgery should be performed without discontinuation of aspirin. Aspirin might be discontinued as a last measure only with very high bleeding risk and a comparably low ischaemic risk. However, such surgical procedures should be performed in hospitals where 24/7 catheterization laboratories are available so as to treat patients immediately in case of peri-operative ischaemic events.

Although generally not recommended, bridging with i.v. compounds (eptifibatide/tirofiban or cangrelor) might be applicable in rare cases when DAPT cannot be interrupted before NCS (e.g. in patients with very high risk of stent thrombosis, history of recurrent MI, recent PCI) (see Figure 5 and Figure 7).282

Bridging with intravenous antiplatelet agents. ASA, acetylsalicylic acid; FU, follow-up; LD, loading dose; NCS, non-cardiac surgery; o.d., once a day.
Figure 7

Bridging with intravenous antiplatelet agents. ASA, acetylsalicylic acid; FU, follow-up; LD, loading dose; NCS, non-cardiac surgery; o.d., once a day.

aTirofiban: 0.1 µg/kg/min; if creatinine clearance <50 mL/min, adjust to 0.05 µg/kg/min. Eptifibatide: 2.0 µ/kg/min; if creatinine clearance is <50 mL/min, adjust to 1.0 µg/kg/min. bUntil oral P2Y12 inhibitor therapy is possible. cInitiate within 72 h from P2Y12 inhibitor discontinuation at a dose of 0.75 μg/kg/min for a minimum of 48 h and a maximum of 7 days.

For patients receiving antiplatelet therapy, who have excessive or life-threatening peri-operative bleeding, transfusion of platelets is recommended as a bail-out strategy. However, ticagrelor and its active metabolite may also inhibit aggregation of transfused platelets. Experimental data indicate that administration of albumin binds ticagrelor and reduces its inhibitory effect on platelet aggregation.283 A monoclonal antibody fragment (PB2452) for neutralizing ticagrelor is in development but is not yet clinically available.284

5.3.1.4. Platelet function guided peri-operative management of antiplatelet therapy

Platelet function testing has several theoretical advantages in the peri-operative setting, including: (i) the identification of patients on antiplatelet therapy who are at increased risk of surgery-related bleeding; (ii) individualized timing of elective surgery after antiplatelet therapy cessation; and (iii) guiding therapy in bleeding complications.285–287 However, neither the optimal assay nor a universal cut-off value associated with bleeding has been defined and validated in patients undergoing NCS.

Recommendation Table 13

Recommendations for use of antiplatelet therapy in patients undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 13

Recommendations for use of antiplatelet therapy in patients undergoing non-cardiac surgery

graphic
graphic

5.3.2. Oral anticoagulants

Approximately one in four patients taking anticoagulant therapy will require a surgical or invasive procedure within 2 years.290 Peri-operative management of oral anticoagulant (OAC) therapy depends on surgery- and patient-related factors and the specific OAC agent (vitamin K antagonist [VKA] or a non-vitamin K antagonist oral anticoagulant [NOAC]). See Figure 8 for a summary of recommendations for the management of OACs in patients undergoing NCS.

Recommendations for management of oral anticoagulation therapy in patients undergoing non-cardiac surgery.
Figure 8

Recommendations for management of oral anticoagulation therapy in patients undergoing non-cardiac surgery.

CHA2DS2-VASc, congestive heart failure, hypertension, age ≥75 years, diabetes mellitus, stroke, vascular disease, age 65–74 years, sex category (female); N, no; NCS, non-cardiac surgery; NOAC, non-vitamin K antagonist oral anticoagulant; VKA, vitamin K antagonist; VTE, venous thromboembolism. Y, yes; aMechanical aortic valve replacement (AVR) and any thromboembolic risk factor (atrial fibrillation, previous thromboembolism, severe left ventricular dysfunction, hypercoagulable state), or older-generation mechanical AVR, or a mechanical mitral valve replacement. bRecent stroke <3 months, high risk of VTE recurrences (e.g. antithrombin 3 deficiency or protein C and/or S deficiency), left ventricular apex thrombus, atrial fibrillation with a very high stroke risk. cBridging with unfractionated heparin or low molecular weight heparin. dE.g. >3 months after stroke/VTE. eFor NOAC management during NCS, see Figures 9 and 10.

Surgery-related factors include urgency of the intervention and the procedure-related bleeding risk (reflecting both the risk of bleeding occurrence and the risk of adverse outcome if bleeding occurs) (see Table 8). Procedures where mechanical compression is unfeasible carry a high risk of serious bleeding complications.

Patient-related factors include age, individual thrombotic risk, history of bleeding complications, renal function, concomitant medication, comorbidity, etc. Patients requiring a reversal agent need careful monitoring of haemostatic parameters and evaluation of thrombotic and bleeding risk in the peri-operative phase, as reversal might be insufficient or prothrombotic rebound might occur. In the latter case, an interdisciplinary decision should be made with regard to early resumption of anticoagulation treatment.

5.3.2.1. Vitamin K antagonists

Three drugs are currently used: warfarin (half-life 36–48 h), acenocoumarol (half-life 12 h), and phenprocoumon (half-life 100 h).

Vitamin K antagonists in patients with mechanical heart valves

Maintenance of therapeutic international normalized ratio (INR) is crucial for patients with mechanical heart valves (MHVs). Minor surgical procedures and procedures where bleeding is easily controlled can be performed without VKA interruption. The INR should be monitored and maintained at the lower level of the therapeutic range. Major surgical procedures needing INR ≤1.5 require VKA interruption, and heparin bridging should be considered. However, the evidence to support bridging therapy is limited and derived from cohort studies with poor or no comparator groups.291 Furthermore, the current-generation mechanical prosthetic valves in the aortic position are associated with a lower risk of thromboembolism compared with the older ones.291 Randomized controlled trials of bridging vs. no bridging therapy for patients with AF who do not have an MHV have shown higher risk of bleeding without a change in incidence of thromboembolic events, and increasing concerns have been raised that bridging therapy exposes patients to higher bleeding risks without reducing the risk of thromboembolism.292,293 The recently published PERI-OP trial compared bridging therapy vs. placebo in patients with either an MHV, AF, or atrial flutter who required interruption of OAC therapy for surgery, and found no significant benefit for post-operative dalteparin bridging to prevent major thromboembolism.294 The results were consistent for the AF (n = 1166) and MHVs groups (n = 350). Therefore, in patients with MHVs with a low risk of thromboembolism (e.g. mechanical bileaflet aortic valve in patients with sinus rhythm), bridging may not be needed. In patients with MHVs with a high risk of thromboembolism (mechanical aortic valve replacement [AVR] and any thromboembolic risk factor, or an older-generation mechanical AVR, or a mechanical mitral or tricuspid valve replacement), bridging with heparin should be considered during the peri-operative time interval when the INR is subtherapeutic (Figure 8). In all situations, the risks of bleeding should be weighed against the benefits of thromboembolism prevention.

Intravenous unfractionated heparin (UFH) is the only heparin treatment approved for bridging in patients with MHVs. Subcutaneous low molecular weight heparin (LMWH), although used off-label, has supplanted the use of UFH as a bridging therapy, owing to the lower incidence of thrombocytopenia, greater convenience, more predictable dose–response relationship, and significant cost saving resulting from outpatient administration. A meta-analysis of nine studies of 1042 patients with MHVs showed no differences between LMWH and UFH in the risks of thromboembolic events or major bleeding events.295 When LMWH is used, it should be given at a therapeutic dose twice a day and adjusted for renal impairment, when applicable. Anti-factor Xa (FXa) activity monitoring with target levels from 0.5–1.0 U/mL may be useful when the dosage may be difficult to determine (e.g. in patients with renal dysfunction or obesity). Vitamin K antagonist bridging strategies are shown in Supplementary data, Figure S2.

Vitamin K antagonists for atrial fibrillation/venous thromboembolism

In patients using VKA for AF or venous thromboembolism (VTE), invasive procedures with a low bleeding risk can be performed without VKA interruption.296–299 The INR should be monitored and maintained at the lower level of the therapeutic range. When interruption is necessary due to high bleeding risk procedures, the BRIDGE trial in AF patients showed that 3–5 days of warfarin interruption without bridging was superior to heparin bridging, having the same incidence of arterial and venous thromboembolism and significantly lower incidence of major bleeding.292

Bridging therapy may be considered for patients with a high thrombotic risk (i.e. AF with CHA2DS2-VASc (congestive heart failure, hypertension, age ≥75 years, diabetes mellitus, stroke, vascular disease, age 65–74 years, sex category [female]) score >6, recent cardioembolic stroke <3 months, or high risk of VTE recurrence, weighing the risk of bleeding against the risk of thromboembolism291,294,300 (see Supplementary data, Figure S3).

Restarting vitamin K antagonists after invasive procedures or surgery

Patients who have interrupted VKA treatment before surgery should restart the OAC 12–24 h after the invasive procedure, if the bleeding is well-controlled and gastric and intestinal reabsorption have been re-established. The restarting dose should be the maintenance dose plus a boosting dose of 50% for 2 days. Patients managed with bridging therapy should start LMWH or UFH together with VKA 24 h after surgery, if the bleeding is well-controlled and maintained, until the INR has reached the therapeutic range. In patients undergoing surgery with a high bleeding risk, therapeutic dose LMWH should be delayed for 48–72 h after haemostasis has been secured.

Reversal of vitamin K antagonists

Reversal of VKA can be managed with vitamin K, prothrombin complex concentrates (PPCs), and plasma administration. Vitamin K (from 2–10 mg depending on the INR value) can be used orally, with a predictable reduction in INR in 18–24 h or i.v. (in 25–50 mL normal saline over 15–30 min) for more rapid INR reduction (4–6 h). It should be noted that coagulation factors can still be below normal despite INR normalization, which means that bleeding risk might not yet be normalized. In patients needing reversal for immediate major surgery, PPCs or plasma should be used. Four-factor PPCs are the preferred option301 and are dosed on the basis of INR and body weight (INR 2–4 at 25 U/kg, INR 4–6 at 35 U/kg, INR >6 at 50 U/kg, with a maximum dose of 5000 U at 100 kg of body weight). When four-factor PPCs are unavailable, three-factor PPCs or plasma may be used. Patients requiring a reversal agent need careful monitoring of haemostatic parameters and evaluation of thrombotic and bleeding risks in the peri-operative phase, as reversal might be insufficient or a prothrombotic rebound might occur. In the latter case, an interdisciplinary decision should be made with regard to early resumption of anticoagulation treatment.

5.3.2.2. Non-vitamin K antagonist oral anticoagulants

Four drugs are currently used: dabigatran (factor IIa inhibitor), apixaban, rivaroxaban, and edoxaban (FXa inhibitors). The pharmacokinetic and pharmacodynamic characteristics of these drugs are shown in Table 8.

Unplanned surgery in patients on non-vitamin K antagonist oral anticoagulants and reversal for emergency procedures

When an urgent surgical intervention is required, it is recommended that NOAC therapy is immediately interrupted. Peri-operative management of NOAC therapy in specific procedural settings and suggested strategies for potential reversal of NOAC anticoagulant effect are shown in Figures 9–1199,240,302 (see Supplementary data, Table S5).

Peri-operative management of non-vitamin K antagonist oral anticoagulant according to the periprocedural risk of bleeding.
Figure 9

Peri-operative management of non-vitamin K antagonist oral anticoagulant according to the periprocedural risk of bleeding.

NCS, non-cardiac surgery; NOAC, non-vitamin K antagonist oral anticoagulant. aIn patients/circumstances favouring NOAC accumulation (e.g. renal dysfunction, older age, concomitant medication), the NOAC should be paused 12–24 h earlier. bIn patients on rivaroxaban or edoxaban taking the dose in the evening, the evening dose may be skipped. cNOACs have predictable weaning of the anticoagulant effect. Owing to the increase in bleeding risk associated with bridging, it is generally not recommended to use bridging in patients taking NOACs. Very few circumstances when bridging with heparin may be considered in patients taking a NOAC include high thromboembolic risk conditions, such as: 1) patients with a recent (within 3 months) thromboembolic event (stroke, systemic embolism, or VTE); 2) patients who experienced a thromboembolic event during previous interruption of NOAC therapy.

Timing of last non-vitamin K antagonist oral anticoagulant dose before elective NCS according to renal function.
Figure 10

Timing of last non-vitamin K antagonist oral anticoagulant dose before elective NCS according to renal function.

GFR, glomerular filtration rate; LMWH, low molecular weight heparin; NCS, non-cardiac surgery; NOAC, non-vitamin K antagonist oral anticoagulant; UFH, unfractionated heparin.

Suggested strategy for potential reversal of non-vitamin K oral anticoagulants effect.
Figure 11

Suggested strategy for potential reversal of non-vitamin K oral anticoagulants effect.

aPTT, activated partial thromboplastin time; dTT, diluted thrombin time; FXa, factor Xa; N, no; NOAC, non-vitamin K antagonist oral anticoagulant; PT, prothrombin time; UFH, unfractionated heparin. Y, yes; aConditions that are potentially life-threatening or that may threaten the survival of limb or organ. bConditions that can be managed and procedure delayed for several days. c>24 h in case of significantly reduced renal function (i.e. eGFR <50 mL/min). dIf specific reversal agent is unavailable, consider non-specific haemostatic agents (prothrombin complex concentrate [PCC] or activated PCC [aPCCs]). Idarucizumab has only been tested in patients undergoing urgent surgery. Andexanet has not been tested in patients requiring urgent surgery. Andexanet binds all FXa inhibitors (including UFH) non-specifically. eUpon re-check.

Whereas the open-label prospective trial testing the specific reversal agent idarucizumab in patients on dabigatran enrolled participants experiencing acute major bleeding or requiring urgent surgical intervention,303 the trial with the reversal agent andexanet alpha for FXa inhibitors included only patients with acute major bleeding under therapy, but not those requiring urgent surgery.304 However, the off-label use of andexanet alpha in life-threatening situations requiring an immediate intervention may be considered, bearing in mind that andexanet alpha non-specifically binds all FXa inhibitors, which may have important implications for further treatment, including the administration of UFH or LMWH.240 When specific reversal agents are unavailable, prothrombin complex concentrate (PCC) or activated PCC should be considered, although there is a lack of evidence on their efficacy and safety for emergency procedures in patients taking a NOAC.290,305 Performing immediate or urgent surgery under general rather than spinal anaesthesia is prudent, in order to reduce the risk of epidural haematoma.

Prior to unplanned surgery, the full panel of coagulation blood tests (see Supplementary data, Table S6) should be obtained in order to assess the patient’s coagulation status. The indication for reversal (and/or non-specific haemostatic) agents is primarily governed by the patient’s clinical presentation, but initial assessment of coagulation status may have important implications for treatment in the next few hours or days. Specific coagulation tests such as diluted thrombin time (dTT) or ecarin clotting assay for dabigatran and antiFXa chromogenic assays for FXa inhibitors, and the assessment of NOAC plasma levels, may help in interpreting routine coagulation tests and waning of anticoagulant effect.240

Planned interventions in patients on non-vitamin K oral anticoagulants

Invasive surgical interventions may require temporary discontinuation of NOAC therapy, while many less-invasive procedures with a relatively low risk of bleeding may be performed under minimally interrupted or uninterrupted NOAC therapy (Figure 9).240

Bridging

In patients taking a NOAC, peri-operative bridging using heparin or LMWH was associated with increased risk of bleeding without reduction in thromboembolic events.290,306–308 Therefore, when NOAC interruption is required for surgery, bridging is not recommended, except in a few high thrombotic risk circumstances (see Figure 9). However, post-operative thromboprophylaxis with LMWH should be considered in patients in whom NOAC therapy cannot be quickly restarted. In patients receiving bridging with LMWH, monitoring of antiFXa activity and dose adjustment to a target level of 0.5–1.0 U/mL may be considered.

Laboratory testing before surgery

Pre-operative assessment of anticoagulation status in patients on a NOAC undergoing planned surgery provides a direct assessment of residual drug concentration. Shorter NOAC interruption time intervals in patients undergoing low-risk procedures may result in mildly or moderately elevated NOAC levels, as seen in the Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) trial,309 whereas creatinine clearance <50 mL/min, standard NOAC dose (compared with reduced dose), body weight <70 kg, and female sex were associated with elevated NOAC levels among patients undergoing high-risk surgery. The use of amiodarone, verapamil, or diltiazem was also associated with elevated pre-operative NOAC levels in the prospective Per-procedural Concentration of Direct Oral Anticoagulants (CORIDA) trial.310 Importantly, elevated NOAC levels were not found to be independently associated with bleeding complications.309,310

The evidence base for modifying the duration of pre-operative NOAC interruption time intervals according to residual NOAC plasma levels is unavailable, and ‘safe’ plasma levels of NOACs for different procedures are largely unknown. The time-based NOAC interruption (Figure 9) appears safe in most patients undergoing surgery.311,312 When NOACs are interrupted for >72 h, the likelihood of any residual NOAC plasma levels is very low.309,310

Considerations for specific procedures

Before interventions that carry a very high risk of bleeding—such as spinal or epidural anaesthesia, or lumbar puncture requiring intact haemostasis---interruption of NOACs for up to five half-lives (i.e. 3 days for FXa inhibitors or 4–5 days for dabigatran) should be considered, whereas NOACs can usually be restarted 24 h after the intervention.313,314

Dental procedures are generally considered to be associated with minor bleeding risk, and adequate local haemostasis is usually easily achieved. Hence, most dental procedures can be performed in an outpatient setting, with uninterrupted NOAC (or a single skipped dose) and using specific local haemostatic measures (such as the application of oxidized cellulose or absorbable gelatine sponge, sutures, tranexamic acid mouthwash or compressive gauze). Most professional statements on dental surgery advise uninterrupted NOAC, but these recommendations are mostly based on expert consensus, while some studies are currently ongoing.315–317

When to restart non-vitamin K antagonist oral anticoagulants after interventions

In general, NOAC can be restarted 6–8 h after interventions with rapid and complete haemostasis. When the bleeding risk with full-dose anticoagulation resumption outweighs the risk of thromboembolic events, therapeutic anticoagulation may be postponed to >48–72 h after the procedure, using prophylactic post-operative thromboprophylaxis until the resumption of full-dose NOAC is deemed safe (Figure 9).240 Post-operative heparin administration should also be considered in patients unable to take oral therapy. Off-label use of reduced-dose NOACs to attenuate the risk of post-operative bleeding is not recommended, as there is no evidence informing such an approach.

5.3.2.3. Combination therapy (antiplatelet and anticoagulant)

In general, according to the 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS), dual antithrombotic therapy should be adopted in most patients with AF and a recent PCI.99 Elective surgery should be postponed until the period when antiplatelet therapy can be safely discontinued in combination therapy (6 months after elective PCI or 12 months after ACS).268 Peri-operative handling of NOACs should follow the above recommendations (Figures 9 and 10). In urgent/emergency surgery with high bleeding risk, operative measures to reduce bleeding and/or reversal strategies of anticoagulation might be applied. In patients receiving combination therapy for other indications (e.g. TAVI and AF), according to recent trial results, antiplatelet therapy can be safely discontinued before NCS.318 In patients receiving low-dose OACs as part of a vascular protection strategy, rivaroxaban should be paused for at least 24 h before surgery and resumed according to the post-operative bleeding risk.

Recommendation Table 14

Recommendations for interruption and resumption of anticoagulants in patients undergoing non-cardiac surgery

graphic
graphic
graphic
graphic
Recommendation Table 14

Recommendations for interruption and resumption of anticoagulants in patients undergoing non-cardiac surgery

graphic
graphic
graphic
graphic

5.4. Peri-operative thromboprophylaxis

Trends show that the case-fatality of peri-operative VTE has declined over the past few decades.319,320 Its causal relationship with preventable mortality has been challenged by a recent meta-analysis.321 Thus, peri-operative VTE should be regarded as a marker of increased mortality risk rather than a causal factor. Careful pre-operative assessment is essential to identify patients with increased VTE risk who might benefit from peri-operative thromboprophylaxis. Procedure-related (e.g. type of surgery and likelihood of post-operative immobilization) and patient-related factors contribute to the risk of VTE. For non-orthopaedic surgical patients at low risk of VTE, mechanical methods of VTE prophylaxis (graduated compression stockings, intermittent pneumatic compression, or venous foot pump) rather than pharmacologic prophylaxis or no prophylaxis are recommended. Patients with CV disease (e.g. patients with recent MI or HF) have increased risk of peri-operative VTE.322 The Caprini score has been developed for risk stratification,323 and validated in different surgical settings (see Supplementary data, Table S7).324–327

Thromboprophylaxis should be considered for patients with moderate (i.e. 5–8 points) and high scores (i.e. ≥9 points). Thromboprophylaxis should be initiated during the hospital stay until 12 h before NCS and continued post-operatively based on individual risk assessment for bleeding. In most cases, thromboprophylaxis should be continued until the patient becomes fully mobilized or until hospital discharge (usually up to 10 days). Extended pharmacological VTE prophylaxis beyond discharge is not routinely recommended in most non-orthopaedic surgical patients. Although there are insufficient data regarding thromboprophylaxis after cancer surgery (particularly major abdominal and/or pelvic surgery for cancer), the consensus is to extend treatment duration with preferred use of LMWH for 3–4 weeks. Decisions on prophylaxis in populations for which the Caprini score has not been validated (such as orthopaedic surgery) should be based on individual and procedure-specific risk factors. Among those, a previous VTE is the strongest risk predictor (see  Supplementary data, Table S7).328 For special situations and populations (e.g. neurosurgery, elderly, obese), please refer to available specific practice guidelines.329–332

Large phase 3 and phase 4 studies comparing NOACs with LMWH have shown similar results regarding efficacy and safety after major orthopaedic surgery.333 The usual time period for thromboprophylaxis after total knee and hip arthroplasty was up to 14 days and 35 days in RCTs, respectively,334–339 but large-scale data suggest safety of foreshortening the duration restricted to the hospital stay after fast-track surgery.340 Recent practice guidelines and a meta-analysis suggest a rationale for the use of aspirin as thromboprophylaxis in modern elective total hip and knee arthroplasty.341,342 However, there is a need for more adequately powered clinical trials with appropriate end-points comparing aspirin with other pharmacological methods. Aspirin should not be used as the sole initial agent for VTE prophylaxis, but switching to aspirin following a short course (e.g. 5 days) of rivaroxaban may be suitable for selected low-risk patients.343 It is recommended to implement patient care programmes—including post-operative mobilization, electronic prophylaxis recommendations, and teaching sessions regarding daily use of thromboprophylaxis—as these have been shown to be beneficial in reducing the risk of post-operative VTE complications.344

Recommendation Table 15

Recommendations for thromboprophylaxis

graphic
graphic
Recommendation Table 15

Recommendations for thromboprophylaxis

graphic
graphic

5.5. Patient blood management

Major surgery is associated with a high risk of peri-operative blood loss. Preferred treatment of acute anaemia related to peri-operative blood loss is transfusion of allogenic blood products. However, a large body of evidence indicates that inappropriate transfusion of red blood cells (RBCs) may be associated with inherent complications and impaired surgical outcome. Therefore, it is important to identify at-risk patients pre-operatively and manage peri-operative bleeding in any patients undergoing major surgery.

A hallmark study including >200 000 major surgical patients showed that even mild anaemia significantly increased the risk of morbidity—including respiratory, urinary, wound, septic, and thromboembolic complications—and mortality across all age groups.345 Moreover, Baron and colleagues analysed >39 000 surgical patients and showed that anaemia was significantly associated with increased mortality rate, hospital length of stay, and post-operative admission to intensive care.346 Up to 48% of surgical patients suffer from anaemia, and therefore anaemia should be considered to be a risk factor any time during hospitalization.347 Von Heymann and colleagues analysed 4494 cardiac surgical patients and showed that pre-operative anaemia and intra-operative transfusion were independently associated with decreased long-term survival.348 In addition, long-term survival was decreased by 50% in anaemic patients receiving blood transfusion compared with those without blood transfusion.

Anaemia may contribute to myocardial ischaemia, particularly if CAD is present. Iron deficiency (ID) is the underlying cause of anaemia in ∼50% of all cases.347 It was recently shown that ID was associated with increased risk of 90 day mortality both in patients with (4–14%) and without (2–5%) anaemia.349 In addition, the incidence of serious adverse events, major cardiac and cerebrovascular events, allogenic blood transfusion requirements, and length of stay were increased in patients with ID.

Based on the possibility of preserving the patients’ own blood resources and to enable safe handling of donor blood, the World Health Assembly (WHA) has endorsed the Patient Blood Management (PBM) approach (WHA63.12). Patient Blood Management is a patient-centred and multidisciplinary approach to manage anaemia, minimize iatrogenic blood loss and bleeding, and harness tolerance to anaemia in an effort to improve patient outcome.350–355 A comprehensive PBM programme addressing all three PBM pillars was associated with reduced transfusion need of RBC units, and lower complication and mortality rates.350

5.5.1. Pre-operative anaemia—diagnosis and treatment

A serum ferritin level <30 ng/mL, transferrin saturation <20%, and/or microcytic hypochromic red cells (mean corpuscular volume <80 fl, mean corpuscular haemoglobin <27 g/dL) are indicative of ID. In the presence of inflammation or transferrin saturation <20%, a ferritin level of <100 ng/mL points to functional ID (iron sequestration) (Table 10).

Table 10

Laboratory parameters for the diagnosis of absolute iron-deficiency anaemia

ParameterNormalIron deficiency
Mean corpuscular haemoglobin (g/dL)28–33<27
Mean cellular volume (fL)80–96<80
Transferrin saturation (%)16–45<20
Ferritin (ng/mL)18–360<30a
Reticulocytes haemoglobin (ng/mL)18–360<30
ParameterNormalIron deficiency
Mean corpuscular haemoglobin (g/dL)28–33<27
Mean cellular volume (fL)80–96<80
Transferrin saturation (%)16–45<20
Ferritin (ng/mL)18–360<30a
Reticulocytes haemoglobin (ng/mL)18–360<30
a

In cases of chronic kidney disease, chronic heart failure or infections, iron deficiency is diagnosed with ferritin level <100 ng/mL or transferrin saturation <20%.358

Table 10

Laboratory parameters for the diagnosis of absolute iron-deficiency anaemia

ParameterNormalIron deficiency
Mean corpuscular haemoglobin (g/dL)28–33<27
Mean cellular volume (fL)80–96<80
Transferrin saturation (%)16–45<20
Ferritin (ng/mL)18–360<30a
Reticulocytes haemoglobin (ng/mL)18–360<30
ParameterNormalIron deficiency
Mean corpuscular haemoglobin (g/dL)28–33<27
Mean cellular volume (fL)80–96<80
Transferrin saturation (%)16–45<20
Ferritin (ng/mL)18–360<30a
Reticulocytes haemoglobin (ng/mL)18–360<30
a

In cases of chronic kidney disease, chronic heart failure or infections, iron deficiency is diagnosed with ferritin level <100 ng/mL or transferrin saturation <20%.358

Apart from compromised bone marrow function, most types of anaemia are correctable within a period of 2–4 weeks. Oral and i.v. iron therapy can be used to treat ID. Intravenous iron products consist of an iron core embedded in a carbohydrate shell, which influences the stability of the drug, for example: iron sucrose comprises a less stable shell, allowing a maximum dose of 200 mg per infusion, whereas ferric carboxymaltose, ferric derisomaltose, and ferumoxytol have a stable shell associated with slow iron release and allowing a higher dose. Administration of i.v. iron has been shown to effectively reverse anaemia in ID patients.356,357

Intravenous iron is efficacious and safe359 and should be used in patients in whom oral iron is not tolerated, or if surgery is planned in short notice after the diagnosis of ID. A prospective observational study of 1728 major surgical patients showed that the prevalence of ID was 50%, 46.3%, and 52.7% in patients with haemoglobin <8.0, 8.0–8.9, and 9.0–9.9 g/dL, respectively.357 Furthermore, all iron supplemented iron-deficient anaemic patients required fewer RBC transfusions during the post-operative period, and a reduced intra-operative transfusion rate was observed if iron was supplemented >7 days before surgery. In addition, the length of stay was significantly reduced by 2.8 days for iron supplemented patients. In the recent PREVENTT trial studying patients with anaemia undergoing major abdominal surgery, pre-operative iron transfusion failed to improve outcomes;360 however, due to a fault in the study design, all anaemic patients received i.v. iron but not all (∼50–70%) were suffering from ID.

Recombinant human erythropoietin (rHuEPO) has frequently been used together with iron supplementation to increase pre-operative haemoglobin concentrations. A recent Cochrane review found that the administration of rHuEPO + iron to anaemic patients prior to NCS, compared with control treatment, reduced the need for RBC transfusion and increased the haemoglobin concentration pre-operatively; however, there were no important differences in the risk of adverse events or mortality within 30 days, or in length of hospital stay.361 Well-designed, adequately powered RCTs are required to more precisely estimate the impact of this combined treatment.

Pre-operative management of patients with anaemia can be simplified by making use of standard operating procedures or algorithms in which thresholds for diagnosis and treatment are depicted.362 An example of such an algorithm can be found in the PBM programme363 (see Supplementary data, Figure S4) and in the British Committee for Standards in Haematology (BCSH) Guidelines on the Identification and Management of Pre-Operative Anaemia.364

Recommendation Table 16

Recommendations for intra- and post-operative complications associated with anaemia

graphic
graphic
Recommendation Table 16

Recommendations for intra- and post-operative complications associated with anaemia

graphic
graphic

5.5.2. Bleeding and reduction of iatrogenic diagnostic/surgery-related blood loss

Blood loss associated with laboratory testing can either cause or aggravate hospital-acquired anaemia, which is associated with increased length of stay and complications. In 1867 patients undergoing cardiac surgery, an average of 115 tests per patient were performed, with a cumulative median volume of 454 mL.365 A reduction in blood drawn for laboratory analyses can be achieved by lowering the frequency of sampling and using paediatric-size collection tube sizes, for example. To decrease blood loss, blood-saving bundles could be used (e.g. a closed-loop arterial blood sampling system, smaller sampling tubes, reduction of frequency of blood drawings, and sample numbers). Such a strategy decreased mean blood loss per intensive care unit (ICU) day from 43.3 mL to 15.0 mL (P < 0.001),366 mainly due to the introduction of closed-loop arterial blood sampling systems. In addition, units of transfused RBCs per 100 observation days decreased from 7 to 2.3 (P < 0.001).366

A reduction in surgery-related blood loss starts from the pre-operative stage, with appropriate cessation strategies for anticoagulation and antiplatelet therapy. Intra-operative approaches to avoid blood loss include: (i) advanced anaesthetic; (ii) advanced surgical techniques with meticulous haemostasis, such as minimally invasive surgery and laparoscopic surgery; (iii) judicious use of diathermy dissection; (iv) physician’s mindfulness regarding limiting blood loss; and (v) application of topical haemostatic agents.367–369

Adequate coagulation management to minimize blood loss needs to be a pre-condition before RBC transfusion is considered. In this respect, the use of a coagulation algorithm is recommended,370,371 encompassing pre-operative assessment,372 and ensuring basic conditions for haemostasis, reversal of anticoagulants, point-of-care diagnostics in bleeding patients, and optimized coagulation management with the use of clotting factor concentrates.373,374

Tranexamic acid is an antifibrinolytic agent that is widely used for prophylaxis and treatment of bleeding caused by hyperfibrinolysis. A meta-analysis including 129 trials encompassing more than 10 000 patients to assess the effect of tranexamic acid on blood transfusion showed that administration of tranexamic acid reduced allogeneic blood transfusion by 38% (P < 0.001).375 In the recent POISE-3 study, 9535 patients undergoing NCS were randomized to tranexamic acid (1 g i.v. bolus) or placebo at the start and end of surgery. The incidence of the primary efficacy outcome (composite bleeding outcome) at 30 days was significantly lower with tranexamic acid than with placebo (HR, 0.76; 95% CI, 0.67–0.87).376 With respect to the primary safety outcome (composite CV outcome), the results did not meet the non-inferiority criteria (HR, 1.02; 95% CI, 0.92–1.14; P = 0.04 for non-inferiority).

The use of (washed) cell recovery is highly recommended in surgical settings where blood loss is routinely or anticipated to be >500 mL, as it reduces the rate of exposure to allogeneic RBCs, risk of infection, and length of stay. A meta-analysis including 47 trials encompassing 3433 patients of all surgical disciplines showed that the use of washed cell salvage reduced the rate of exposure to allogeneic RBC transfusion by 39% (P < 0.001), risk of infection by 28% (P = 0.03), and length of stay by 2.31 days (P < 0.001).377

Recommendation Table 17

Recommendations for intra- and post-operative complications associated with blood loss

graphic
graphic
graphic
graphic
Recommendation Table 17

Recommendations for intra- and post-operative complications associated with blood loss

graphic
graphic
graphic
graphic

5.5.3. Optimal blood component use with patient-centred clinical decision support

In order to optimize utilization of allogeneic blood products and to ensure guideline-adherent transfusion strategies, computerized physician order-entry systems should be considered.390,391 For example, Kaserer and colleagues evaluated the effectiveness of a monitoring and feedback programme and compared transfusion rates of >210 000 patients before and after implementation;392 overall, transfusion of RBCs was reduced by 40%.

Informed consent should be obtained from patients prior to transfusion of allogeneic blood products. It is necessary to effectively communicate the risks and benefits of the various potential interventions to the patient. It may further be recommended that any transfusion of allogeneic blood products should be mentioned in the discharge summary. In addition, the patient’s own preferences and values should be considered when developing a medical plan.

Recommendation Table 18

Recommendations for intra- and post-operative complications associated with allogeneic blood transfusion

graphic
graphic
Recommendation Table 18

Recommendations for intra- and post-operative complications associated with allogeneic blood transfusion

graphic
graphic

6. Specific diseases

Patients with CVD have an increased risk of peri-operative CV complications.45 Both the risk of complications and the peri-operative management depend on the specific type of CVD.

6.1. Coronary artery disease

6.1.1. Risk for patients with coronary artery disease

The peri-operative risk of CV complications for patients with established CAD depends on the baseline CV risk, type of surgery, and degree of urgency of the NCS. Older patients have a higher risk than younger patients, and patients with a recent ACS have a higher risk than those with CCS. The presence of comorbidities may also influence the risk.

6.1.2. Pre-operative risk assessment and management

The diagnostic evaluation and pre-operative management of patients with CCS undergoing NCS are outlined in Section 4. In particular, the value of CCTA and ICA are discussed in Sections 4.5.3.1. and 4.5.3.2.

In patients in need of immediate NCS, the operation must be performed without further delay and the time for pre-operative assessment is limited.

In patients scheduled for elective NCS who present with an ACS, the management of ACS should follow the guidelines for ACS patients in the non-surgical setting.98,171 In such settings, it would be reasonable to consider treating the culprit lesion only before NCS. Potential changes in timing of surgery and the peri-operative management (e.g. type of surgery, anaesthesia, medical therapy, and peri-operative monitoring) should be considered.

In patients with known CAD, it is recommended to collect information regarding previous invasive and non-invasive diagnostic examinations, and therapeutic interventions for CAD, within a sufficient time interval before the NCS, ideally at the time the proposal for NCS is made.

6.1.3. Revascularization strategies

The indication for coronary revascularization depends on the clinical presentation of CAD (ACS vs. CCS), urgency, and cardiac risk of NCS. In general, there is clear evidence that routine revascularization improves outcomes in ACS patients and less support for such a strategy in patients with CCS. The process of decision-making related to revascularization in CCS should be individualized, in order to prioritize revascularization in case of involvement of a significant amount of ischaemic myocardium or refractory symptoms, while medical management is a valuable option in patients with less relevant CAD manifestations.

6.1.3.1. Chronic coronary syndromes

The rationale for coronary revascularization before NCS is to prevent peri-operative myocardial ischaemia leading to acute MI, haemodynamic instability, and arrhythmia. Data from autopsy studies following fatal peri-operative MI showed that more than two-thirds of the patients had significant left main or three-vessel disease.397 A retrospective registry based on the Coronary Artery Surgery Study (CASS) study found that coronary artery bypass graft (CABG) reduced the risk of peri-operative mortality and MI in patients undergoing major NCS, particularly in subjects with three-vessel disease and reduced left ventricular ejection fraction (LVEF).398 However, the evidence in support of routine prophylactic revascularization before NCS is based on relatively small clinical trials and retrospective registries unrepresentative of current clinical practice.

In the Coronary Artery Revascularization Prophylaxis (CARP) trial, 510 patients with CCS were randomized to either optimal medical therapy or coronary revascularization (surgical or percutaneous) before major vascular surgery.399 In this study, patients receiving coronary revascularization compared with those who were medically treated did not differ in terms of acute MI after 30 days (8.4% vs. 8.4%, respectively) and mortality after 2.7 years (22% vs. 23%, respectively). Of note, this study excluded patients with significant left main disease, while one-third of the patients presented with three-vessel disease.399 In another randomized trial, 426 patients without evidence of CAD and scheduled for CEA were randomized to either routine coronary angiography with provisional revascularization before CEA or CEA without previous coronary angiography. There were no significant differences between the treatment groups in all-cause mortality, acute MI, and stroke at 30 days.172 A subsequent meta-analysis including 3949 patients showed no clinical benefit associated with routine prophylactic revascularization before NCS.400 A recent retrospective analysis on 4414 patients undergoing total joint arthroplasty found that the risk of adverse CV events was increased in patients with CAD, regardless of coronary revascularization before surgery. However, in patients receiving coronary revascularization, the risk of adverse CV events decreased, as the interval between revascularization and total joint arthroplasty was >2 years.401

The lack of evidence in support of routine prophylactic revascularization in CCS does not preclude a decision-process based on individual risk–benefit assessment in patients with a significant amount of ischaemic myocardium (as in the case of left main disease) and/or with refractory symptoms. The International Study of Comparative Health Effectiveness with Medical and Invasive Approaches (ISCHEMIA) trial randomized 5179 patients with stable CAD and moderate or severe ischaemia to an initial invasive strategy (consisting of angiography plus revascularization, as appropriate) or to an initial strategy of medical therapy alone, with bail-out angiography if medical therapy failed.402 This trial did not find a significant difference in the primary composite end-point of death and MI between treatment groups. Interestingly, these neutral results were inapplicable to patients with severe left main disease, since these patients were excluded after a pre-randomization CCTA. It has yet to be demonstrated whether CCTA represents a valuable tool with which to select patients with stable CAD and/or moderate or severe ischaemia who might benefit from an initial invasive strategy. A large angiography-based registry of 9016 CCS patients with high-risk coronary anatomy (three-vessel disease with ≥70% stenosis in all three epicardial vessels or left main disease ≥50% stenosis) showed improved outcomes (all-cause mortality or MI) in patients undergoing revascularization (both for PCI or CABG) vs. conservative medical therapy (HR, 0.62; 95% CI, 0.58–0.66; P < 0.001).403

The 2018 ESC/EACTS Guidelines on myocardial revascularization and the 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes generally apply to this population of patients, as long as the NCS can be postponed long enough to allow safe discontinuation of DAPT.146,404 Similarly, the choice between PCI and CABG should follow the general rules outlined in the above-mentioned guidelines.268,404 The use of intravascular imaging for planning and optimization of PCI is encouraged.405,406

6.1.3.2. Acute coronary syndromes

There are no trials specifically addressing the strategy of revascularization in ACS patients scheduled for NCS. High- and very high-risk patients should be treated according to the 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation,98 with an early (<24 h) or immediate (<2 h) invasive strategy, respectively. In low-risk groups amenable to a selective invasive strategy, the decision-making should be consistent with the approach for CCS patients.

In a post hoc analyses of the HIP-ATTACK (HIP Fracture Accelerated Surgical TreaTment And Care tracK) trial, patients with increased baseline troponin before randomization showed lower risk of mortality with accelerated surgery (within 6 h from the diagnosis) vs. standard of care (HR, 0.38; 95% CI, 0.21–0.66).57

The selection of the type of revascularization (PCI or CABG) should be based on the coronary anatomy and complexity of atherosclerosis, and the presence of diabetes.268,404 When PCI is chosen, the use of a DES is recommended.407 In case of a life-threatening clinical condition requiring undeferrable NCS and concomitant ACS-STEMI with an indication for coronary revascularization, a minimalistic approach with plain balloon angioplasty and delayed stenting might be considered.408,409  Figure 12 shows a summary of diagnostic and therapeutic pathways in patients with CAD scheduled for NCS.

Management of patients with acute or chronic coronary syndrome scheduled for non-cardiac surgery.
Figure 12

Management of patients with acute or chronic coronary syndrome scheduled for non-cardiac surgery.

ACS, acute coronary syndrome; BNP, B-type natriuretic peptide; CABG, coronary artery bypass graft; CAD, coronary artery disease; CCS, chronic coronary syndrome; ECG, electrocardiogram; FFR, fractional flow reserve; hs-cTn, high-sensitivity cardiac troponin; ICA, invasive coronary angiography; iwFR, instantaneous wave-free ratio; N, no; NCS, non-cardiac surgery; NT-proBNP, N-terminal proBNP; PCI, percutaneous coronary intervention; TTE, transthoracic echocardiography. Y, yes; The figure provides a schematic representation of diagnostic tools and therapy to be implemented according to surgical risk and underlying cardiac condition. aBiomarkers: hs-cTn T/I (Class I) ± BNP/NT-proBNP (Class IIa). bFunctional capacity based on Duke Activity Status Index (DASI) or the ability to climb two flights of stairs. cICA ± PCI/CABG on a case-by-case basis according to the Heart Team.

Recommendation Table 19

Recommendations for the timing of non-cardiac surgery and revascularization in patients with known coronary artery disease

graphic
graphic
Recommendation Table 19

Recommendations for the timing of non-cardiac surgery and revascularization in patients with known coronary artery disease

graphic
graphic

6.2. Chronic heart failure

6.2.1. Risk for patients with heart failure

Heart failure is an established risk factor for post-operative mortality across a broad range of surgical specialties.410–412 Several tools for risk calculations in patients undergoing NCS include HF as a predictor of adverse post-operative events.411,413

The risk of adverse post-operative events associated with HF depends on whether the LV systolic function is preserved or reduced, on the haemodynamic compensation, and on the presence of symptoms.414,415 In patients undergoing NCS, there is a risk of acute decompensated HF, with rapid onset or worsening of symptoms and/or signs of HF, precipitated by fluid accumulation and/or comorbid conditions.412

Patients with peri-operative acute or chronic HF are at increased risk of mortality during NCS. In a recently published analysis of 21 560 996 hospitalizations for NCS, the presence of any diagnosis of HF was associated with significantly higher in-hospital all-cause mortality compared with absence of HF (4.8% vs. 0.78%; adjusted OR, 2.15; 95% CI, 2.09–2.22).416 Among patients with a chronic HF diagnosis, peri-operative mortality was greater in those with acute exacerbation of chronic HF compared to those with compensated chronic HF. In a recent large-scale cohort study of individuals undergoing ambulatory surgery, the crude 90 day mortality was 2.0% among patients with HF and 0.4% among patients without HF.417 The crude risk of 30 day post-operative complications was 5.7% and 2.7%, respectively. Of note, the risk of mortality progressively increased with decreasing systolic function. It is not recommended to perform elective NCS in patients with decompensated HF.

The value of pre-operative assessment of LV function with TTE and measurement of natriuretic peptides (BNP or NT-proBNP) is discussed in Section 4.4. The TTE should not be older than 6 months, or performed just before NCS in the case of clinical worsening.

6.2.2. Pre- and post-operative management strategies

In order to reduce the risk of acute decompensation and the risk of mortality, optimal guideline-directed medical treatment of HF before scheduled NCS is recommended.412 Special attention should be given to the fluid balance, since high-volume infusion is often needed in the peri-operative period. Invasive monitoring of the arterial pressure aiming to obtain oximetric and metabolic parameters during NCS is frequently needed for intermediate- to high-risk NCS among HF patients. Furthermore, dynamic variables derived from the arterial pressure waveform (cardiac output, pulse pressure variation, stroke volume variation) are useful for guiding protocolled goal-directed therapy. Use of more invasive tools, such as right heart catheterization or transoesophageal echocardiography (TEE), might be considered on an individual patient level (see Section 7.1).

Baseline medication should be continued throughout the peri-operative period, in accordance with the recommendations provided in Section 5.2. It is recommended to perform ECG, measure biomarkers of myocardial injury (cTn T/I), and perform echocardiography to tailor the optimal treatment strategy in patients with acute decompensated HF post-operatively.

The management of patients with cardiac implantable electronic devices (CIEDs) undergoing NCS is discussed in Section 6.4.5. In patients with resynchronization devices (cardiac resynchronization therapy [CRT]), it is recommended to keep the device on to provide better haemodynamic stability.

6.2.3. Hypertrophic obstructive cardiomyopathy

Patients with hypertrophic cardiomyopathy with LV outflow tract obstruction (HOCM) have an increased risk of complications during NCS and require additional attention.418 It is recommended to perform TTE before NCS in order to determine the extent of the hypertrophy, obstruction, and diastolic function.419 Avoidance of prolonged pre-operative fasting and dehydration is important to maintain stroke volume and reduce the risk of increased obstruction. Furthermore, it is important to avoid vasodilating anaesthetic agents and maintain neutral fluid balance during the peri-operative period. The heart rate should be kept low (60–65 beats per minute [b.p.m.]) and AF should be avoided. Medication used to treat LV outflow tract obstruction should remain on board during NCS.

6.2.4. Patients with ventricular assist devices undergoing non-cardiac surgery

Ventricular assist devices (VADs) play an important role in the treatment of patients with end-stage HF who require a bridge to heart transplantation or as a permanent destination therapy. As the number of patients receiving VAD as destination therapy increases,420,421 the need for NCS in this specific subset of patients is expected to increase in years to come. Non-cardiac surgery should be performed in surgical centres that have access to VAD teams (Table 11).

Table 11

Peri-operative approach to patients with ventricular assist devices undergoing non-cardiac surgery

Pre-operativeIntra-operativePost-operative
  • Multidisciplinary team identified (primary surgical and anaesthesia teams, cardiac surgery, HF cardiologist, VAD personnel)

  • Pre-operative medical optimization when possible or necessary

  • Physical examination focused on the sequelae of HF

  • Baseline ECG, echocardiogram, and laboratory values

  • Manage pacemaker/ICD settings when indicated

  • CT examination to evaluate possible driveline interference with the operative field

  • Hold, bridge, or reverse anticoagulation when indicated, after VAD team consultation

  • Standard American Society of Anesthesiologists monitors

  • Cerebral tissue oxygenation, processed electroencephalogram, arterial line with ultrasound guidance, central venous catheter if fluid shifts are expected, PA catheter only if severe pulmonary hypertension, TEE available

  • Monitor VAD control console

  • External defibrillator pads in place

  • Optimize pre-load, support RV function, avoid increase in afterload

  • Gradual peritoneal insufflations and position changes

  • Standard post-anaesthesia care unit unless ICU is otherwise indicated

  • Extubation criteria are unchanged

  • Avoid hypoventilation, optimize oxygenation

  • Resume heparin infusion when post-op bleeding risk is acceptable

Pre-operativeIntra-operativePost-operative
  • Multidisciplinary team identified (primary surgical and anaesthesia teams, cardiac surgery, HF cardiologist, VAD personnel)

  • Pre-operative medical optimization when possible or necessary

  • Physical examination focused on the sequelae of HF

  • Baseline ECG, echocardiogram, and laboratory values

  • Manage pacemaker/ICD settings when indicated

  • CT examination to evaluate possible driveline interference with the operative field

  • Hold, bridge, or reverse anticoagulation when indicated, after VAD team consultation

  • Standard American Society of Anesthesiologists monitors

  • Cerebral tissue oxygenation, processed electroencephalogram, arterial line with ultrasound guidance, central venous catheter if fluid shifts are expected, PA catheter only if severe pulmonary hypertension, TEE available

  • Monitor VAD control console

  • External defibrillator pads in place

  • Optimize pre-load, support RV function, avoid increase in afterload

  • Gradual peritoneal insufflations and position changes

  • Standard post-anaesthesia care unit unless ICU is otherwise indicated

  • Extubation criteria are unchanged

  • Avoid hypoventilation, optimize oxygenation

  • Resume heparin infusion when post-op bleeding risk is acceptable

CT, computed tomography; ECG, electrocardiogram; HF, heart failure; ICU, intensive care unit; PA, pulmonary artery; RV, right ventricular; TEE, transoesophageal echocardiography; VAD, ventricular assist device.

Adapted from Roberts et al.421

Table 11

Peri-operative approach to patients with ventricular assist devices undergoing non-cardiac surgery

Pre-operativeIntra-operativePost-operative
  • Multidisciplinary team identified (primary surgical and anaesthesia teams, cardiac surgery, HF cardiologist, VAD personnel)

  • Pre-operative medical optimization when possible or necessary

  • Physical examination focused on the sequelae of HF

  • Baseline ECG, echocardiogram, and laboratory values

  • Manage pacemaker/ICD settings when indicated

  • CT examination to evaluate possible driveline interference with the operative field

  • Hold, bridge, or reverse anticoagulation when indicated, after VAD team consultation

  • Standard American Society of Anesthesiologists monitors

  • Cerebral tissue oxygenation, processed electroencephalogram, arterial line with ultrasound guidance, central venous catheter if fluid shifts are expected, PA catheter only if severe pulmonary hypertension, TEE available

  • Monitor VAD control console

  • External defibrillator pads in place

  • Optimize pre-load, support RV function, avoid increase in afterload

  • Gradual peritoneal insufflations and position changes

  • Standard post-anaesthesia care unit unless ICU is otherwise indicated

  • Extubation criteria are unchanged

  • Avoid hypoventilation, optimize oxygenation

  • Resume heparin infusion when post-op bleeding risk is acceptable

Pre-operativeIntra-operativePost-operative
  • Multidisciplinary team identified (primary surgical and anaesthesia teams, cardiac surgery, HF cardiologist, VAD personnel)

  • Pre-operative medical optimization when possible or necessary

  • Physical examination focused on the sequelae of HF

  • Baseline ECG, echocardiogram, and laboratory values

  • Manage pacemaker/ICD settings when indicated

  • CT examination to evaluate possible driveline interference with the operative field

  • Hold, bridge, or reverse anticoagulation when indicated, after VAD team consultation

  • Standard American Society of Anesthesiologists monitors

  • Cerebral tissue oxygenation, processed electroencephalogram, arterial line with ultrasound guidance, central venous catheter if fluid shifts are expected, PA catheter only if severe pulmonary hypertension, TEE available

  • Monitor VAD control console

  • External defibrillator pads in place

  • Optimize pre-load, support RV function, avoid increase in afterload

  • Gradual peritoneal insufflations and position changes

  • Standard post-anaesthesia care unit unless ICU is otherwise indicated

  • Extubation criteria are unchanged

  • Avoid hypoventilation, optimize oxygenation

  • Resume heparin infusion when post-op bleeding risk is acceptable

CT, computed tomography; ECG, electrocardiogram; HF, heart failure; ICU, intensive care unit; PA, pulmonary artery; RV, right ventricular; TEE, transoesophageal echocardiography; VAD, ventricular assist device.

Adapted from Roberts et al.421

Recommendation Table 20

Recommendations for management of heart failure in patients undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 20

Recommendations for management of heart failure in patients undergoing non-cardiac surgery

graphic
graphic

6.3. Valvular heart disease

6.3.1. Risk for patients with valvular heart disease

Valvular heart disease increases the risk of peri-operative CV complications during NCS. The magnitude of risk is highly variable and depends on the severity of VHD and type of NCS. It is particularly increased in patients with obstructive valve pathology, for example symptomatic AS or MS, where peri-operative volume shifts and arrhythmia may lead to rapid decompensation.424

6.3.2. Pre-operative management strategies and risk-reduction strategy

Clinical and echocardiographic evaluation is recommended in all patients with known or suspected VHD who are scheduled for elective intermediate-or-high-risk NCS. Patients in whom mild-to-moderate VHD was diagnosed >1 year earlier should have clinical and echocardiographic re-assessment. Heart team discussion may be helpful in patients with significant VHD. The risks of valvular intervention and the risk of NCS-related complications should be estimated and communicated to the patient and the surgical team.

6.3.2.1. Aortic valve stenosis

The peri-operative risk associated with AS during NCS depends upon the presence of symptoms, stenosis severity, and coexisting cardiac disease (e.g. CAD, mitral insufficiency, or reduced LVEF). Severe symptomatic AS is a significant risk factor for post-operative MI and HF, and a predictor for 30 day and long-term mortality after NCS.425,426 Careful peri-operative management is essential in patients undergoing intermediate- and high-risk NCS, albeit the significance of AS in patients undergoing low-risk NCS might have been overemphasized in studies that pre-date the more recent advances in anaesthesia, surgical techniques, and post-operative management. Aortic valve replacement has been associated with reduced in-hospital and 30 day mortality and morbidity among patients with AS undergoing intermediate- to high-risk NCS.425,426 However, the decision regarding the timing of AVR in relation to NCS should be weighted according to the baseline risk profile and the risk associated with the NCS. The choice of surgical aortic valve replacement (SAVR) vs. TAVI should follow the 2021 ESC/EACTS Guidelines for the management of valvular heart disease245 and patient’s informed preference (Figure 13).

Management of patients with severe aortic valve stenosis scheduled for non-cardiac surgery.
Figure 13

Management of patients with severe aortic valve stenosis scheduled for non-cardiac surgery.

BAV, balloon aortic valvuloplasty; ECG, electrocardiogram; LV, left ventricular; LVEF, left ventricular ejection fraction; N, no; NCS, non-cardiac surgery; SAVR, surgical aortic valve replacement; TAVI, transcatheter aortic valve implantation; TTE, transthoracic echocardiography; TEE, transoesophageal echocardiography. Y, yes; The figure provides a schematic representation of diagnostic assessment or therapy to be implemented according to surgical risk and underlying cardiac condition. aThis applies to treatment of complications (e.g. atrial fibrillation, heart failure). No medical therapy is recommended for aortic stenosis per se.

In patients with severe symptomatic AS, in whom NCS can be deferred, aortic valve intervention (SAVR or TAVI) is recommended before NCS. In patients requiring time-sensitive NCS, TAVI is a reasonable option.427 In patients with severe symptomatic AS in need of time-sensitive NCS in whom TAVI or SAVR are unfeasible, balloon aortic valvuloplasty (BAV) may be considered before NCS as a bridge to definitive aortic valve repair. Asymptomatic patients with severe AS and normal LVEF can safely undergo low- to intermediate-risk NCS, unless the NCS is associated with large volume shifts.245,428

6.3.2.2. Mitral valve stenosis

Non-cardiac surgery can be performed with a relatively low risk of complications in patients with mild MS (valve area >1.5 cm2) and in asymptomatic patients with moderate-to-severe MS (valve area ≤1.5 cm2) and systolic pulmonary artery pressure (SPAP) <50 mmHg on echocardiography.429 As transmitral gradients are flow-sensitive, tachycardia and fluid overload can cause pulmonary oedema during NCS. In this regard, arterial vasodilators should be avoided and surveillance for peri-operative AF is of paramount importance. Management of anticoagulation for patients with high thrombotic risk is discussed in Section 5.3.2. In asymptomatic patients with moderate-to-severe MS and SPAP >50 mmHg, and in symptomatic patients, the peri-operative risk of CV events is increased. In this case, a percutaneous mitral commissurotomy (PMC) should be considered before high-risk NCS. Otherwise, a multidisciplinary team should manage patients with moderate-to-severe MS who are ineligible for PMC, and NCS should be performed only if necessary. Intermediate-risk NCS may be performed in asymptomatic patients with severe MS with appropriate intra-operative and post-operative haemodynamic monitoring, if valve morphology is unsuitable for PMC.

6.3.2.3. Aortic valve regurgitation

In patients with mild-to-moderate aortic valve regurgitation (AR), NCS can be performed without additional risk. Patients with severe AR in whom valvular intervention is needed should be treated before intermediate-or-high-risk elective NCS (see Recommendation Table 21 and the 2021 ESC/EACTS Guidelines for the management of valvular heart disease).245

6.3.2.4. Mitral valve regurgitation

In patients with severe symptomatic mitral valve regurgitation (MR), the type of valve disease (primary or secondary) and LV function should be assessed. Patients with secondary MR, especially of ischaemic aetiology, are at increased risk of CV complications during NCS.430 Patients with severe symptomatic MR fulfilling the intervention criteria should be referred for valve treatment before intermediate- or high-risk elective NCS. In case of symptomatic moderate-to-severe secondary MR, patients meeting the Cardiovascular Outcomes Assessment of the MitraClip Percutaneous Therapy for Heart Failure Patients With Functional Mitral Regurgitation (COAPT) criteria should be considered for transcatheter edge-to-edge treatment before NCS (Figure 14).245,431 In patients with severe primary MR with symptoms or asymptomatic with LV dysfunction, valve repair is the recommended therapy (see Recommendation Table 21 and the 2021 ESC/EACTS Guidelines for the management of valvular heart disease245).

Management of patients with secondary mitral valve regurgitation scheduled for non-cardiac surgery.
Figure 14

Management of patients with secondary mitral valve regurgitation scheduled for non-cardiac surgery.

ECG, electrocardiogram; GDMT, guideline-directed medical therapy; N, no; NCS, non-cardiac surgery; TTE, transthoracic echocardiography; TEE, transoesophageal echocardiography; TEER, transcatheter edge-to-edge repair. Y, yes; aCoronary angiography ± PCI/CABG on a case-by-case according to the expert team.

In patients with reduced LVEF and concomitant AR or MR, intra-operative haemodynamic monitoring, heart rate control, and careful fluid balance are crucial to avoid haemodynamic deterioration, especially during high-risk surgery.

Recommendation Table 21

Recommendations for management of valvular heart disease in patients undergoing non-cardiac surgery

graphic
graphic
graphic
graphic
Recommendation Table 21

Recommendations for management of valvular heart disease in patients undergoing non-cardiac surgery

graphic
graphic
graphic
graphic
6.3.2.5. Patients with prosthetic valve(s)

Patients who have undergone previous surgical correction of VHD and have a prosthetic valve can undergo NCS provided that there is no evidence of valve dysfunction. In current practice, the main problem is the need for a modification of the anticoagulation regimen in the peri-operative period; this is discussed in detail in Section 5.3.

6.3.2.6. Prophylaxis of infective endocarditis

Prophylaxis against infective endocarditis in patients requiring NCS should be consistent with the 2015 ESC Guidelines for the management of infective endocarditis.432

6.4. Known or newly diagnosed arrhythmias

Arrhythmias pose a significant burden to patients undergoing NCS, contributing to excessive morbidity and mortality.433,434

6.4.1. Peri-operative management—general measures

Arrhythmias—namely, supraventricular tachycardia (SVT) and ventricular tachycardia (VT)—may accompany acute surgical illness, but should not defer urgent surgical procedures, unless the arrhythmia is life-threatening. All patients with known arrhythmia undergoing elective surgery should have a 12-lead ECG performed pre-operatively and undergo a cardiology check-up. Prevention of potential arrhythmic triggers is crucial: electrolyte and acid-base imbalance, myocardial ischaemia (also caused by excessive blood loss and anaemia), and large volume shifts, which can provoke subsequent autonomic hyperactivity, should be avoided in the pre-, intra-, and post-operative periods. Patients with a systolic HF should receive optimal treatment, as such therapy reduces the risk of total mortality and sudden cardiac death (SCD).435 Patients already taking AADs should generally not stop taking these drugs. Patients at high-risk of malignant arrhythmias should have continuous ECG monitoring through the whole peri-operative period, with particular emphasis placed on patients who have had implantable cardioverter–defibrillators (ICDs) deactivated during NCS.

6.4.2. Supraventricular arrhythmias

Supraventricular arrhythmias do not usually cause deferral of surgery. In rare cases, presence of pre-excitation and AF conducted rapidly over an accessory pathway flag a patient at risk of SCD and may indicate the need for ablation, if surgery is not emergent.

Supraventricular premature beats usually do not require therapy. Identification and correction of potential triggers (electrolyte and acid-base imbalance, volume overload, etc.) are highly recommended. Peri-operative SVT typically responds well to vagal manoeuvres, or to a bolus of adenosine if unsuccessful. If SVT persists or reoccurs, i.v. beta-blockers, verapamil, or diltiazem can be used for rhythm conversion or temporal slowing of atrioventricular conduction.436 Prompt cardioversion should be performed in rare cases of haemodynamically unstable SVT. If prophylactic therapy is needed to prevent recurrent SVT, beta-blockers or non-dihydropyridine CCBs (verapamil, diltiazem) can be used, and flecainide/propafenone or amiodarone can be considered if ineffective. Rarely, when SVT recurs despite therapy or becomes incessant, ablation should be considered in patients undergoing high-risk, non-emergent surgery. Recently published results of an RCT confirmed the superiority of radiofrequency (RF) ablation over AADs for persistent atrioventricular nodal re-entry tachycardia. Large registries and meta-analyses have demonstrated efficacy and safety of RF ablation in WPW syndrome and other SVTs, with a single-procedure success rate of >90.437–441

6.4.3. Atrial fibrillation/flutter

The majority of patients with AF receive lifelong OAC therapy for the prevention of stroke and systemic embolism,99 and the peri-operative management of OAC therapy will depend on the type of surgery (see Section 5.3.2).99,240 Sometimes AF is asymptomatic442 and may be first detected on admission for surgery, or may first occur in the pre-operative period. The initial management of newly diagnosed AF includes prevention of thromboembolism, and symptom control, and should not be deferred awaiting consultation with a cardiologist.99 In patients with newly diagnosed AF who require systemic OAC therapy for stroke prevention, the choice of anticoagulant in the pre-operative period depends on the type of surgery (see Section 5.3.2). Optimal rate control (i.e. resting heart rate <110 b.p.m.)99 is mandatory in all patients with AF, whereas rhythm control (i.e. achieving and maintenance of sinus rhythm) in the pre-operative period may be considered only if symptoms persist despite optimal rate control.

Rate control can be achieved using beta-blockers or non-dihydropyridine CCBs (verapamil, diltiazem). Amiodarone can be used as first-line therapy in patients with HF, whereas digoxin is usually ineffective in high adrenergic conditions such as surgery. Pharmacological cardioversion of symptomatic recent-onset AF can be attempted using flecainide or propafenone; in patients without significant LV hypertrophy, LV systolic dysfunction, or IHD, the use of flecainide or propafenone results in prompt (3–5 h) and safe restoration of sinus rhythm in >50% of patients. Intravenous amiodarone administration has a limited and delayed effect but can slow the heart rate within 12 h; i.v. vernakalant is the most rapidly cardioverting drug, including patients with mild HF and/or IHD.99,443 Dofetilide is not used in Europe, and ibutilide is effective to convert atrial flutter to sinus rhythm.99,444 In patients with AF and haemodynamic instability, emergency cardioversion (most commonly electrical direct current cardioversion) is indicated.99 Alternatively, pharmacological cardioversion using i.v. AADs should be attempted, if consistent with the patient’s clinical status. Systemic OAC therapy to prevent thromboembolic events should be started as soon as possible.99 Post-operative AF is discussed in Section 8.6.

Management of atrial flutter follows the same principles as AF with respect to OAC therapy. Rate control is usually an initial approach in patients with atrial flutter;436 however, drugs that slow atrioventricular conduction (digoxin, beta-blockers, or non-dihydropyridine CCBs) are usually less effective than in AF. In patients with a high ventricular rate, electrical cardioversion is frequently needed.445–448 Amiodarone may be an alternative used to control rate, especially in HF or critically compromised patients.449 Dofetilide and ibutilide are effective in converting atrial flutter to sinus rhythm, whereas class IA and IC drugs and amiodarone are less efficient and should not be used.450–453

6.4.4. Ventricular arrhythmias

Premature ventricular contractions (PVC) and non-sustained VT are frequent in the general population and patients undergoing NCS. Specific clinical features have been suggested as predictors of increasing incidence of PVC.454 These arrhythmias have historically been considered benign; however, recent studies have suggested that they may be associated with an adverse outcome, even in patients with apparently normal hearts, especially if frequent (e.g. >10–20%).455–459 In patients with heart disease, the prognostic impact of PVC and non-sustained VT depends on type and extent of heart damage.460–466 In patients undergoing urgent NCS, they do not require treatment unless frequent and symptomatic. If haemodynamically compromising, up-titration of beta-blockers is recommended; amiodarone (300 mg i.v. bolus) should be considered if beta-blockers are not tolerated or contraindicated.467 Further diagnostics to rule out significant heart disease is necessary in patients awaiting elective NCS, especially if frequent, complex (non-sustained VT), symptomatic, or in those with a positive family history of SCD.

Polymorphic VT and ventricular fibrillation (VF) can be provoked by ischaemia, electrolyte imbalance, or may be manifestations of primary electrical disease, such as long-QT or Brugada syndrome. Monomorphic VT is often associated with the presence of scarred myocardium. Hence, peri-operative VT or VF in a patient awaiting surgery should lead to a diagnostic work-up to exclude severe ventricular dysfunction (see Section 4.5.1), and to rule out CAD requiring prompt revascularization (see Section 6.1) and other potential causes of arrhythmia (primary electrical disease, dyselectrolytaemia). Monomorphic VT in patients without overt structural or electrical heart disease (idiopathic VT, most commonly arising from outflow tract) is associated with good prognosis and may be left untreated or, if symptomatic, may be treated with beta-blockers, verapamil, or sodium channel blockers. Patients with haemodynamically compromising VT should undergo electrical cardioversion (after i.v. sedation, if conscious) and VF should be terminated with prompt defibrillation. Recurrent VT and VF in the setting of acute ischaemia may be effectively treated with beta-blockers and amiodarone, and myocardial revascularization in case of obstructive CAD.468 Up-titration of beta-blockers to the maximal tolerated doses can prevent arrhythmia recurrence.469

Haemodynamically stable, sustained VT should be cardioverted as a first-line treatment; i.v. procainamide or flecainide may be considered in patients without HF or myocardial ischaemia. In cases in which these drugs are unavailable, i.v. amiodarone can be used. In selected cases, when monomorphic VT recurs in patients with scarred myocardium with no reversible causes, despite optimal therapy, invasive electrophysiological study and ablation should be performed pre-operatively, if NCS can be deferred. After extensive endocardial VT ablation, treatment with an OAC for a limited period of time might be reasonable.470,471 A summary of diagnostic and therapeutic pathways in patients with SVT or VT is shown in Table 12.

Table 12

Peri-operative management of patients with arrhythmias

Type of arrhythmiaSVTIdiopathic VT in structurally/functionally normal heartVT in structural heart disease
Diagnostics
  • ECG ± TTEa

  • ECG ± TTE

  • ECG + TTE + biomarkersb

  • ± Coronary angiography

  • ± Cardiac CT/MRI

Acute management
  • Vagal manoeuvres

  • I.v. adenosine, beta-blocker, CCB

  • Electrical cardioversion if unstable

  • Vagal manoeuvres

  • I.v. beta-blockers/ verapamil

  • Electrical cardioversion if unstable

  • Treatment of underlying heart disease

  • I.v. beta-blocker (uptitration), amiodarone

  • Electrical cardioversion if unstable

Prevention of recurrence
  • Per oral beta-blocker, CCB

  • Catheter ablation if recurrent despite OMT (only before high-risk NCS)

  • No treatment or

  • Per oral beta-blocker, CCB, class I AAD

  • Catheter ablation in case of recurrence despite AADs or drug-intolerance before high-risk NCS

  • Per oral beta-blocker, amiodarone

  • Catheter ablation if recurrent despite OMT

Type of arrhythmiaSVTIdiopathic VT in structurally/functionally normal heartVT in structural heart disease
Diagnostics
  • ECG ± TTEa

  • ECG ± TTE

  • ECG + TTE + biomarkersb

  • ± Coronary angiography

  • ± Cardiac CT/MRI

Acute management
  • Vagal manoeuvres

  • I.v. adenosine, beta-blocker, CCB

  • Electrical cardioversion if unstable

  • Vagal manoeuvres

  • I.v. beta-blockers/ verapamil

  • Electrical cardioversion if unstable

  • Treatment of underlying heart disease

  • I.v. beta-blocker (uptitration), amiodarone

  • Electrical cardioversion if unstable

Prevention of recurrence
  • Per oral beta-blocker, CCB

  • Catheter ablation if recurrent despite OMT (only before high-risk NCS)

  • No treatment or

  • Per oral beta-blocker, CCB, class I AAD

  • Catheter ablation in case of recurrence despite AADs or drug-intolerance before high-risk NCS

  • Per oral beta-blocker, amiodarone

  • Catheter ablation if recurrent despite OMT

AAD, antiarrhythmic drug; CCB, calcium channel blocker; CT, computer tomography; ECG, electrocardiogram; MRI, magnetic resonance imaging; NCS, non-cardiac surgery; OMT, optimal medical therapy; SVT, supraventricular tachycardia; TTE, transthoracic echocardiography; VT, ventricular tachycardia.

a

Before high-risk surgery.

b

High-sensitivity cardiac troponin T/I, and/or BNP/ N-terminal pro-BNP.

Table 12

Peri-operative management of patients with arrhythmias

Type of arrhythmiaSVTIdiopathic VT in structurally/functionally normal heartVT in structural heart disease
Diagnostics
  • ECG ± TTEa

  • ECG ± TTE

  • ECG + TTE + biomarkersb

  • ± Coronary angiography

  • ± Cardiac CT/MRI

Acute management
  • Vagal manoeuvres

  • I.v. adenosine, beta-blocker, CCB

  • Electrical cardioversion if unstable

  • Vagal manoeuvres

  • I.v. beta-blockers/ verapamil

  • Electrical cardioversion if unstable

  • Treatment of underlying heart disease

  • I.v. beta-blocker (uptitration), amiodarone

  • Electrical cardioversion if unstable

Prevention of recurrence
  • Per oral beta-blocker, CCB

  • Catheter ablation if recurrent despite OMT (only before high-risk NCS)

  • No treatment or

  • Per oral beta-blocker, CCB, class I AAD

  • Catheter ablation in case of recurrence despite AADs or drug-intolerance before high-risk NCS

  • Per oral beta-blocker, amiodarone

  • Catheter ablation if recurrent despite OMT

Type of arrhythmiaSVTIdiopathic VT in structurally/functionally normal heartVT in structural heart disease
Diagnostics
  • ECG ± TTEa

  • ECG ± TTE

  • ECG + TTE + biomarkersb

  • ± Coronary angiography

  • ± Cardiac CT/MRI

Acute management
  • Vagal manoeuvres

  • I.v. adenosine, beta-blocker, CCB

  • Electrical cardioversion if unstable

  • Vagal manoeuvres

  • I.v. beta-blockers/ verapamil

  • Electrical cardioversion if unstable

  • Treatment of underlying heart disease

  • I.v. beta-blocker (uptitration), amiodarone

  • Electrical cardioversion if unstable

Prevention of recurrence
  • Per oral beta-blocker, CCB

  • Catheter ablation if recurrent despite OMT (only before high-risk NCS)

  • No treatment or

  • Per oral beta-blocker, CCB, class I AAD

  • Catheter ablation in case of recurrence despite AADs or drug-intolerance before high-risk NCS

  • Per oral beta-blocker, amiodarone

  • Catheter ablation if recurrent despite OMT

AAD, antiarrhythmic drug; CCB, calcium channel blocker; CT, computer tomography; ECG, electrocardiogram; MRI, magnetic resonance imaging; NCS, non-cardiac surgery; OMT, optimal medical therapy; SVT, supraventricular tachycardia; TTE, transthoracic echocardiography; VT, ventricular tachycardia.

a

Before high-risk surgery.

b

High-sensitivity cardiac troponin T/I, and/or BNP/ N-terminal pro-BNP.

Recommendation Table 22

Recommendations for management of known or newly diagnosed arrhythmias

graphic
graphic
graphic
graphic
Recommendation Table 22

Recommendations for management of known or newly diagnosed arrhythmias

graphic
graphic
graphic
graphic

6.4.5. Bradyarrhythmias

Temporary cardiac pacing during the peri-operative period should be limited to patients undergoing urgent NCS, if bradycardia is haemodynamically compromising despite i.v. chronotropic drugs, or provokes episodes of ventricular tachyarrhythmia.481 In patients undergoing elective NCS, surgery should be deferred if possible, and permanent pacemaker should be implanted, if indications for pacing are fulfilled.481 Prophylactic pacing in the settings of asymptomatic bifascicular block, with or without first-degree atrioventricular block, is generally not indicated, and chronotropic drugs (atropine, isoprenaline, adrenaline, or, alternatively, aminophylline, dopamine, or glucagon in beta-blocker or CCB overdosing) are usually effective. Patients with bifascicular bundle branch block or prolonged His-ventricular interval are at an increased risk of developing complete heart block.482,483 Equipment needed to perform emergent transcutaneous pacing and personnel able to perform such a procedure should be immediately available during NCS in patients with bifascicular block; alternatively, a permanent pacemaker may be implanted.481

6.4.6. Management of patients with cardiac implantable electronic devices

Patients with CIEDs can undergo NCS, pending adequate peri-operative device management. A pre-operative check should have been performed at least once within the 12 months preceding surgery for pacemaker patients and within 6 months for patients with ICD, in the absence of any malfunction (remote monitoring can also be used for check-ups).484,485 In pacing-dependent patients, patients with biventricular pacing for CRT, and ICD-recipients undergoing elective NCS associated with risk of electromagnetic interference (EMI) (e.g. involving the use of unipolar electrocoagulation, especially above the umbilicus), CIED check and reprogramming should be performed immediately before surgery. In pacemaker-dependent patients, devices should be reprogrammed to non-sensing or asynchronous pacing mode to protect against inhibition of the pacemaker. This can be performed in the majority of pacemaker models by placing a magnet over the pacemaker can.486,487 However, magnet mode in modern pacemakers, except Medtronic and Sorin/Livanova/Microport, is programmable and may not be asynchronous pacing; thus, magnet application is not a universal remedy against EMI-induced malfunction. Furthermore, asynchronous pacing may lead to pacing on the T wave, which can provoke VT/VF. However, the risk of clinically significant EMI is low487–489 and a practical solution would be to monitor the patient via plethysmography or an arterial line, and limit the use of electrocautery if pauses occur during ECG monitoring.

Patients with leadless pacemakers may safely undergo surgery, with precautions similar to patients with conventional pacemakers, avoiding EMI and after reprogramming the pacemaker into non-sensing mode in pacing-dependent patients (due to its intracardiac location and lack of a Hall-effect sensor/reed switch, this device cannot be temporarily reprogrammed to an asynchronous mode with a magnet applied over the body of the pacemaker).490,491

In patients with ICDs undergoing NCS with anticipated risk of EMI, arrhythmia detection or antiarrhythmic therapies through the device should be switched off before NCS,492 or a magnet should be put over the device.488,489,492 All modern ICDs will respond to magnet application by inhibiting antitachycardia therapy, while the brady pacing is left intact. Deactivation by programming mandates telemetry and cardioversion equipment until reactivation, which may be impractical. Furthermore, there is a risk that the patient is discharged without the device being reactivated. These factors would favour the use of a magnet instead of deactivation. In some patients where the device is inaccessible for magnet application, a magnet cannot be used and reprogramming is mandatory. From this point onwards, throughout the whole procedure until reactivation of the ICD, the patient should have continuous ECG monitoring, and personnel skilled in early detection of arrhythmias, defibrillation, and cardiopulmonary resuscitation manoeuvres should be present. As soon as possible after NCS, it is recommended that the ICD is checked and therapies switched on.485

Patients with subcutaneous ICD can undergo surgery after switching off the antiarrhythmic therapy or magnet application; however, if thoracic surgery is planned, especially with a median sternotomy, the surgeon should be aware of the presence of the ICD and the course of the subcutaneous electrode. This can prevent mechanical damage of the lead, the direct use of electrocautery on the electrode, or the placement of sternal wires in close proximity to the sensing electrodes.493

In pacing-dependent patients, patients with CRT, and ICD patients, EMI with the device should be avoided (e.g. with electrocautery). Use of bipolar electrocautery, short bursts of impulses limited to several seconds (<5 s), with the lowest effective energy, and operating with a pen or stylus away from the device (>15 cm) can minimize the risk of interference with the device. In the case of unipolar electrocoagulation, the electrosurgical unit should be connected in a way that keeps the current circuit away from the CIED can and electrodes. However, the manufacturer’s recommendations should be considered (usually recommending placement of the indifferent electrode on the opposite site of the body to the one operated on, but possibly close to the surgical site, on a well-vascularized, muscular area). Consequently, the indifferent return pad should be placed as far away from the CIED as possible, keeping the surgical site between the CIED and the return electrode (Figure 15).494–497

Optimal location of return electrode during unipolar electrosurgery in patients with cardiac implantable electronic devices, depending on the surgery site.
Figure 15

Optimal location of return electrode during unipolar electrosurgery in patients with cardiac implantable electronic devices, depending on the surgery site.

CIED, cardiac implantable electronic device. Use of bipolar electrocautery, short (<5 s) bursts of impulses, with the lowest effective energy, operating with pen or stylus away (>15 cm) from the device can minimize the risk of interference with the device. (A) Surgery site on ipsilateral site above CIED. (B) Surgery on ipsilateral site below CIED. (C) Surgery on contralateral site.494

In patients with implantable loop recorders (especially those not undergoing remote monitoring and regular downloads of the CIED memory), the device memory download is to be considered before procedures associated with possible EMI, or involving the anatomical location close to the device, to avoid misrecognizing and recording noise as arrhythmia or erasing the memory.498,499

Recommendation Table 23

Recommendations for management of bradyarrhythmia and patients carrying cardiac implantable devices

graphic
graphic
graphic
graphic
Recommendation Table 23

Recommendations for management of bradyarrhythmia and patients carrying cardiac implantable devices

graphic
graphic
graphic
graphic

6.5. Adult congenital heart disease

Adults with congenital heart disease (ACHD) account for >60% of the population with congenital heart disease (CHD).500,501 Accordingly, ACHD represent an increasing proportion of NCS admissions502 and might be at high risk of CV events.

Pre-operative risk assessment in ACHD needs to focus on the underlying disease, type of surgery, residua, and sequelae.503 Coexistence of HF, pulmonary hypertension, arrhythmia, hypoxaemia, damage to other organs, and endocarditis may considerably influence the baseline risk of these patients from no additional risk to very high risk of worse prognosis.503,504 Thus, original medical and surgical reports should be obtained along with current data, which should include symptoms, exercise capacity, oxygen saturation, laboratory values (BNP, haemoglobin, creatinine, etc.), and medication.

In a recent report, absolute mortality in ACHD patients undergoing NCS exceeded 4%.502 Mortality and peri-operative morbidity were greater in ACHD compared with a matched comparison cohort, and patients with severe ACHD had the highest mortality rate. It is well known that patients with pulmonary hypertension and with Eisenmenger syndrome have a higher risk of complications.505 A large registry confirmed these findings: patients with severe CHD had an increased risk of 30 day mortality, overall mortality, and reintubation, while patients with intermediate CHD had a moderate increase in overall mortality and risk of reintubation.506 Based on this study and recent guidelines,503 the classification in Table 13 is proposed for risk stratification.

Table 13

Risk stratification for non-cardiac surgery in adults with congenital heart disease

Minor riskPatients with small, uncorrected defects, and no need for medication or any other treatment
Patients with successfully corrected CHD with no symptoms, no relevant residua, and no need for medication
Intermediate riskPatients with corrected or uncorrected conditions with residual haemodynamic abnormality, with or without medication
Severe riskPatients with uncorrected cyanotic heart disease, pulmonary hypertension, other complex CHD, ventricular dysfunction requiring medication, and patients listed for heart transplantation
Minor riskPatients with small, uncorrected defects, and no need for medication or any other treatment
Patients with successfully corrected CHD with no symptoms, no relevant residua, and no need for medication
Intermediate riskPatients with corrected or uncorrected conditions with residual haemodynamic abnormality, with or without medication
Severe riskPatients with uncorrected cyanotic heart disease, pulmonary hypertension, other complex CHD, ventricular dysfunction requiring medication, and patients listed for heart transplantation

CHD, congenital heart disease.

Table 13

Risk stratification for non-cardiac surgery in adults with congenital heart disease

Minor riskPatients with small, uncorrected defects, and no need for medication or any other treatment
Patients with successfully corrected CHD with no symptoms, no relevant residua, and no need for medication
Intermediate riskPatients with corrected or uncorrected conditions with residual haemodynamic abnormality, with or without medication
Severe riskPatients with uncorrected cyanotic heart disease, pulmonary hypertension, other complex CHD, ventricular dysfunction requiring medication, and patients listed for heart transplantation
Minor riskPatients with small, uncorrected defects, and no need for medication or any other treatment
Patients with successfully corrected CHD with no symptoms, no relevant residua, and no need for medication
Intermediate riskPatients with corrected or uncorrected conditions with residual haemodynamic abnormality, with or without medication
Severe riskPatients with uncorrected cyanotic heart disease, pulmonary hypertension, other complex CHD, ventricular dysfunction requiring medication, and patients listed for heart transplantation

CHD, congenital heart disease.

A consultation by an ACHD specialist is necessary, especially in patients with intermediate or severe ACHD scheduled for intermediate- or high-risk NCS. It is recommended that elective surgery in intermediate- and severe-risk ACHD patients should take place in centres experienced in the management of ACHD patients. It is generally recommended to perform the least invasive procedures and anaesthesia with as low an impact on haemodynamics as possible.

Optimal peri-operative care in ACHD undergoing NCS starts with a proper pre-operative evaluation. Adults with congenital heart disease can present with multiorgan involvement (kidneys, liver, lung, and endocrine system), which should be considered during the diagnostic work-up.503 In many cases, patients with ACHD have a lifelong indication for OAC therapy or antithrombotic treatment, mainly due to arrhythmias or increased thromboembolic risk associated with specific ACHD. Oral anticoagulant therapy in the peri-operative phase should be re-evaluated on a case-by-case basis. The CHA2DS2-VASc score has not been validated in ACHD patients and should not be used in this group.

Continuous haemodynamic monitoring can be necessary in patients with ACHD and should include invasive BP monitoring, especially in cases with ACHD of moderate or severe complexity. It is worth mentioning that, according to the type of ACHD or surgery (e.g. coarctation of the aorta or following a Blalock-Taussig shunt), the location of the arterial line for continuous BP and gas exchange monitoring requires careful attention. In patients with persistent right-to-left shunts, air filters must be used for venous access. Ventilator management and extubation can be complicated by the presence of restrictive lung disease.507 Antibiotic prophylaxis for endocarditis should be given according to the 2015 ESC Guidelines for the management of infective endocarditis.432 Furthermore, post-operative care in an ICU with experience in handling ACHD patients is often necessary.

A prolonged monitoring period in this setting should be considered, with a special focus on arrhythmias and an optimal volume management, as it has been shown that up to 50% of adverse events were attributable to lapses in the post-operative monitoring and care.508 Two groups at special risk are patients with chronic cyanosis and those after the Fontan procedure. Chronic cyanosis is associated with multiorgan involvement. Furthermore, bleeding risk is higher due to multiple collaterals, platelet dysfunction, and alterations in the coagulation cascade.509 In patients with Eisenmenger syndrome, conditions that increase pulmonary vascular resistance such as hypothermia, metabolic acidosis, hypercapnia, and hypovolaemia must be avoided.509 This is also true for patients after the Fontan procedure, where venous return relies on low pulmonary pressures. If intra-abdominal pressure rises too high in these patients, venous return is drastically reduced, with a subsequent drop in cardiac output. These haemodynamic aspects should be carefully considered in cases of laparoscopic or open NCS.

Recommendation Table 24

Recommendations for management of patients with adult congenital heart disease undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 24

Recommendations for management of patients with adult congenital heart disease undergoing non-cardiac surgery

graphic
graphic

6.6. Pericardial diseases

Active pericardial disease is infrequent at the time of NCS, but potentially life-threatening. The clarification of underlying aetiology is of utmost importance for peri-operative management (viral or bacterial infection, malignant, systemic autoimmune, metabolic, or autoreactive disease). The treatment of these conditions should follow the recommendations provided in the 2015 ESC Guidelines for the diagnosis and management of pericardial diseases.510

Acute pericarditis is a clear indication to postpone an elective surgical procedure. However, in cases of undeferrable NCS, attention is required regarding drug-to-drug interactions. The frequently used colchicine is predominantly metabolized through the liver, while renal excretion accounts for only 10–20%. Colchicine may increase sensitivity to central nervous system depressants and exert a respiratory depressant effect.511 Adverse events of peri-operative significance include diarrhoea, worsening renal failure, and, very rarely, bone marrow suppression, hepatotoxicity, paralysis, convulsions, and cardiopulmonary collapse. Immunosuppressive drugs such as steroids and interleukin-1 receptor antagonist agents suppress the immune system, and may increase the risk of infection and delay wound healing in the peri-operative phase.511

Imminent cardiac tamponade is an absolute contraindication for all surgical procedures, especially when a general anaesthesia is required.510,512 Before NCS, pericardial effusion should first be percutaneously drained, under local anaesthesia. In cases of small or moderate chronic pericardial effusion and constrictive pericarditis, attention should be paid to take measurements to increase cardiac pre-load. The pre-load should be optimized peri-operatively with i.v. fluids prior to general anaesthetic induction to facilitate ventricular filling. Manipulation and medication diminishing venous return to the heart should be avoided or minimized. Positive pressure ventilation might cause a dramatic decline in pre-load and should be avoided. If spontaneous breathing is not possible, ventilation with minimal inspiratory pressures (low tidal volumes, high respiratory rate) should be considered. Anaesthetics that minimize changes in heart rate, systemic vascular resistance, venous return, and myocardial contractility should be selected.189 Ketamine, a sympathetic stimulant that preserves spontaneous ventilation, is the drug of first choice. Combinations of opiates, benzodiazepines, and nitric oxide, with or without low doses of volatile anaesthetics, are acceptable for maintenance of anaesthesia. Muscle relaxants with minimal circulatory effects should be preferred, although the modest increase in heart rate observed with administration of pancuronium is also acceptable.

Recommendation Table 25

Recommendations for pericardial diseases

graphic
graphic
Recommendation Table 25

Recommendations for pericardial diseases

graphic
graphic

6.7. Pulmonary disease and pulmonary arterial hypertension

The coexistence of pulmonary disease in cardiac patients undergoing NCS may increase the operative risk. Pre-existing pulmonary disease has a significant impact on peri-operative risk, but the most common effect is an increase in the risk of post-operative pulmonary complications. These complications particularly occur after abdominal or thoracic surgery, and the risk seems to be increased in smokers. Certain respiratory conditions are associated with CV pathology and may require special cardiac risk assessment and management, in addition to dealing with the pulmonary disease per se. Three such conditions are chronic obstructive pulmonary disease (COPD), obesity hypoventilation syndrome, and pulmonary arterial hypertension (PAH).

6.7.1. Pulmonary disease

Chronic obstructive pulmonary disease is as a major cause of morbidity and mortality.513 Although patients with COPD have an increased risk of CVD, there is no evidence that COPD is related to a higher risk of peri-operative cardiac complications. However, post-operative pulmonary complications result in significant mortality and morbidity. Pre-operative evaluation, using specific post-operative pulmonary complication tools, can be used to stratify at-risk patients and enable optimal pre-operative and peri-operative management.514 In patients with COPD undergoing NCS, the pre-operative treatment goals are to optimize pulmonary function and minimize post-operative respiratory complications; this includes using the pre-operative period for education, including possible cessation of smoking (2 months before surgery), instruction concerning chest physiotherapy and lung expansion manoeuvres, muscular endurance training, and re-nutrition if required. Beta-adrenergic agonists and anticholinergic agents should be continued until the day of surgery in all symptomatic COPD patients with bronchial hyper-reactivity. In some cases, short-term systemic/inhaled steroids may be considered. Where there is active pulmonary infection, appropriate antibiotics should be administered for at least 10 days and, if possible, elective NCS should be deferred.515

Obesity hypoventilation syndrome is defined as the triad of obesity, daytime hypoventilation, and sleep-disordered breathing. Although distinct from simple obesity and sleep apnoea, it is estimated that 90% patients with obesity hypoventilation syndrome also have obstructive sleep apnoea (OSA). The prevalence of obesity hypoventilation syndrome is 0.15–3% of adults and 7–22% in patients undergoing bariatric surgery.516 Obesity hypoventilation syndrome is associated with even higher morbidity, including HF (and obesity-related cardiomyopathy), angina pectoris, pulmonary hypertension (30–88%) and cor pulmonale, and increased peri-operative mortality.516 Patients at high risk of obesity hypoventilation syndrome who are undergoing NCS should be referred for additional specialist investigation of sleep-disordered breathing and pulmonary hypertension, with pre-operative initiation of appropriate positive airway pressure therapy, and planning of peri-operative techniques (anaesthetic and surgical) and post-operative positive airway pressure management within an appropriate monitored environment.516,517

6.7.2. Pulmonary arterial hypertension

Pulmonary arterial hypertension is associated with increased morbidity and mortality in patients undergoing NCS.518 A meticulous pre-operative diagnostic work-up in this subset of patients should include assessment of functional status and severity of disease, in addition to comorbidities and the type of NCS. Echocardiography and right heart catheterization (if clinically indicated) are key components in the pre-operative work-up. The morbidity and mortality associated with PAH derive from the haemodynamic response of the right ventricle to acute increases in afterload.519

In patients with severe PAH, peri-operative mortality ranging between 3–18% has been reported, depending on the severity of the underlying disease, and the nature and urgency of the surgical procedure. Emergency procedures are also associated with a high risk of complications.520,521 Patient-related and surgery-related factors should be considered when assessing peri-operative risk in patients with PAH (Table 14).522 Owing to the potential for anaesthesia and surgery to be complicated by acute right HF and pulmonary hypertensive crisis, elective NCS in patients with PAH should be adequately discussed in a multidisciplinary team. Ideally, patients with PAH undergoing NCS should have optimal medical treatment before surgery and be managed in a centre experienced in PAH. Patients scheduled for NCS should be discussed by a pneumologist, cardiologist, surgeon, and an anaesthesiologist.523 The management of patients with PAH in the peri-operative setting should follow the recommendations provided in the 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension.524

Table 14

Patient-related and surgery-related factors to be considered when assessing peri-operative risk in patients with pulmonary arterial hypertension

Patient-related peri-operative risk factors in patients with PAHSurgery-related peri-operative risk factors in patients with PAH
  • Functional class >II

  • Reduced six-minute walk distance

  • Coronary heart disease

  • Previous pulmonary embolism

  • Chronic renal insufficiency

  • Severe right ventricular dysfunction

  • Emergency surgery

  • Duration of anaesthesia >3 h

  • Intra-operative requirement for vasopressors

Patient-related peri-operative risk factors in patients with PAHSurgery-related peri-operative risk factors in patients with PAH
  • Functional class >II

  • Reduced six-minute walk distance

  • Coronary heart disease

  • Previous pulmonary embolism

  • Chronic renal insufficiency

  • Severe right ventricular dysfunction

  • Emergency surgery

  • Duration of anaesthesia >3 h

  • Intra-operative requirement for vasopressors

PAH, pulmonary arterial hypertension.

Adapted from Olsson et al.522

Table 14

Patient-related and surgery-related factors to be considered when assessing peri-operative risk in patients with pulmonary arterial hypertension

Patient-related peri-operative risk factors in patients with PAHSurgery-related peri-operative risk factors in patients with PAH
  • Functional class >II

  • Reduced six-minute walk distance

  • Coronary heart disease

  • Previous pulmonary embolism

  • Chronic renal insufficiency

  • Severe right ventricular dysfunction

  • Emergency surgery

  • Duration of anaesthesia >3 h

  • Intra-operative requirement for vasopressors

Patient-related peri-operative risk factors in patients with PAHSurgery-related peri-operative risk factors in patients with PAH
  • Functional class >II

  • Reduced six-minute walk distance

  • Coronary heart disease

  • Previous pulmonary embolism

  • Chronic renal insufficiency

  • Severe right ventricular dysfunction

  • Emergency surgery

  • Duration of anaesthesia >3 h

  • Intra-operative requirement for vasopressors

PAH, pulmonary arterial hypertension.

Adapted from Olsson et al.522

Several novel therapies aimed at reducing pulmonary pressures are used pre-operatively in patients undergoing NCS. Of these therapies, endothelin receptor antagonists, phosphodiesterase inhibitors, and prostacyclin analogues are the most used.525 This medication should be continued during the peri-operative phase because therapy disruption may lead to a critical rebound of the PAH. Inhaled nitric oxide allows selective pulmonary vasodilatation with rapidity of action and it can be given to patients who develop worsening pulmonary hypertension post-operatively to maintain right ventricular (RV) function and haemodynamic stability.526

Recommendation Table 26

Recommendations for patients with pulmonary arterial hypertension undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 26

Recommendations for patients with pulmonary arterial hypertension undergoing non-cardiac surgery

graphic
graphic

6.8. Arterial hypertension

The prevalence of arterial hypertension in adults in Europe is ∼30–45%.527 Of these patients, <40% have well-controlled BP (<140/90 mmHg). A large observational study has shown that patients with untreated hypertension 1 month before surgery had a 69% increased risk of 90 day post-operative mortality.183 Further, overall CV risk assessment, including the search for hypertension-mediated organ damage, is of paramount importance in hypertensive patients, and mandatory when there is newly detected BP elevation.528

Postponing surgery is usually not advised in patients with grade 1 or 2 hypertension. In contrast, in subjects with a systolic BP ≥180 mmHg and/or diastolic BP ≥110 mmHg, deferring the intervention until BP is under control is advisable, except for emergency surgery.236,527,529,530 It also seems important to avoid large peri-operative BP fluctuations. In a recent randomized trial among patients undergoing abdominal surgery, an individualized intra-operative treatment strategy with systolic BP values kept within a 10% difference from the pre-operative office measurement resulted in a reduced risk of post-operative organ dysfunction.528 In a meta-analysis including 130 862 patients undergoing surgery, intra-operative hypotension was associated with increased risk of morbidity (OR, 2.08; 95% CI, 1.56–2.77), mortality (OR, 1.94; 95% CI, 1.32–2.84), cardiac complications (OR, 2.44; 95% CI, 1.52–3.93), and AKI (OR, 2.68; 95% CI, 1.31–5.55).531 In patients with hypertension, hypoperfusion may occur at higher BP levels and peri-operative BP control should be tailored to pre-operative levels.528

In patients referred for elective NCS, control of BP should be prioritized, especially in patients with systolic BP >160 mmHg. The management of patients with hypertension in the pre-operative setting should follow the recommendation provided in the 2018 ESC/European Society of Hypertension (ESH) Guidelines for the management of arterial hypertension.529 These guidelines advocate an approach using RAAS inhibitors (in patients aged <70 years) or CCBs (in patients aged >70 years) as single therapy in moderate hypertension and both in combination where dual therapy is needed, adding a diuretic and an aldosterone antagonist if additional medication is needed for adequate control. Beta-blockers are restricted to patients where it is specifically indicated.529 In patients with hypertension and a clear indication for beta-blocker, third-generation beta-blockers—such as carvedilol, celiprolol, labetolol, and nebivolol—have superior antihypertensive effects compared with other beta-blockers and fewer adverse effects, but there are no RCTs reporting outcomes in hypertensive patients.529 In a large observational study, a beta-blocker prescription prior to NCS was associated with lower 30 day mortality in patients with three or four cardiac risk factors.188 However, for patients with no cardiac risk factors, the risk of death was significantly increased with beta-blockers.188,532

Most patients with stage 3 hypertension on the day of surgery will be classified as hypertensive urgencies. In these cases, the 2018 ESC/ESH Guidelines for the management of arterial hypertension recommend ACEIs, CCBs, or diuretics. Neither ACEIs nor diuretics are recommended on the day of surgery. The use of CCBs is supported by a meta-analysis of 11 studies of CCBs during NCS.220 Further, a study of 989 well-controlled patients with hypertension without hypertension-related organ damage, testing the use of fast-acting nasal nifedipine when stage 3 hypertension (systolic BP >180 and/or diastolic BP >110 mmHg) was diagnosed the day of NCS, found no difference in outcome between administration of nifedipine and surgery on the same day compared with deferral for hypertension control before resuming surgery.533 Immediate nifedipine treatment was associated with shorter hospital stays. As this was the first study testing the need for deferral of stage 3 hypertension, it challenged the need for this practice. For patients with hypertensive emergencies (systolic BP >180 and/or diastolic BP >110 mmHg and organ damage), the 2018 ESC/ESH Guidelines for the management of arterial hypertension recommend labetolol, nitroglycerin, nitroprusside, etc., according to the affected organ.529

The timing of administration of the antihypertensive drugs, and their continuation or discontinuation in the peri-operative period is discussed in Section 5.2.

Recommendation Table 27

Recommendations for pre-operative management of hypertension

graphic
graphic
Recommendation Table 27

Recommendations for pre-operative management of hypertension

graphic
graphic

6.9. Peripheral artery disease

Patients with peripheral artery disease (PAD) usually have advanced atherosclerotic disease affecting multiple vascular beds in varying degrees and have a worse prognosis compared with patients without PAD.534–538 Patients with PAD generally differ in their risk profiles, according to whether they undergo vascular or non-vascular NCS.

6.9.1. Peripheral artery disease and non-vascular non-cardiac surgery

Decisions on pre-operative treatment of pre-existing PAD and AAA in patients scheduled for non-vascular NCS should be made on an individual basis, taking into account symptoms and risks of surgery. Non-cardiac surgery should be prioritized in patients needing revascularization for PAD, but careful peri-operative monitoring of deterioration in lower extremity perfusion is warranted, particularly in those patients with chronic limb-threatening ischaemia (e.g. peripheral BP of ≤50–70 mmHg in the foot joint and ≤30–50 mmHg in the toes).539 For patients with AAA, pain control is essential to ensure stable BP, minimizing rupture risk. Patients with large AAA (i.e. >5 cm in diameter for women and >5.5 cm for men) should be evaluated for aortic aneurysm repair (preferably EVAR)540–542 before non-vascular NCS is planned, particularly in the case of malignant tumours, depending on the stage of malignant disease.

6.9.2. Peripheral artery disease and vascular non-cardiac surgery

The 2017 ESC Guidelines on the diagnosis and treatment of peripheral arterial diseases535 and the European Society for Vascular Surgery (ESVS) 2019 Clinical Practice Guidelines on the management of abdominal aorto-iliac artery aneurysms35 provide detailed evidence and recommendations on the screening of patients before vascular NCS and their treatment modality selection. Notably, there is randomized evidence against prophylactic coronary revascularization before major vascular surgery in CCS patients due to lack of benefit in improvement of peri-operative and long-term outcomes (2.7 years mean follow-up).399 Assessment of functional capacity might offer guidance to select candidates for cardiac assessment prior to major NCS, although severe walking impairment may challenge this test. The previously established risk predictive model to detect mortality in patients undergoing elective AAA repair may be helpful.543 Low-dose rivaroxaban plus aspirin initiated within 10 days after a lower limb revascularization procedure reduces post-operative thrombotic events (acute limb ischaemia, amputation, MI, ischaemic stroke, and CV death) in patients undergoing lower limb revascularization, whether it is carried out using an endovascular or open surgical approach.544 Handling of other co-drugs should follow the recommendations detailed in Section 5.2.

Recommendation Table 28

Recommendations for management of patients with peripheral artery disease and/or abdominal aortic aneurysm undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 28

Recommendations for management of patients with peripheral artery disease and/or abdominal aortic aneurysm undergoing non-cardiac surgery

graphic
graphic

6.10. Cerebrovascular disease

Patients undergoing NCS should be questioned about previous neurological symptoms, and those with symptoms suggestive of transient ischaemic attack (TIA) or stroke in the preceding 6 months should undergo pre-operative neurological consultation and neurovascular and brain imaging, if appropriate. In the absence of dedicated studies addressing this issue, the criteria for carotid revascularization in symptomatic and asymptomatic patients is described in detail in the 2017 ESC Guidelines on the diagnosis and treatment of peripheral arterial diseases, in collaboration with the ESVS; these guidelines should also guide the management of patients with carotid disease who are undergoing NCS.535

In patients with symptomatic carotid disease, the benefit of carotid revascularization is particularly high in patients with recent (<3 months) TIA or stroke, and ≥70% carotid artery stenosis. Thus, carotid revascularization should be performed first and elective NCS should be postponed.545

The question as to whether patients with severe asymptomatic carotid occlusive disease who are undergoing elective major NCS require pre-operative carotid revascularization remains a matter of debate. Importantly, the purpose of carotid revascularization in this setting is more the long-term prevention of stroke than peri-operative stroke risk reduction; therefore, if carotid revascularization is indicated, this may be performed before or after the planned NCS. Independent of the revascularization strategy, patients with carotid artery stenosis benefit from aggressive CV risk-factor modification to prevent peri-operative myocardial ischaemia.

Recommendation Table 29

Recommendations for management of patients with suspected or established carotid artery disease undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 29

Recommendations for management of patients with suspected or established carotid artery disease undergoing non-cardiac surgery

graphic
graphic

6.11. Renal disease

Renal disease is associated with several cardiac comorbidities, including hypertension, HF, CAD, and arrhythmias.546 Consistently, renal disease portends a significant increase in the post-operative risk of CV events, including MI, stroke, and progression of HF in patients undergoing NCS. For this reason, most risk indices for the quantification of pre-operative risk in patients undergoing NCS include renal function.

Chronic kidney disease (CKD) is defined as impaired kidney function or raised proteinuria, confirmed on two or more occasions at least 3 months apart. The kidney function can be assessed through the estimated glomerular filtration rate (eGFR) calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) formula, including sex, age, ethnic origin, and serum creatinine concentration. A cut-off glomerular filtration rate (GFR) value <60 mL/min/1.73 m2 significantly correlates with MACEs. Identification of cardiac patients at risk of worsening of renal function in the peri-operative phase of NCS is of paramount importance, in order to initiate supportive measures such as maintenance of adequate intravascular volume for renal perfusion and use of vasopressors.547

Patients with cardiac comorbidities are prone to develop AKI after major NCS, due to worsening of haemodynamic status associated with fluids or blood loss, and withdrawal or continuation of cardio-active therapies. The peri-operative management of patients undergoing NCS and treated with cardio-active drugs has been discussed in Section 5.2. Acute kidney injury reduces long-term survival in patients with normal baseline renal function.548 Of interest, ∼30–40% of all cases of AKI occur after surgery and the incidence of post-operative AKI ranges 18–47%. Risk factors for the development of post-operative AKI following NCS include cardiac (decompensated or chronic HF, hypertension, cardio-active drugs) and non-cardiac triggers (age, sex, emergent, and/or intraperitoneal surgery, mild pre-operative renal insufficiency, pre-operative creatinine elevation, CKD, and DM).549,550 The combination of a low cardiac output/high venous pressure and/or the administration of iodinated contrast media during diagnostic and operative procedures represent the most frequent causes of AKI in hospitalized cardiac patients, regardless of pre-existing impaired renal function. Contrast-induced AKI is defined as a rise in serum creatinine of 44 mmol/L (0.5 mg/dL) or a 25% relative rise from baseline at 48 h (or 5–10% at 12 h) following contrast administration. It occurs in up to 15% of patients with CKD who are undergoing radiographic procedures.551 Although most cases of contrast-induced AKI are self-limiting, with renal function returning to normal within 7 days of the procedure, these patients occasionally (0.5–12% of cases) develop overt renal failure associated with increased morbidity and mortality. To reduce the risk of contrast-induced AKI in subjects requiring contrast-enhanced radiography, the Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guidelines recommend: pre-operative hydration with i.v. isotonic fluids; the use of the minimum volume of contrast media; and the use of low-osmolar or iso-osmolar contrast media, regardless of pre-existing renal disease.547 In the post-operative phase, kidney function should be monitored by applying established AKI scoring systems to raise awareness and enable early intervention.552

Recommendation Table 30

Recommendations for management of patients with renal disease undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 30

Recommendations for management of patients with renal disease undergoing non-cardiac surgery

graphic
graphic

6.12. Obesity

The prevalence of people being overweight and having obesity is reaching epidemic proportions in Western countries557 and is the second leading cause of preventable death following tobacco use.558 Obesity is defined as a body mass index (BMI) of ≥30 kg/m2, morbid obesity as a BMI ≥35 kg/m2, and super-morbid obesity as a BMI ≥50 kg/m2. Obese individuals have a higher prevalence of CV risk factors and a higher risk of death,559 and are a population who are at increased risk of adverse events in the case of surgical procedures. There are specific recommendations for the pre-operative risk assessment of obese patients undergoing NCS, regardless of the presence of pre-existing cardiac conditions.560 However, while obesity accelerates the propensity for CVD, it seems that many types of CVD may have a better prognosis in the overweight population compared with their leaner counterparts, a phenomenon that is known as the ‘obesity paradox’.561,562 Similarly, in cases of NCS, mildly obese patients present lower mortality risk compared with underweight and normal weight patients, both post-operatively and at long-term follow-up.563 This finding may be related to the lower prevalence of PMI in mildly obese patients undergoing NCS.564

It has been suggested that cardiorespiratory fitness (CRF), rather than BMI, should be used to assess CV risk in obese patients. While the classification based on BMI is simple, highly reproducible, and widely adopted in clinical practice, it does not reflect fat distribution and body composition. Cardiorespiratory fitness refers to the ability of the circulatory and respiratory systems to supply oxygen to skeletal muscles during sustained physical activity, which is of paramount importance, especially in patients with cardiac diseases. The primary measure of CRF is VO2 max.565 A cohort study of nearly 10 000 patients with CAD followed for almost 15 years showed that those with relatively good CRF had favourable prognosis regardless of body composition;566 however, a lower CRF was found to be a major predictor of mortality, regardless of BMI.561,567 Whether specific optimization and/or treatment strategies might have a positive impact on the outcome of obese patients with pre-existing or newly diagnosed cardiac comorbidities and scheduled for NCS is a matter of ongoing controversy. Studies assessing the effect of weight loss interventions (low-energy diets with or without an exercise component) on clinical outcomes in patients undergoing NCS found inconsistent results in terms of peri-operative morbidity or mortality.568,569

Recommendation Table 31

Recommendations for management of patients with obesity undergoing non-cardiac surgery

graphic
graphic
Recommendation Table 31

Recommendations for management of patients with obesity undergoing non-cardiac surgery

graphic
graphic

6.13. Diabetes

Due to the progressive ageing of the population undergoing surgical procedures and the increasing prevalence of obesity worldwide, the prevalence of diabetes among patients undergoing NCS is expected to increase in years to come.4,571 Several studies have demonstrated that diabetic patients undergoing NCS have a higher prevalence of CAD than non-diabetic patients. Furthermore, patients with diabetes are more likely to have silent ischaemia because of altered neural pain pathways in the heart.572 For this reason, patients with diabetes appear to have a greater risk of post-operative myocardial ischaemia. Different reasons exist behind the relation between DM and increased peri-operative mortality in patients undergoing NCS. First, patients with diabetes are known to have more comorbidity and/or advanced CAD at the time of intervention. Second, diabetes is a clear risk factor for stroke. Diabetes is associated with post-operative congestive HF and wound infections. Many patients with diabetes have impaired renal function. The presence or a new diagnosis of impaired glucose metabolism in patients scheduled for NCS should follow the recommendations provided for the general population in the 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases,573 including increased awareness regarding possible subclinical organ damage.

The glycated haemoglobin (HbA1c) test should be performed in all patients with diabetes or impaired glucose metabolism scheduled for NCS, if this measurement has not been performed in the previous 3 months. There is evidence to support that pre-admission optimal treatment of hyperglycaemia in patients scheduled for elective NCS is effective in reducing the post-operative risk of CV events, including MI, stroke, and progression of HF.574 In contrast, no clear association has been shown between intra-operative blood glucose levels and the subsequent risk of surgical site infection, MI, stroke, and death in patients undergoing NCS.575 The risk of acidosis associated with metformin use is also debated.576 Nevertheless, repeated blood glucose monitoring on the day of surgery is recommended, with a general consensus to maintain peri-operative glucose levels <10.0 mmol/L without causing hypoglycaemia (target level 5.6–10.0 mmol/L). This can be achieved either with subcutaneous doses of rapid-acting insulin analogues or with i.v. insulin.577 Handling of SGLT2 inhibitors in relation to surgery has been discussed in Section 5.2.

Recommendation Table 32

Recommendations for management of patients with diabetes mellitus undergoing non-cardiac surgery

graphic
graphic
graphic
graphic
Recommendation Table 32

Recommendations for management of patients with diabetes mellitus undergoing non-cardiac surgery

graphic
graphic
graphic
graphic

6.14. Cancer

Due to their generally older age, cancer patients have a high prevalence of CV risk factors and CVD, being a population at increased risk of adverse events in cases of NCS. It is therefore important to optimize treatment of CV risk factors and known CVD before NCS, following the general and disease-specific recommendations provided in other sections of these guidelines. Furthermore, NCS may be particularly challenging in cancer patients because of previous administration of potentially cardiotoxic chemotherapy or fibrosis due to previous radiation. For example, the widely used anthracyclines have a dose-dependent relation with the incidence of HF and the use of trastuzumab can lead to important cardiotoxicity and should be taken into account in the pre-operative assessment. Furthermore, radiation therapy to areas that included the heart may lead to premature CAD and VHD; previous thoracic radiotherapy may predispose younger patients to heart disease who would otherwise not have an elevated risk. Post-operative AF is frequently observed in patients undergoing cancer surgery, with the highest incidence reported for lung surgery. Patients with cancer are at elevated risk of thrombosis due to both the disease itself and patient- and treatment-related factors. In a small study of patients undergoing planned open surgery for abdominal or pelvic cancer, enoxaparin prophylaxis for 4 weeks compared with 1 week reduced the incidence of thrombosis (4.8% in the enoxaparin group vs.12.0% in the placebo group; P = 0.02).582 Although later studies have shown somewhat conflicting results, the consensus is to recommend extending thromboprophylaxis after major abdominal and/or pelvic surgery for cancer to 4–5 weeks, with preferred use of LMWH.583 A summary of patient-related and cancer therapy-related factors that could influence peri-operative risk is shown in Table 15. Further information is available in the 2022 ESC Guidelines on cardio-oncology.584

Table 15

Factors that could influence peri-operative risk during cancer surgery and preventive strategies

Factors that could influence peri-operative risk during cancer surgeryPreventive strategies
  • Patient-related factors

  • Lifestyle risk factors—smoking, obesity, sedentary lifestyle

  • Poorly controlled CV risk factors—hypertension, diabetes

  • Pre-existing CVD, including cancer therapy-related cardiovascular toxicity

  • Cardiac medication increasing peri-operative bleeding risk (e.g. antiplatelets and anticoagulants)

  • Historical primary malignancy

  • Current cancer type, stage, and location

  • Arrhythmias (due to myocardial cancer invasion, induced QT-prolongation, AF, or imbalance of autonomic nervous system)

  • Optimal management of CV risk factors and CVD

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

  • ECG monitoring for arrhythmias

  • Correction of all proarrhythmic conditions

  • Neoadjuvant cancer therapy

  • Previous cardiotoxic cancer treatments (especially anthracycline chemotherapy and/or trastuzumab; immune checkpoint inhibitors, VEGFi, fluoropyrimidine and thoracic radiotherapy)

  • Cancer treatments increasing peri-operative bleeding risk (e.g. antiangiogenics, BTKi)

  • Cancer treatments increasing risk of arrhythmias

  • Ensure optimal CV monitoring of neoadjuvant therapy

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

Factors that could influence peri-operative risk during cancer surgeryPreventive strategies
  • Patient-related factors

  • Lifestyle risk factors—smoking, obesity, sedentary lifestyle

  • Poorly controlled CV risk factors—hypertension, diabetes

  • Pre-existing CVD, including cancer therapy-related cardiovascular toxicity

  • Cardiac medication increasing peri-operative bleeding risk (e.g. antiplatelets and anticoagulants)

  • Historical primary malignancy

  • Current cancer type, stage, and location

  • Arrhythmias (due to myocardial cancer invasion, induced QT-prolongation, AF, or imbalance of autonomic nervous system)

  • Optimal management of CV risk factors and CVD

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

  • ECG monitoring for arrhythmias

  • Correction of all proarrhythmic conditions

  • Neoadjuvant cancer therapy

  • Previous cardiotoxic cancer treatments (especially anthracycline chemotherapy and/or trastuzumab; immune checkpoint inhibitors, VEGFi, fluoropyrimidine and thoracic radiotherapy)

  • Cancer treatments increasing peri-operative bleeding risk (e.g. antiangiogenics, BTKi)

  • Cancer treatments increasing risk of arrhythmias

  • Ensure optimal CV monitoring of neoadjuvant therapy

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

AF, atrial fibrillation; BTKi, Bruton tyrosine kinase inhibitors; CV, cardiovascular; CVD, cardiovascular disease; ECG, electrocardiogram; VEGFi, vascular endothelial grow factor inhibitor; VTE, venous thromboembolism.

Table 15

Factors that could influence peri-operative risk during cancer surgery and preventive strategies

Factors that could influence peri-operative risk during cancer surgeryPreventive strategies
  • Patient-related factors

  • Lifestyle risk factors—smoking, obesity, sedentary lifestyle

  • Poorly controlled CV risk factors—hypertension, diabetes

  • Pre-existing CVD, including cancer therapy-related cardiovascular toxicity

  • Cardiac medication increasing peri-operative bleeding risk (e.g. antiplatelets and anticoagulants)

  • Historical primary malignancy

  • Current cancer type, stage, and location

  • Arrhythmias (due to myocardial cancer invasion, induced QT-prolongation, AF, or imbalance of autonomic nervous system)

  • Optimal management of CV risk factors and CVD

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

  • ECG monitoring for arrhythmias

  • Correction of all proarrhythmic conditions

  • Neoadjuvant cancer therapy

  • Previous cardiotoxic cancer treatments (especially anthracycline chemotherapy and/or trastuzumab; immune checkpoint inhibitors, VEGFi, fluoropyrimidine and thoracic radiotherapy)

  • Cancer treatments increasing peri-operative bleeding risk (e.g. antiangiogenics, BTKi)

  • Cancer treatments increasing risk of arrhythmias

  • Ensure optimal CV monitoring of neoadjuvant therapy

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

Factors that could influence peri-operative risk during cancer surgeryPreventive strategies
  • Patient-related factors

  • Lifestyle risk factors—smoking, obesity, sedentary lifestyle

  • Poorly controlled CV risk factors—hypertension, diabetes

  • Pre-existing CVD, including cancer therapy-related cardiovascular toxicity

  • Cardiac medication increasing peri-operative bleeding risk (e.g. antiplatelets and anticoagulants)

  • Historical primary malignancy

  • Current cancer type, stage, and location

  • Arrhythmias (due to myocardial cancer invasion, induced QT-prolongation, AF, or imbalance of autonomic nervous system)

  • Optimal management of CV risk factors and CVD

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

  • ECG monitoring for arrhythmias

  • Correction of all proarrhythmic conditions

  • Neoadjuvant cancer therapy

  • Previous cardiotoxic cancer treatments (especially anthracycline chemotherapy and/or trastuzumab; immune checkpoint inhibitors, VEGFi, fluoropyrimidine and thoracic radiotherapy)

  • Cancer treatments increasing peri-operative bleeding risk (e.g. antiangiogenics, BTKi)

  • Cancer treatments increasing risk of arrhythmias

  • Ensure optimal CV monitoring of neoadjuvant therapy

  • Optimize preventive strategies with respect to VTE and arterial thromboembolic events

AF, atrial fibrillation; BTKi, Bruton tyrosine kinase inhibitors; CV, cardiovascular; CVD, cardiovascular disease; ECG, electrocardiogram; VEGFi, vascular endothelial grow factor inhibitor; VTE, venous thromboembolism.

6.15. Coronavirus disease 2019

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In a recent observational study, among 140 231 patients scheduled for NCS, 2.2% of patients had a pre-operative diagnosis of SARS-CoV-2 infection.585 Patients undergoing surgery with peri-operative or recent SARS-CoV-2 appear to be at increased risk of post-operative VTE and mortality compared with patients with no history of SARS-CoV-2 infection.586 Furthermore, the possible myocardial injury associated with COVID-19 infection might increase the intrinsic peri-operative risk of adverse cardiac events associated with surgical procedures.587,588

To date, there is no specific CV screening to be performed after COVID-19 infection in patients scheduled for NCS. The pre-operative evaluation of CV risk associated with NCS in this specific subset of patients should incorporate, beyond the general risk assessment outlined in these guidelines, the severity of COVID-19 disease, history of CV complications during COVID-19 infection, and functional capacity after recovery. This information is deemed of importance to further optimize timing of surgery and the treatment of COVID-19-associated cardiovascular conditions affecting the peri-operative risk of NCS.589

The optimal timing of NCS in patients with a history of COVID-19 infection is largely unknown. Generally, elective NCS should be postponed until complete recovery and optimization of coexisting comorbidities. Registry data of patients undergoing NCS after COVID-19 infection report increased risks of mortality and morbidity up to 7 weeks post-COVID diagnosis.586 Another study has reported greater risk of post-operative complications up to 8 weeks post-diagnosis.590 However, it should be noted that almost all available data come from study periods with zero to low prevalence of vaccination, and no robust data exist on patients recovering from the more recent Delta and Omicron variants.

A joint statement on elective surgery and anaesthesia for patients after COVID-19 infection from the American Society of Anaesthesiologists and Anaesthesia Patient Safety Foundation591 suggests delaying elective surgery for 7 weeks after a SARS-CoV-2 infection in unvaccinated patients who are asymptomatic at the time of surgery. The clinical course of COVID-19 infection, presence and intensity of cardiopulmonary symptoms during the acute and late phases, and pre-existing comorbidities should be taken into consideration. In addition, persistence of COVID-19 symptoms—such as fatigue, shortness of breath, and chest discomfort—should be given attention, as this condition is associated with higher post-operative mortality independent of the timing of COVID-19 diagnosis.588,589 The evidence is currently insufficient to make recommendations for those who become infected after vaccination.

7. Peri-operative monitoring and anaesthesia

7.1. Peri-operative monitoring

Despite the absence of prospective RCTs investigating the prognostic relevance of peri-operative monitoring, previous evidence clearly indicates that the routine use of monitoring improves safety of surgical procedures. Mandatory intra-operative monitoring of the CV and respiratory systems, temperature, neuromuscular transmission, and depth of anaesthesia is recommended.592,593

Routine CV monitoring includes ECG, automated non-invasive BP measurement at regular intervals, and peripheral oxygen saturation with pulse oximetry. Near-infrared spectroscopy has recently been introduced to assess regional tissue perfusion and oxygenation. In selected cases, more invasive monitoring techniques can be applied such as continuous arterial BP measurement via an arterial catheter and monitoring of cardiac output. Mean arterial pressure and heart rate remain stable, even with variation of up to 30% of total blood volume.594 Right heart catheterization can be used to continuously measure central venous pressure and/or pulmonary artery pressure, pulmonary artery wedge pressure (as a reflection of LV diastolic pressure), and cardiac output. However, these are static variables that do not reliably reflect the CV filling status, and have been shown to accurately guide fluid therapy in 50% of patients.595

While routine use of pulmonary catheterization is discouraged during NCS, using dynamic variables, such as stroke volume variation or pulse pressure variation, has become the gold standard. In addition, TEE is increasingly being used as an intra-operative monitoring technique in major surgery in cardiac compromised patients and during cardiac surgery. Decisions on the extent of peri-operative monitoring and implementation of specific strategies during NCS should always be based on an individual patient-directed assessment, taking into consideration the severity of surgery and the patient’s physical condition. Basic peri-operative monitoring of the respiratory system consists of pulse oximetry and capnography; both methods are non-invasive and easily applicable. Pulse oximetry enables in vivo measurement of arterial oxygen saturation, and capnometry measures end-tidal carbon dioxide concentration during inspiration and expiration; these monitors also provide information about the global haemodynamic status.

Patients, independent of the anaesthetic technique and information provided by the different monitoring systems, must be regularly controlled.596 In addition, blood loss and urine output should be checked, when appropriate, and overall clinical status.597 Special attention is required for the activation and setting of audible alarms, as inadequate use or failure to respond to intra-operative alarms may result in patient hazard and undesirable outcomes.598

7.2. Anaesthesia

The decision on the optimal peri-operative strategy should be based on close exchange of clinical information between anaesthesiologists, cardiologists, surgeons, and other relevant specialists. In addition, it is mandatory that any proposed strategy is presented to and discussed with the patient. An informed discussion with the patient describing the planned patient pathway and expectations during the pre-, peri-, and post-operative phases of care, and what to expect from staff and surroundings, should be given using a clear, concise, and simple description. The ESA published Pre-operative evaluation of adults undergoing elective noncardiac surgery: updated Guideline from the European Society of Anaesthesiology in 2018.560 This current section focuses on issues that are specifically important to patients with CV risk factors and diseases, taking into account the most recent developments in peri-operative management of these patients.

7.2.1. Intra-operative haemodynamics

Most anaesthetic techniques reduce sympathetic tone, leading to a decrease in venous return due to increased compliance of the venous system, vasodilatation, and decreased BP. Therefore, maintenance of adequate organ flow and perfusion pressure is of key importance in anaesthesiological management, especially in the CV-compromised patient. The importance of keeping stable peri-operative haemodynamics has been recognized for many years.599

In the past few years, several studies have focused on the relationship between intra-operative hypotension and post-operative patient outcome. A recent systematic review identified 42 studies looking at associations between various absolute and relative intra-operative hypotension definitions and post-operative adverse outcomes after NCS.214 The reported associations suggest that organ injury (myocardial injury, stroke, AKI) might occur when the mean arterial pressure decreases to <80 mmHg for ≥10 min, and that this risk increases with BP becoming progressively lower. However, most of the included studies had a retrospective observational design with a large variability in patient characteristics. In addition, the definitions of intra-operative hypotension varied widely across included studies. A recent study on the incidence of intra-operative hypotension as a function of the chosen cut-off definition described 48 different definitions of intra-operative hypotension. When applying these definitions to a cohort of 15 509 consecutive adult patients undergoing NCS under general anaesthesia, any episode of systolic BP <80 mmHg was found in 41% of the patients, and 93% of the patients had at least one episode of systolic BP >20% below baseline. The relation between threshold values from the literature and incidence of intra-operative hypotension showed a sigmoidal shaped cumulative incidence curve, with intra-operative hypotension occurrence frequencies varying from 5–99%.600 It seems that no universal target BP to define intra-operative hypotension can currently be defined. In addition, in studies on intra-operative hypotension, both the threshold to define hypotension and the method chosen to model intra-operative hypotension affected the association of intra-operative hypotension with outcome.601 As a consequence, different studies on intra-operative hypotension are uncomparable and clinical conclusions on reported results remain hazardous. A recent expert consensus statement concluded that intra-operative mean arterial pressures <60–70 mmHg are associated with myocardial injury, AKI, and death. These complications are a function of hypotension severity and duration.602 It remains to be established whether correction of intra-operative hypotension is also associated with improved post-operative outcome. To date, only one study has specifically addressed the question of whether an individualized BP management strategy reduces post-operative complications in a multicentre RCT including 292 patients. An individualized management strategy of targeting a systolic BP within 10% of the patient’s normal resting value resulted in significantly lower rates of post-operative organ dysfunction compared with standard practice (38.1% vs. 51.7%, respectively).528 These findings support the benefits of personalizing care, especially in surgical patients at high-risk of cardiac complications. It is important to underscore the importance of a physiopathological approach in understanding the underlying mechanisms of intra-operative hypotension, taking into account the extent and severity of the patient’s comorbidities; only then will a tailored treatment targeting the cause of intra-operative hypotension be possible (Figure 16).

Pathophysiological approach to address intra-operative hypotension.
Figure 16

Pathophysiological approach to address intra-operative hypotension.

aE.g. peri-operative myocardial failure.

The severity of intra-operative hypotension is defined both by the threshold decrease from baseline and the duration of the hypotensive episode. The underlying mechanisms for intra-operative hypotension need to be identified: sympathicolysis with vasodilation, hypovolaemia, or other more complex causes such as peri-operative myocardial failure. Once the underlying mechanisms are identified, a targeted therapeutic strategy can be applied. This includes considering whether administration of specific chronic vasoactive medication such as ACEIs or ARBs should be interrupted 24 h prior to surgery.216 A detailed analysis of the strategies for the different chronic CV medication is discussed in Section 5. Of note, post-operative outcome is not only negatively influenced by the occurrence of intra-operative hypotension, but also by hypotensive events during the initial four post-operative days.603 For adult non-cardiac surgical patients, there is insufficient evidence to recommend a general upper limit of arterial pressure at which therapy should be initiated, although pressures >160 mmHg have been associated with myocardial injury and MI.602

Intra-operative tachycardia may adversely affect the myocardial oxygen balance and thus result in peri-operative myocardial injury. A retrospective analysis of 41 140 patients found that a heart rate ≥90 b.p.m. was associated with an increased risk of myocardial injury.604 These findings were similar to observations in the Vascular Events in Noncardiac Surgery Patients Cohort Evaluation (VISION) trial.605 An analysis of high-risk surgical patients found that an HR >87 b.p.m., recorded at rest before pre-operative cardiopulmonary exercise testing, was strongly associated with distinct CV phenotypes, which may explain the unintended, adverse consequences of non-personalized treatments aimed at reducing intra-operative tachycardia in isolation from other haemodynamic factors.606 Finally, a recent study evaluated the association between intra-operative tachycardia and a composite of post-operative myocardial injury and all-cause mortality. The major finding of this study was the lack of an association between intra-operative heart rate >90 b.p.m. and the composite outcome; HR >80 b.p.m. or >100 b.p.m. were also not associated with the composite outcome.607 Taken together, the assumed association between intra-operative tachycardia and adverse peri-operative outcome remains a subject of ongoing controversy. However, it seems advisable to consider intra-operative tachycardia as an indicator of haemodynamic impairment.

7.2.2. Choice of anaesthetic agent

The choice of the anaesthetic agent has been considered to be of little importance in terms of patient outcome, provided that vital functions are adequately supported. Evidence from surgical myocardial revascularization studies indicates that a volatile vs. i.v. anaesthetic regimen is associated with less post-operative troponin release without impact on clinical outcomes.599,608 A recent large multicentre randomized trial found a neutral effect on 12 month mortality associated with volatile vs. i.v. anaesthetic regimen.609 In NCS, incidence of post-operative cardiac events is not influenced by the choice of a volatile or an i.v. anaesthetic regimen.610

7.3. Locoregional techniques

The analgesic effects of neuraxial analgesia are well-established. The main peri-operative indications for epidural analgesia include major open abdominal surgery and thoracotomy. Possible additional benefits of epidural analgesia, such as accelerated recovery and decreased post-operative complications, remain a matter of debate.611,612

Neuraxial analgesia may induce sympathetic blockade. When reaching the thoracic dermatome level 4, a reduction in cardiac sympathetic drive may occur, with subsequent reduction in myocardial contractility, heart rate, and change in cardiac loading conditions. There are no studies specifically investigating the changes in outcomes related to neuraxial anaesthetic techniques in patients with cardiac disease. Cardiac patients often take various types of drugs that interfere with coagulation; therefore, care should be taken to ensure sufficient coagulation ability when neuraxial blocks are applied.613

Current research is focusing on alternatives for neuraxial analgesia with similar effects on peri-operative pain control in patients with cardiac comorbidities undergoing NCS; these include alternative analgesic techniques such as i.v. analgesia, continuous wound infiltration, paravertebral bloc, and selective nerve blocks.

7.4. Peri-operative goal-directed haemodynamic therapy

Goal-directed therapy aims to optimize CV performance, in order to achieve normal or even supranormal oxygen delivery to tissues, by optimizing pre-load and inotropic function using pre-defined haemodynamic targets. In contrast to clinical signs or arterial pressure-orientated standard therapy, goal-directed therapy is based on flow or fluid responsiveness of haemodynamic variables, such as stroke volume, response to fluid challenges, stroke volume or pulse pressure variation, or similar optimization of cardiac output. Goal-directed therapy was initially based on the use of a pulmonary artery catheter. Less-invasive techniques have recently been developed, including: transoesophageal Doppler, transpulmonary dilution techniques, and advanced pressure waveform analysis. Early goal-directed fluid therapy—in the right patient cohort and with a clearly defined protocol—has been shown to decrease post-operative mortality and morbidity.614–618

7.5. Post-operative management

Several studies have demonstrated that it is possible to stratify the risk of post-operative complications and mortality with a simple surgical Apgar score. This post-event stratification might enable patients to be redirected to higher-intensity care units. The importance of such risk stratification is underscored by the results of the EuSOS group. In this 7 day cohort study, 46 539 consecutive adult NCS patients in 498 hospitals across 28 European nations were included: 1855 patients (4%) died before hospital discharge and 1358 (73%) of those patients were not admitted to critical care at any stage after surgery.7 This concept of failure-to-rescue has gained a lot of attention in peri-operative medicine in the last few years and strategies have been proposed to address this issue.619–622

Severe post-operative pain occurs in 5–10% of patients, increases sympathetic drive, and delays recovery.623,624 A recent study demonstrated that time-weighted average pain scores within 72 h after surgery were significantly associated with myocardial injury in patients undergoing NCS;625 this finding underscores the importance of effective post-operative analgesia to reduce post-operative CV risk.

The place of non-steroidal anti-inflammatory drugs (NSAIDs) in the treatment of post-operative pain in cardiac patients undergoing NCS is a point of debate. Diclofenac has been shown to carry the highest CV risk of any of the non-selective NSAIDs.626,627 The CV risk of ibuprofen seems to be comparable with that of celecoxib.628 Naproxen has a better CV safety profile than diclofenac and ibuprofen.629,630 One randomized trial found that parecoxib and valdecoxib did not increase thromboembolic events in patients undergoing NCS. A meta-analysis of 32 randomized trials did not find an increased CV risk when comparing parecoxib/valdecoxib with placebo,631 and a single-centre observational study with >10 000 patients undergoing arthroplasty found no association between NSAID use and post-operative MI.632 In contrast, another meta-analysis of three randomized trials including 2604 major surgery patients detected a 2.3-fold increase in MACEs in the group with COX-2 inhibitors. In a position paper, The ESC Working Group on Cardiovascular Pharmacotherapy advises that non-aspirin NSAIDs should generally not be used in patients with established or at high risk of CVD.633

Recommendation Table 33

Recommendations for peri-operative monitoring and anaesthesia

graphic
graphic
Recommendation Table 33

Recommendations for peri-operative monitoring and anaesthesia

graphic
graphic

8. Peri-operative cardiovascular complications

Specific challenges apply to detecting CV complications that occur peri-operatively. First, due to anaesthesia and analgesia, PMI, which is the most common CV complication, is largely asymptomatic in ∼90% of patients and is therefore missed in routine clinical practice in the absence of surveillance for PMI.41,101,111,413,636–641 Second, post-operative pain, nausea, surgical wounds, and drains may interfere with the early identification of acute cardiac disorders, such as PMI, Takotsubo syndrome, tachyarrhythmias, and acute HF. Third, cardiologists are usually not directly involved in post-operative care; therefore, the early detection and early treatment of cardiac complications is performed by non-cardiologists, sometimes with little training in the early detection of acute cardiac disorders. Given the relatively high prevalence of cardiac complications, their high morbidity and mortality, and the availability of effective therapy, high awareness combined with surveillance for PMI in high-risk patients (known CAD, PAD, insulin-dependent DM, or symptoms suggestive of cardiac disorders) undergoing intermediate- or high-risk NCS is recommended to overcome these challenges.41,101,109–111,118,413,636–639,642,643 All measures need to be carefully aligned with the responsible surgeon.

Chronic cardiac disorders, such as CAD, seem to provide a substrate for cardiac complications during and after surgery.41,413,636,637 Several related chronic conditions (e.g. diabetes and renal insufficiency), which are likely to be surrogates for undiagnosed cardiac disease, are also strongly associated with peri-operative cardiac complications (Figure 17).41,413,636,637

Factors associated with peri-operative cardiovascular complications. SNS, sympathetic nervous system.
Figure 17

Factors associated with peri-operative cardiovascular complications. SNS, sympathetic nervous system.

Acute conditions such as trauma, surgery, and anaesthesia itself induce activation of the sympathetic nervous system, inflammation, stress, hypercoagulable, and catabolic states, all of which may trigger cardiac complications.41,413,636,637 While the risk of CV complications after NCS is highest in the immediate peri-operative period, it seems to remain increased for a prolonged ‘vulnerable period’ of 3–5 months.8

8.1. Peri-operative myocardial infarction/injury

Peri-operative MI (PMI) is defined as acute cardiomyocyte injury (post-operative hs-cTn T/I release) with or without accompanying symptoms, and with or without ECG or imaging evidence of acute myocardial ischaemia. Peri-operative MI can only be reliably and rapidly detected using PMI surveillance with hs-cTn T/I measurements before and serially after surgery (e.g. 24 and 48 h post-operatively). In the BASEL-PMI study, circa 15% of patients with pre-existing CAD/PAD or aged >65 years undergoing major NCS developed PMI.8 As most PMI occurs during the operation itself or in the immediate post-operative period, during which high doses of anaesthetics and/or analgesics are required, ∼90% of patients with PMI do not report typical symptoms and are therefore missed in routine clinical practice.41,101,111,413,636–639 This is of major concern, as the mortality risk associated with PMI is also high in patients without symptoms.41,101,111,413,636–639 Similarly, the mortality risk associated with PMI is high in patients without additional ECG and/or imaging evidence of myocardial ischaemia.8,41,101,109–111,118,413,564,636–639,641 Overall, 30 day mortality in patients developing PMI is ∼10%.8,41,101,109–111,118,413,564,636–639,641 No single intervention has yet been proven to be unequivocally beneficial in the prevention of PMI.185,644

It is important to highlight that PMI is not a homogenous disease. Several different pathophysiologies and clinical phenotypes may underlie PMI (Figure 18). At least one additional criterion (ischaemic pain; ischaemic ECG changes; imaging evidence of new loss of viable myocardium, or new regional wall motion abnormality in a pattern consistent with an ischaemic aetiology; and thrombus in coronary angiography) is required for patients with PMI to also meet the definition of peri-operative myocardial infarction, according to the fourth universal definition.643 Because is often initially unclear whether the patient will end up fulfilling the criteria for peri-operative myocardial infarction, the broad term PMI is preferred in the initial assessment. In order to properly interpret the aetiology of elevated post-operative hs-cTn T/I concentrations, a baseline pre-operative concentration is necessary to determine whether the increase is acute or chronic (see Section 4).643 To identify the underlying pathophysiology and define causal therapy, systematic work-up and early differentiation of primarily non-cardiac causes (e.g. severe sepsis, PE) vs. the different cardiac causes—including type-1 MI, type-2 MI, tachyarrhythmia, and acute HF—is of major importance (Figures 18 and 19). Transthoracic echocardiography is helpful in the work-up of most patients with PMI.

Differential diagnosis of elevated post-operative cardiac troponin concentrations.
Figure 18

Differential diagnosis of elevated post-operative cardiac troponin concentrations.

CK, creatinine kinase; cTn l, cardiac troponin I; cTn T, cardiac troponin T; MI, myocardial infarction; PMI, peri-operative myocardial infarction/injury. Please be aware that the accuracy of physicians’ judgement in the classification of type-1 vs. type-2 MI in the peri-operative setting may be lower vs. the non-operative setting.647

Systematic work-up (aetiology) and therapy of peri-operative myocardial infarction/injury.
Figure 19

Systematic work-up (aetiology) and therapy of peri-operative myocardial infarction/injury.

CCTA, coronary computed tomography angiography; ECG, electrocardiogram; Hb, haemoglobin; ICA, invasive coronary angiography; MI, myocardial infarction; N, no; ST, ST-segment. Y, yes; Most patients with type-2 MI and silent type-1 MI should be scheduled for stress imaging or CCTA/ICA as outpatients after discharge, depending on symptoms prior to or after surgery and known CAD. aOr active bleeding. bOr other type-2 MI trigger such as hypoxaemia, tachycardia, hypertension. cDual antiplatelet therapy after coronary stenting. dPossibly in combination with dabigatran 110 mg b.i.d.

The term myocardial injury following NCS (MINS) has been used for a selected subset of patients with cardiac PMI, in whom cardiomyocyte injury was deemed most likely due to CAD with myocardial ischaemia (i.e. secondary to supply–demand mismatch or thrombosis), in the absence of an atypical surgical physiological stress, and no evidence of a cardiac non-CAD aetiology, e.g. rapid AF, acute HF).101,109,638,641,642,645,646 In approximately half of all patients with PMI, the underlying pathophysiology cannot be reliably ascertained based on the commonly available documentation and is assumed to be likely type-2 MI due to undocumented or relative hypotension, but may also include missed type-1 MI, or missed PE in cases in which CT angiography has not yet been performed post-operatively.110,647 It is therefore important to highlight that PMI surveillance also facilitates the detection of primarily non-cardiac disorders with immediate therapeutic consequences, such as PE, that would otherwise have been missed.

The prevalence of PMI depends on patient-related, procedural-related, and post-operative factors, and the required minimum extent of acute myocardial injury, quantified by absolute increase in hs-cTn T/I (e.g. the 99th percentile ULN) above the pre-operative hs-cTn T/I concentration.8,41,101,109–111,118,413,564,636–639,641,648 An absolute increase in more than the ULN above pre-operative concentrations has consistently been shown to be associated with a relevant increase in 30 day and long-term mortality, and other non-fatal post-operative cardiac complications, and can easily be determined for each hs-cTn T/I assay. This threshold is therefore recommended for clinical use.8,41,101,109–111,118,413,564,636–639,641,648 However, further studies are warranted regarding optimal thresholds. Emerging data suggest that surveillance for PMI is cost-effective.117,119

Identification of the most likely pathophysiology is critical for selection of the most appropriate therapy. Evidence from a large single-centre retrospective cohort suggests that involvement of a cardiologist in the work-up and therapy of these patients was associated with lower mortality.649 Mortality substantially differs among the different phenotypes: 30 day all-cause mortality and the composite of 30 day CV death, life-threatening arrhythmia, acute MI occurring after day 3, and cardiac decompensation are highest in patients with acute HF and primarily extra-cardiac PMI, such as severe sepsis or PE, intermediate for type-1 MI and tachyarrhythmias, and modestly elevated in likely type-2 MI.110 Type-2 MI patients are usually treated as for type-1 MI, although the evidence for this is limited.

In a randomized, placebo-controlled trial, 1754 patients (mean age 70 years) who had developed MINS after NCS (mainly orthopaedic, general, and vascular surgery) were randomly assigned (1:1) to receive dabigatran 110 mg orally b.i.d. or matched placebo within 35 days of MINS;650 60% of patients were already on aspirin or a P2Y12 inhibitor. The median peak measured hs-cTn concentration associated with the diagnosis of MINS was 82 ng/L. More than 90% of MINS events occurred without a clinical symptom or sign of cardiac ischaemia. Dabigatran/placebo was initiated a median of 6 days after the operation and the average time on study drug was ∼9 months. Among patients with MINS randomly assigned to receive dabigatran (n = 877) or placebo (n = 877), the composite primary efficacy outcome of a major vascular complication—including vascular mortality, MI, non-haemorrhagic stroke, peripheral arterial thrombosis, amputation, and symptomatic VTE—occurred in fewer patients randomized to dabigatran than placebo (97 [11%] of 877 patients assigned to dabigatran vs. 133 [15%] of 877 patients assigned to placebo; HR, 0.72; 95% CI, 0.55–0.93; P = 0.0115). There was no increase in major bleeding. Based on these data, in patients with MINS and at low risk of bleeding, initiation of dabigatran 110 mg orally b.i.d. may be considered about 1 week after NCS.

8.2. Spontaneous myocardial infarction (after day 2)

The incidence of post-operative spontaneous MI after day 2 seems to be about 0.5% within 30 days, and 1–2% within 365 days for patients undergoing major NCS with established CAD, PAD, or aged >65 years.8 In the immediate post-operative period (<5 days), bleeding is a major concern and limits the use of antiplatelet and anticoagulant therapy, depending on the site and extent of surgery. Otherwise, the same principles as for MI therapy should generally be applied following the recommendations of speciality guidelines.98,171

8.3. Takotsubo syndrome

The incidence of peri-operative Takotsubo syndrome remains unknown, as none of the studies with PMI screening used echocardiography in all patients during PMI work-up. Increased awareness in the non-operative setting has led to a substantial increase in the detection of Takotsubo syndrome, and the use of TTE in the work-up of PMI is strongly encouraged. Anecdotal evidence suggests that it is likely that the incidence is also higher than expected in the peri-operative setting.8,41,101,109–111,118,413,564,636–639,641,644

8.4. Acute heart failure

The incidence of post-operative acute HF seems to be 1–2% within 30 days and 4–6% within 365 days in patients with established CAD, PAD, or aged >65 years undergoing major NCS.8 Pre-existing diagnosed or undiagnosed chronic HF and volume loading in the peri-operative and post-operative periods are important contributors. In the absence of studies specifically investigating acute HF post-operatively, the general principles of acute HF diagnostic work-up and therapy should be applied.651

8.5. Venous thromboembolism

The incidence of VTE in the peri-operative phase is currently unknown and likely underreported due to the lack of systematic screening methods and the limited validity of diagnostic tools (e.g. D-dimer, typical pain symptoms) in this setting. It is associated with high peri-operative mortality (∼17%).322 Risk factors for post-operative VTE/PE include type of surgery (e.g. high-risk hip arthroplasty, open prostatectomy, open surgery for malignancy), acute renal insufficiency, MI, and post-operative infection.322 Stratification of the extent of embolism (e.g. massive, submassive, and subsegmental; high risk, intermediate high/low risk, and low risk) is important to predict mortality and guide therapeutic strategy.652 Pulmonary embolism should be suspected in patients with PMI without a clear cause. Close haemodynamic monitoring and monitoring of RV function (echocardiography, CT) is essential to determine which PE patients require aggressive therapy. There is currently a lack of evidence regarding adequate antithrombotic therapy in patients with post-operative PE, since recent major surgery or trauma was a contraindication in previous trials of thrombolytic or anticoagulant therapy.653 Small case series support the use of systemic thrombolysis, surgical thrombectomy,654 or catheter-directed therapies in massive PE. In general, anticoagulation, preferably LMWH or fondaparinux, should be initiated as early as possible.652 Oral anticoalgulant therapy, preferably NOAC due to the lower bleeding risk, should be initiated, depending on post-operative renal function and bleeding risk, as early as possible for at least 3 months.652

Rescue thrombolytic therapy is recommended for patients with PE and haemodynamic deterioration on anticoagulation treatment in the post-operative phase, if possible, according to bleeding risk.652 As an alternative to thrombolytic therapy for massive PE, surgical embolectomy or percutaneous catheter-directed treatment should be considered for patients with haemodynamic deterioration on anticoagulation treatment, particularly in patients with high bleeding risk.

8.6. Atrial fibrillation and other relevant arrhythmias

Post-operative AF is defined as new-onset AF in the immediate post-operative period; its incidence ranges between 2–30%, with peak incidence 2–4 days post-operatively.655,656

Although many post-operative AF episodes are self–terminating and some are asymptomatic, post-operative AF has been associated with a four- to five-fold risk of AF recurrence in the 5 years following cardiac surgery, while the risk of recurrence after NCS is less well described.656–660 Importantly, post-operative AF is a risk factor for stroke, MI, and death compared with non-post-operative AF patients.656,658,661 Post-operative AF may also lead to haemodynamic instability, prolonged hospital stay, infections, renal complications, bleeding, increased in-hospital death, and greater healthcare costs.662–664 The essential principles of the prevention and management of post-operative AF are outlined in Figure 20.

Prevention and management of post-operative atrial fibrillation.
Figure 20

Prevention and management of post-operative atrial fibrillation.

AAD, antiarrhythmic drug; AF, atrial fibrillation; b.p.m., beats per minute; CCB, calcium channel blocker; HR, heart rate; LVEF, left ventricular ejection fraction. N, no; Y, yes. aDepending on the CHA2DS2VASC-score, and post-operative bleeding risk. bIn the acute post-operative phase, unless blood pressure is high, combination of low-dose beta-blocker and loading with digoxin is preferred to avoid hypotension. cShould include a cardiology visit before month 3. Adapted from the 2020 ESC Guidelines on the Diagnosis and Management of Atrial Fibrillation.99

Central illustration: the complex interplay between the intrinsic risk of surgery and the patient risk of peri-operative cardiovascular complications.
Figure 21

Central illustration: the complex interplay between the intrinsic risk of surgery and the patient risk of peri-operative cardiovascular complications.

8.6.1. Prevention of post-operative atrial fibrillation

Pre-operative use of beta-blockers is associated with reduced incidence of post-operative AF,204,665–667 but not major adverse events such as death, stroke, or AKI.668 Importantly, in a large RCT, peri-operative metoprolol was associated with increased mortality risk.185 In a meta-analysis, amiodarone (oral or i.v.) was equally effective in reducing post-operative AF as beta-blockers,207 whereas their combination was better than beta-blockers alone.208 Lower cumulative doses of amiodarone (<3000 mg) may be effective, with fewer adverse events.669–671 Data for other interventions—such as statins,672,673 magnesium,674 sotalol,666 colchicine,675 or corticosteroids676—are not robust.677,678

8.6.2. Management of post-operative atrial fibrillation

8.6.2.1. Rate and/or rhythm control

In haemodynamically unstable patients with post-operative AF, an emergent electrical or possibly pharmacological (i.e. i.v. administration of amiodarone666 or vernakalant,679 if consistent with the clinical situation) cardioversion is indicated.99 In haemodynamically stable patients with post-operative AF, ensuring optimal ventricular rate control during the arrhythmia is mandatory (using, for example beta-blockers or verapamil, as needed). As post-operative AF is often self-terminating, non–emergent cardioversion may not be needed. If performed in patients with AF lasting ≥48 h, non-emergency cardioversion of post-operative AF should follow the principles of pericardioversion thromboprophylaxis (that is, can be performed only after the left atrial thrombosis is excluded by TEE or postponed for 3 weeks of therapeutic OAC therapy). Of note, a RCT of patients with post-operative AF after cardiac surgery showed no net clinical advantage of rhythm (i.e. achieving and maintenance of sinus rhythm) vs. rate control strategy.680 Therefore, rate or rhythm control treatment decisions in patients with haemodynamically stable post-operative AF should be based on patient’s symptoms and shared informed treatment decision-making.99

8.6.2.2. Prevention of atrial fibrillation-related thromboembolic complications

In all patients with post-operative AF, it should be considered to initiate therapeutic anticoagulation as soon as possible during in-hospital treatment, depending on the individual stroke risk (CHA2DS2-VASc) and the bleeding risk after surgery. In a large meta-analysis, patients with post-operative AF had 62% higher risk of early stroke and 37% higher risk of long-term stroke compared with patients without post-operative AF (long-term stroke rates were 2.4% in post-operative AF vs. 0.4% in patients without AF), and a 44% and 37% higher risk of early and long-term mortality, respectively.661 Importantly, post-operative AF was more strongly associated with the long-term stroke risk in patients undergoing NCS (HR, 2.00; 95% CI, 1.70–2.35) than in patients undergoing cardiac surgery (HR, 1.20; 95% CI, 1.07–1.34; P < 0.0001).661

Evidence on the effects of long-term OAC therapy for the prevention of stroke or systemic embolism in patients with post-operative AF is from observational studies.664,681–685 In a recent study, post-operative AF following NCS was associated with similar long-term thromboembolic risk as common, non-surgical AF, and OAC use was associated with comparable lower risk of thromboembolism and all–cause mortality in both groups (mean CHA2DS2-VASc-score 3.0 ± 1.7).686

Based on the available evidence, long-term OAC should be considered in all patients with post-operative AF at risk of stroke. If anticoagulation is parenterally initiated, LMWH or fondaparinux is recommended (over UFH) for most patients. Non-vitamin K antagonist oral anticoagulants should be preferred over VKA for long-term treatment. Re-evaluation of the continuation of OAC may be performed after a period of 3 months. A small RCT (ASPIRE-AF; NCT03968393) on the optimal long-term OAC use among NCS patients developing post-operative AF is ongoing.

8.7. Peri-operative stroke

With respect to NCS, peri-operative stroke has been reported in 0.08–0.70% of patients undergoing general surgery, 0.2–0.9% of patients requiring orthopaedic surgery, 0.6–0.9% of lung operations, and 0.8–3.0% of surgeries involving the peripheral vasculature.687,688 The associated mortality ranges from 18–26%.687,688 A more recent analysis of 523 059 patients undergoing NCS reported a lower incidence of peri-operative stroke (0.1%), although the occurrence of this adverse event was associated with an eight-fold increase in peri-operative mortality within 30 days, corresponding to an absolute risk increase of >20%.689 Peri-operative stroke is mainly ischaemic or cardioembolic, and AF is often the leading underlying condition. Triggers include withdrawal of anticoagulation and the hypercoagulable state related to surgery. Additional aetiologies include atheroembolism, originating from the aorta or the supra-aortic vessels, and local atherothrombosis in the presence of intracranial small vessel disease. Hypoperfusion—related to peri-operative arterial hypotension and/or severe stenosis of the cervicocranial vessels—is an unusual cause of peri-operative stroke.690 Rarely, peri-operative stroke may be due to air, fat, or paradoxical embolisms.

In an attempt to attenuate the risk of peri-operative stroke, antiplatelet/anticoagulant treatments should be continued whenever possible throughout the peri-operative period. Alternatively, the period of drug withdrawal should be kept as short as possible, while weighting thromboembolic and haemorrhagic risks (see Section 5.2). Adequate selection of the anaesthetic technique (regional vs. neuraxial vs. general anaesthesia), prevention and treatment of AF, euglycaemic control (avoiding both hyperglycaemia and hypoglycaemia), and meticulous peri-operative control of BP may all contribute to reducing the risk of peri-operative stroke.

If post-operative stroke occurs, it must trigger immediate action: angio-CT and neurology/neurosurgical consultation with the goal to restore flow in the case of acute thrombotic occlusion.

Recommendation Table 34

Recommendations for peri-operative cardiovascular complications

graphic
graphic
graphic
graphic
Recommendation Table 34

Recommendations for peri-operative cardiovascular complications

graphic
graphic
graphic
graphic

9. Key messages

  • The occurrence of CV complications in the peri-operative phase of NCS has a dramatic impact on prognosis.

  • The risk of CV complications in patients undergoing NCS is determined by patient-related factors, type of surgery or procedure, and the circumstances under which surgery takes place (elective vs. emergency procedure; local or tertiary hospital).

  • Specific patient-related risk factors may be reduced by adequate pre-operative risk assessment and initiation of effective risk-reduction strategies.

  • The quantification of surgical risk as low, intermediate, and high is helpful in identifying the group of patients who should most benefit from preventive, diagnostic, and therapeutic approaches to concomitant CV conditions.

  • Proper selection of type and timing of the surgical procedure may reduce the risk of complications.

  • It is important that patients’ values, quality of life, and preferences regarding the benefits and risks of surgery are taken into consideration, and that well-informed patients are involved in the decisions. Risk should be communicated to the patient in absolute terms (e.g. 1 out of 100).

  • Clinical examination, patient-reported functional capacity, and non-invasive tests represent the cornerstone of pre-operative cardiac assessment.

  • Instrumental and functional cardiac examination tools should be selected in view of the surgical risk, relative diagnostic proficiency, and healthcare resource utilization and costs.

  • The peri-operative evaluation of elderly patients who require elective major NCS should include frailty screening, which has proven to be an excellent predictor of unfavourable health outcomes in the older surgical population.

  • Treatment of pre-existing or newly diagnosed CV conditions (e.g. coronary and peripheral vascular disease, rhythm disorders, and HF) should be individualized according to the pre-operative risk of NCS, and considering the recommendations of speciality guidelines.

  • A multidisciplinary approach to evaluate whether the treatment of concomitant cardiac conditions before scheduled NCS improves peri-operative safety without unnecessary delay is encouraged.

  • Efficient peri-operative management of antithrombotic therapies in patients scheduled for NCS aims to offer the potential benefit of preventing thrombotic events without excessive bleeding complications.

  • It is important to clearly and concisely communicate with patients, with simple verbal and written instructions, about changes in medication in the pre- and post-operative phases.

  • Management in the peri-operative phase of NCS aims to avoid haemodynamic imbalance, while ensuring sufficient cardioprotective action.

  • Healthcare providers are recommended to have high awareness of peri-operative CV complications combined with surveillance for PMI in high-risk patients undergoing intermediate- or high-risk NCS.

  • Routine assessment of treatment quality through specific indicators is important to document and measure the success of preventive and therapeutic strategies in patients undergoing NCS.

10. Gaps in evidence

  • The age cut-off for individuals (considered to be cardiovascularly healthy) benefiting from risk stratification work-out before NCS needs to be evaluated.

  • Further studies are needed to characterize outcome differences in NCS between men and women, and between different countries, in order to individualize peri-operative management and improve patient safety.

  • Evidence on the additive value of cardiac biomarkers, hand-held ultrasound, problem FOCUS, and stress echocardiography for cardiac risk stratification of patients scheduled for NCS presenting with previously unknown cardiac murmur, dyspnoea, oedema, and chest pain is still lacking. The impact of FOCUS on outcomes of urgent or time-sensitive surgery needs further investigation.

  • The impact of stress imaging (echocardiography or MRI) before NCS on reduction of peri-operative CV complications in non-ischaemic heart diseases needs further research.

  • The role of right heart catheterization in patients with advanced HF or patients with severe pulmonary hypertension undergoing NCS is unknown.

  • It is unknown whether artificial intelligence-based systems facilitate prompt detection and response to imminent adverse events in high-risk cardiac patients undergoing high-risk NCS.

  • Systematic and structured research to investigate pathophysiology, causes, and time distribution of serious peri-operative arrhythmic events among patients undergoing NCS is still needed.

  • Strategies for timing of pre-operative CIED control dependent on device type, urgency, and type of NCS, and risk of EMI during NCS need to be developed to ensure maximal patient safety.

  • Benefit of routine myocardial revascularization of high-risk CCS patients (except left main or three-vessel CAD, reduced LV function) before elective intermediate- and high-risk NCS is not well-established.

  • More evidence regarding the need for bridging of anticoagulation in patients with MHVs is needed.

  • There is a lack of evidence regarding the optimal strategies before emergent or time-sensitive NCS for patients on antithrombotic treatment at high risk of thromboembolic events, including the: (i) use of extracorporeal haemoperfusion or NOAC antidotes (ongoing trial NCT04233073); (ii) use of albumin, extracorporeal haemoperfusion, or PB2452-specific antidote to antagonized ticagrelor (ongoing trial NCT04286438 for PB2452); and (iii) premature cessation or bridging during interruption of oral P2Y12-receptor inhibitors (glycoprotein IIb/IIIa receptor inhibitors or cangrelor).

  • There is lack of well-powered studies to evaluate the role of platelet function testing to guide the strategy for treatment of NCS patients on antiplatelet therapy.

  • Evidence regarding the need for and benefit of anticoagulation in NCS patients with post-operative AF is still lacking (ongoing ASPIRE-AF trial: NCT03968393).

  • Prophylactic strategies to reduce the incidence of post-operative AF in NCS patients additional to beta-blocker maintenance in patients already on this treatment need to be evaluated.

  • The optimal cardiac work-up and therapy for patients with PMI within and outside hospital settings need to be evaluated.

  • Studies are needed to investigate the impact of the treatment of peri-operative hypotension on post-operative outcomes, the use of new HF drug classes (SGLT2 inhibitors and vericiguat), and the use of NSAIDs as a temporary treatment for acute post-operative pain.

  • Prospective studies are needed to investigate the incremental value of anaemia algorithms and blood-sparing strategies (use of blood-sparing blood tubes) to reduce the risk of anaemia-associated adverse outcomes among CV patients undergoing NCS.

11. Sex differences

Sex and gender may significantly affect the management and outcomes of patients with specific diseases undergoing NCS. There are sex- and gender-dependent clinical phenotypes of comorbidities and risk factors, which may have an impact on peri-operative morbidity and mortality. However, there is a paucity of data specifically addressing the interplay between sex, age, and comorbidities in patients scheduled for NCS.

The pre-operative assessment before NCS might take sex into consideration, since the age-adjusted incidence of CVD is lower in women than men, and the risk of undetected disease could therefore be lower in women. However, no data exist on sex-specific assessment strategies.

The in-hospital mortality during surgery was recently reported to be lower in women than men.41,691 In contrast, among 609 735 patients who underwent elective NCS between 2009 and 2016, the odds of post-operative 90 day mortality were higher among women with HF than men with HF.692 Further studies are needed to provide more information about outcome differences between men and women in NCS.

Some studies have reported a higher risk of bleeding in women than men, but other studies could not confirm this. No trials have systematically investigated the impact of sex differences regarding efficacy and safety of continuation vs. interruption of antithrombotic therapy in patients undergoing NCS.

The prevalence of anaemia in women of reproductive age is as high as 30% (WHO Global Anaemia estimates),693 resulting in millions of women undergoing surgery every year despite pre-operative anaemia. Furthermore, since women have lower blood volumes and lower haemoglobin values than men, but face the same surgical blood loss as men, they are exposed to far higher risk of post-operative complications. Also, higher transfusion rates and volumes have been reported in women compared with men in elective surgery.694 It is therefore of particular importance that clinicians follow the Patient Blood Management program in women undergoing NCS. Other sex-related differences in physiology, and pharmacokinetics and pharmacodynamics of anaesthetic drugs may influence the anaesthesia plan, pain management, post-operative recovery, and patient satisfaction.

Sex differences regarding presentation, electrophysiological substrate, complications, or long-term outcomes have been reported in patients undergoing CIED implantation,695,696 and female sex is a well-known risk factor for stroke in patients with AF.697 However, no specific data exist that suggest sex differences in risk profile or outcomes of patients with CIEDs or arrhythmias who undergo NCS.

12. ‘What to do’ and ‘what not to do’ messages from the Guidelines

13. Quality indicators

Quality indicators (QIs) are tools that may be used to evaluate care quality, including structural, process, and outcomes of care.698 They may also serve as a mechanism for enhancing adherence to guideline recommendations, through associated quality improvement initiatives and the benchmarking of care providers.699,700 As such, the role of QIs in improving care and outcomes for CV disease is increasingly being recognized by healthcare authorities, professional organizations, payers, and the public.698

The ESC understands the need for measuring and reporting quality and outcomes of CV care and has established methods for the development of the ESC QIs for the quantification of care and outcomes for CV diseases.698 To date, the ESC has developed QI suites for a number of CV diseases701–703 and embedded these in respective ESC Clinical Practice Guidelines (2020 ESC Guidelines for the diagnosis and management of atrial fibrillation; 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure; and 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy).99,412,481,704 Furthermore, the ESC aims to integrate its QIs with clinical registries such as the EURObservational Research Programme (EORP) and the European Unified Registries On Heart Care Evaluation and Randomized Trials (EuroHeart) project705 to provide ‘real-world’ data about the patterns and outcomes of care for CV disease across Europe.

In parallel with the writing of this Clinical Practice Guideline document, a process has been initiated to develop QIs for patients undergoing NCS using the ESC methodology and through collaboration with patient representatives and domain experts. Such QIs may be used for evaluation of the quality of care for this group of patients and enable capture of important aspects of care delivery. The QIs, alongside their measurement specifications and development process, will be published in a separate paper.

14. Central illustration

There is a complex interplay between the intrinsic risk of surgery and the patient-related risk of peri-operative CV complications. This latter risk depends on the baseline general and CV status of patients scheduled for NCS. For each patient, the proper quantification and communication of the surgical risk require close cooperation between cardiologists, surgeons, anaesthesiologists, general practitioners, and other healthcare providers (Figure 21).

15. Supplementary data

Supplementary data is available at European Heart Journal online.

16. Data availability statement

No new data were generated or analysed in support of this research.

17. Author information

Author/Task Force Member Affiliations: Salvatore Cassese, Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Munich, Germany; Trygve S. Hall, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway; Magdy Abdelhamid, Cardiology Department, Faculty of Medicine, Kase Al Ainy, Cairo University, Cairo, Egypt; Emanuele Barbato, Advanced Biomedical Sciences, University Federico II, Naples, Italy, Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium; Stefan De Hert, Department of Anesthesiology and Perioperative Medicine, Ghent University Hospital/Ghent University, Ghent, Belgium; Tobias Geisler, Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany; Lynne Hinterbuchner, Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria; Borja Ibanez, Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain, Cardiology Department, Fundación Jiménez Díaz Hospital, Madrid, Spain, CIBERCV Centro Investigacion Biomedica en Red, Madrid, Spain; Ingrid de Laval (Sweden), ESC Patient Forum, Sophia Antipolis, France; Radosław Lenarczyk, Department of Cardiology, Congenital Heart Diseases and Electrotherapy, The Medical University of Silesia, Division of Medical Sciences in Zabrze, Zabrze, Poland, Silesian Center for Heart Disease, Zabrze, Poland; Ulrich R. Mansmann, Institute for Medical Data Processing, Biometry, and Epidemiology, Ludwig-Maximilian's University, Munich, Germany; Paul McGreavy (United Kingdom), ESC Patient Forum, Sophia Antipolis, France; Christian Mueller, Cardiovascular Research Institute Basel, University Heart Center, University Hospital Basel, Basel, Switzerland, University of Basel, Basel, Switzerland; Claudio Muneretto, Cardiothoracic, University of Brescia Italy, Brescia, Italy; Alexander Niessner, Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria; Tatjana S. Potpara, School of Medicine, Belgrade University, Belgrade, Serbia, Cardiology Clinic, University Clinical Center of Serbia, Belgrade, Serbia; Arsen D. Ristić, Faculty of Medicine, Belgrade University, Belgrade, Serbia, Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia; L. Elif Sade, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America, Cardiology, University of Baskent, Ankara, Turkey; Henrik Schirmer, Department of Cardiology, Akershus University Hospital, Lørenskog, Norway, Institute of Clinical Medicine, Campus Ahus, University of Oslo, Lørenskog, Norway; Stefanie Schüpke, ISAResearch Center, Deutsches Herzzentrum München, Munich, Germany, Heart Alliance, DZHK (German Center for Cardiovascular Research), Munich, Germany; Henrik Sillesen, Vascular Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark, Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Helge Skulstad, Department of Cardiology, Clinic of heart-, lung- and vessel disease, Oslo University Hospital, Oslo, Norway, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Lucia Torracca, Cardiac Surgery, IRCCS Humanitas Research Hospital, Milano, Italy; Oktay Tutarel, Department of Congenital Heart Disease and Paediatric Cardiology, German Heart Centre Munich, TUM School of Medicine, Technical University of Munich, Munich, Germany, DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Peter Van der Meer, Cardiology, University Medical Center Groningen, Groningen, Netherlands; Wojtek Wojakowski, Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland; and Kai Zacharowski, Anaesthesiology, Intensive Care Medicine & Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.

18. Appendix

ESC Scientific Document Group: includes Document Reviewers and ESC National Cardiac Societies.

Document Reviewers: Juhani Knuuti (CPG Review Coordinator) (Finland), Steen Dalby Kristensen (CPG Review Coordinator) (Denmark), Victor Aboyans (France), Ingo Ahrens (Germany), Sotiris Antoniou (United Kingdom), Riccardo Asteggiano (Italy), Dan Atar (Norway), Andreas Baumbach (United Kingdom), Helmut Baumgartner (Germany), Michael Böhm (Germany), Michael A. Borger (Germany), Hector Bueno (Spain), Jelena Čelutkienė (Lithuania), Alaide Chieffo (Italy), Maya Cikes (Croatia), Harald Darius (Germany), Victoria Delgado (Spain), Philip J. Devereaux (Canada), David Duncker (Germany), Volkmar Falk (Germany), Laurent Fauchier (France), Gilbert Habib (France), David Hasdai (Israel), Kurt Huber (Austria), Bernard Iung (France), Tiny Jaarsma (Sweden), Aleksandra Konradi (Russian Federation), Konstantinos C. Koskinas (Switzerland), Dipak Kotecha (United Kingdom), Ulf Landmesser (Germany), Basil S. Lewis (Israel), Ales Linhart (Czech Republic), Maja-Lisa Løchen (Norway), Michael Maeng (Denmark), Stéphane Manzo-Silberman (France), Richard Mindham (United Kingdom), Lis Neubeck (United Kingdom), Jens Cosedis Nielsen (Denmark), Steffen E. Petersen (United Kingdom), Eva Prescott (Denmark), Amina Rakisheva (Kazakhstan), Antti Saraste (Finland), Dirk Sibbing (Germany), Jolanta M. Siller-Matula (Austria), Marta Sitges (Spain), Ivan Stankovic (Serbia), Rob F. Storey (United Kingdom), Jurrien ten Berg (Netherlands), Matthias Thielmann (Germany), and Rhian M. Touyz (Canada/United Kingdom).

ESC National Cardiac Societies actively involved in the review process of the 2022 ESC Guidelines on assessment and management of patients undergoing non-cardiac surgery

Algeria: Algerian Society of Cardiology, Mohammed Amine Bouzid; Armenia: Armenian Cardiologists Association, Hamayak Sisakian; Austria: Austrian Society of Cardiology, Bernhard Metzler; Belarus: Belorussian Scientific Society of Cardiologists, Vadim Shumavets; Belgium: Belgian Society of Cardiology, Agnès Pasquet; Bosnia and Herzegovina: Association of Cardiologists of Bosnia and Herzegovina, Elnur Smajic; Bulgaria: Bulgarian Society of Cardiology, Maria Milanova; Croatia: Croatian Cardiac Society, Boško Skorić; Cyprus: Cyprus Society of Cardiology, Maria Karakyriou; Czechia: Czech Society of Cardiology, Hana Skalicka; Denmark: Danish Society of Cardiology, Michael Maeng; Egypt: Egyptian Society of Cardiology, Bassem Abd Elhamid; Estonia: Estonian Society of Cardiology, Arno Ruusalepp; Finland: Finnish Cardiac Society, Kati Valtola; France: French Society of Cardiology, Ariel Cohen; Georgia: Georgian Society of Cardiology, Archil Chukhrukidze; Germany: German Cardiac Society, Ilka Ott; Greece: Hellenic Society of Cardiology, Nikos Kafkas; Hungary: Hungarian Society of Cardiology, Zoltán Járai; Iceland: Icelandic Society of Cardiology, Thórdís Jóna Hrafnkelsdóttir; Ireland: Irish Cardiac Society, Patricia Campbell; Israel: Israel Heart Society, Alon Eisen; Italy: Italian Federation of Cardiology, Stefano Urbinati; Kazakhstan: Association of Cardiologists of Kazakhstan, Nazipa Aidargaliyeva; Kosovo (Republic of): Kosovo Society of Cardiology, Arlind Batalli; Kyrgyzstan: Kyrgyz Society of Cardiology, Olga Lunegova; Latvia: Latvian Society of Cardiology, Andrejs Erglis; Lebanon: Lebanese Society of Cardiology, Georges Saade; Lithuania: Lithuanian Society of Cardiology, Andrius Macas; Luxembourg: Luxembourg Society of Cardiology, Cristiana Banu; Malta: Maltese Cardiac Society, Tiziana Felice; Moldova: Moldavian Society of Cardiology, Aurel Grosu; Montenegro: Montenegro Society of Cardiology, Mihailo Vukmirovic; Morocco: Moroccan Society of Cardiology, Aida Soufiani; Netherlands: Netherlands Society of Cardiology, Eric Dubois; North Macedonia: North Macedonian Society of Cardiology, Hristo Pejkov; Norway: Norwegian Society of Cardiology, Erlend Aune; Poland: Polish Cardiac Society, Stanisław Bartuś; Portugal: Portuguese Society of Cardiology, Mário Santos; Romania: Romanian Society of Cardiology, Elisabeta Badila; Russian Federation: Russian Society of Cardiology, Olga Irtyuga; San Marino: San Marino Society of Cardiology, Luca Bertelli; Serbia: Cardiology Society of Serbia, Branko Beleslin; Slovakia: Slovak Society of Cardiology, Martin Dúbrava; Slovenia: Slovenian Society of Cardiology, Zlatko Fras; Spain: Spanish Society of Cardiology, José Luis Ferreiro; Sweden: Swedish Society of Cardiology, Claes Held; Switzerland: Swiss Society of Cardiology, Philippe Meyer; Syrian Arab Republic: Syrian Cardiovascular Association, Walid Bsata; Tunisia: Tunisian Society of Cardiology and Cardio-Vascular Surgery, Afef Ben Halima; Turkey: Turkish Society of Cardiology, Murat Biteker, United Kingdom of Great Britain and Northern Ireland: British Cardiovascular Society, Andrew Archbold; Ukraine: Ukrainian Association of Cardiology, Maksym Sokolov; and Uzbekistan: Association of Cardiologists of Uzbekistan, Nodir Zakirov.

ESC Clinical Practice Guidelines (CPG) Committee: Colin Baigent (Chairperson) (United Kingdom), Magdy Abdelhamid (Egypt), Victor Aboyans (France), Sotiris Antoniou (United Kingdom), Elena Arbelo (Spain), Riccardo Asteggiano (Italy), Andreas Baumbach (United Kingdom), Michael A. Borger (Germany), Jelena Čelutkienė (Lithuania), Maja Cikes (Croatia), Jean-Philippe Collet (France), Volkmar Falk (Germany), Laurent Fauchier (France), Chris P. Gale (United Kingdom), Sigrun Halvorsen (Norway), Bernard Iung (France), Tiny Jaarsma (Sweden), Aleksandra Konradi (Russian Federation), Konstantinos C. Koskinas (Switzerland), Dipak Kotecha (United Kingdom), Ulf Landmesser (Germany), Basil S. Lewis (Israel), Ales Linhart (Czech Republic), Maja-Lisa Løchen (Norway), Richard Mindham (United Kingdom), Jens Cosedis Nielsen (Denmark), Steffen E. Petersen (United Kingdom), Eva Prescott (Denmark), Amina Rakisheva (Kazakhstan), Marta Sitges (Spain), and Rhian M. Touyz (Canada /United Kingdom).

References

1

Weiser
 
TG
,
Regenbogen
 
SE
,
Thompson
 
KD
,
Haynes
 
AB
,
Lipsitz
 
SR
,
Berry
 
WR
, et al.  
An estimation of the global volume of surgery: a modelling strategy based on available data
.
Lancet
 
2008
;
372
:
139
144
.

2

Weiser
 
TG
,
Haynes
 
AB
,
Molina
 
G
,
Lipsitz
 
SR
,
Esquivel
 
MM
,
Uribe-Leitz
 
T
, et al.  
Estimate of the global volume of surgery in 2012: an assessment supporting improved health outcomes
.
Lancet
 
2015
;
385
:
S11
.

4

Smilowitz
 
NR
,
Gupta
 
N
,
Guo
 
Y
,
Beckman
 
JA
,
Bangalore
 
S
,
Berger
 
JS
.
Trends in cardiovascular risk factor and disease prevalence in patients undergoing non-cardiac surgery
.
Heart
 
2018
;
104
:
1180
1186
.

5

Lee
 
OH
,
Hong
 
SJ
,
Ahn
 
CM
,
Kim
 
JS
,
Kim
 
BK
,
Ko
 
YG
, et al.  
The incidence of non-cardiac surgery in patients treated with drug-eluting stents according to age
.
J Invasive Cardiol
 
2019
;
31
:
E9
E14
.

6

Vascular Events in Noncardiac Surgery Patients Cohort Evaluation Study Investigators
,
Spence
 
J
,
LeManach
 
Y
,
Chan
 
MTV
,
Wang
 
CY
,
Sigamani
 
A
, et al.  
Association between complications and death within 30 days after noncardiac surgery
.
CMAJ
 
2019
;
191
:
E830
E837
.

7

Pearse
 
RM
,
Moreno
 
RP
,
Bauer
 
P
,
Pelosi
 
P
,
Metnitz
 
P
,
Spies
 
C
, et al.  
Mortality after surgery in Europe: a 7 day cohort study
.
Lancet
 
2012
;
380
:
1059
1065
.

8

Sazgary
 
L
,
Puelacher
 
C
,
Lurati Buse
 
G
,
Glarner
 
N
,
Lampart
 
A
,
Bolliger
 
D
, et al.  
Incidence of major adverse cardiac events following non-cardiac surgery
.
Eur Heart J Acute Cardiovasc Care
 
2020
;
10
:
550
558
.

9

Smilowitz
 
NR
,
Berger
 
JS
.
Perioperative cardiovascular risk assessment and management for noncardiac surgery: a review
.
JAMA
 
2020
;
324
:
279
290
.

10

Kristensen
 
SD
,
Knuuti
 
J
,
Saraste
 
A
,
Anker
 
S
,
Botker
 
HE
,
Hert
 
SD
, et al.  
2014 ESC/ESA Guidelines on non-cardiac surgery: cardiovascular assessment and management: the Joint Task Force on non-cardiac surgery: cardiovascular assessment and management of the European Society of Cardiology (ESC) and the European Society of Anaesthesiology (ESA)
.
Eur Heart J
 
2014
;
35
:
2383
2431
.

11

Kamel
 
H
,
Johnston
 
SC
,
Kirkham
 
JC
,
Turner
 
CG
,
Kizer
 
JR
,
Devereux
 
RB
, et al.  
Association between major perioperative hemorrhage and stroke or Q-wave myocardial infarction
.
Circulation
 
2012
;
126
:
207
212
.

12

CRASH-2 Trial Collaborators
,
Shakur
 
H
,
Roberts
 
I
,
Bautista
 
R
,
Caballero
 
J
,
Coats
 
T
, et al.  
Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomised, placebo-controlled trial
.
Lancet
 
2010
;
376
:
23
32
.

13

Kougias
 
P
,
Sharath
 
S
,
Barshes
 
NR
,
Chen
 
M
,
Mills
 
JL
, Sr
.
Effect of postoperative anemia and baseline cardiac risk on serious adverse outcomes after major vascular interventions
.
J Vasc Surg
 
2017
;
66
:
1836
1843
.

14

Cohn
 
SL
.
The cardiac consult for patients undergoing non-cardiac surgery
.
Heart
 
2016
;
102
:
1322
1332
.

15

Guay
 
J
,
Choi
 
P
,
Suresh
 
S
,
Albert
 
N
,
Kopp
 
S
,
Pace
 
NL
.
Neuraxial blockade for the prevention of postoperative mortality and major morbidity: an overview of Cochrane systematic reviews
.
Cochrane Database Syst Rev
 
2014
:
2014
:
CD010108
.

16

Norderud
 
K
,
Egholm
 
G
,
Thim
 
T
,
Olesen
 
KKW
,
Madsen
 
M
,
Jensen
 
LO
, et al.  
Validation of the European Society of Cardiology and European Society of Anaesthesiology non-cardiac surgery risk score in patients treated with coronary drug-eluting stent implantation
.
Eur Heart J Qual Care Clin Outcomes
 
2019
;
5
:
22
27
.

17

Mangano
 
DT
.
Perioperative medicine: NHLBI working group deliberations and recommendations
.
J Cardiothorac Vasc Anesth
 
2004
;
18
:
1
6
.

18

Glance
 
LG
,
Lustik
 
SJ
,
Hannan
 
EL
,
Osler
 
TM
,
Mukamel
 
DB
,
Qian
 
F
, et al.  
The Surgical Mortality Probability Model: derivation and validation of a simple risk prediction rule for noncardiac surgery
.
Ann Surg
 
2012
;
255
:
696
702
.

19

Muller
 
MD
,
Lyrer
 
P
,
Brown
 
MM
,
Bonati
 
LH
.
Carotid artery stenting versus endarterectomy for treatment of carotid artery stenosis
.
Cochrane Database Syst Rev
 
2020
;
2
:
CD000515
.

20

Bendixen
 
M
,
Jørgensen
 
OD
,
Kronborg
 
C
,
Andersen
 
C
,
Licht
 
PB
.
Postoperative pain and quality of life after lobectomy via video-assisted thoracoscopic surgery or anterolateral thoracotomy for early stage lung cancer: a randomised controlled trial
.
Lancet Oncol
 
2016
;
17
:
836
844
.

21

Falcoz
 
PE
,
Puyraveau
 
M
,
Thomas
 
PA
,
Decaluwe
 
H
,
Hürtgen
 
M
,
Petersen
 
RH
, et al.  
Video-assisted thoracoscopic surgery versus open lobectomy for primary non-small-cell lung cancer: a propensity-matched analysis of outcome from the European Society of Thoracic Surgeon database
.
Eur J Cardiothorac Surg
 
2016
;
49
:
602
609
.

22

Martin
 
D
,
Mantziari
 
S
,
Demartines
 
N
,
Hubner
 
M
,
Study Group
 
ESA
.
Defining major surgery: a Delphi consensus among European Surgical Association (ESA) members
.
World J Surg
 
2020
;
44
:
2211
2219
.

23

Bolliger
 
M
,
Kroehnert
 
JA
,
Molineus
 
F
,
Kandioler
 
D
,
Schindl
 
M
,
Riss
 
P
.
Experiences with the standardized classification of surgical complications (Clavien-Dindo) in general surgery patients
.
Eur Surg
 
2018
;
50
:
256
261
.

24

Bjorck
 
M
,
Earnshaw
 
JJ
,
Acosta
 
S
,
Bastos Goncalves
 
F
,
Cochennec
 
F
,
Debus
 
ES
, et al.  
Editor’s choice – European Society for Vascular Surgery (ESVS) 2020 Clinical Practice Guidelines on the management of acute limb ischaemia
.
Eur J Vasc Endovasc Surg
 
2020
;
59
:
173
218
.

25

Holte
 
K
,
Kehlet
 
H
.
Postoperative ileus: a preventable event
.
Br J Surg
 
2000
;
87
:
1480
1493
.

26

Popescu
 
WM
,
Bell
 
R
,
Duffy
 
AJ
,
Katz
 
KH
,
Perrino
 
AC
.
A pilot study of patients with clinically severe obesity undergoing laparoscopic surgery: evidence for impaired cardiac performance
.
J Cardiothorac Vasc Anesth
 
2011
;
25
:
943
949
.

27

Lestar
 
M
,
Gunnarsson
 
L
,
Lagerstrand
 
L
,
Wiklund
 
P
,
Odeberg-Wernerman
 
S
.
Hemodynamic perturbations during robot-assisted laparoscopic radical prostatectomy in 45° Trendelenburg position
.
Anesth Analg
 
2011
;
113
:
1069
1075
.

28

Hirvonen
 
EA
,
Nuutinen
 
LS
,
Kauko
 
M
.
Hemodynamic changes due to Trendelenburg positioning and pneumoperitoneum during laparoscopic hysterectomy
.
Acta Anaesthesiol Scand
 
1995
;
39
:
949
955
.

29

Nguyen
 
NT
,
Wolfe
 
BM
.
The physiologic effects of pneumoperitoneum in the morbidly obese
.
Ann Surg
 
2005
;
241
:
219
226
.

30

Keus
 
F
,
Gooszen
 
HG
,
van Laarhoven
 
CJ
.
Open, small-incision, or laparoscopic cholecystectomy for patients with symptomatic cholecystolithiasis. An overview of Cochrane Hepato-Biliary Group reviews
.
Cochrane Database Syst Rev
 
2010
;
2010
:
CD008318
.

31

Mamidanna
 
R
,
Burns
 
EM
,
Bottle
 
A
,
Aylin
 
P
,
Stonell
 
C
,
Hanna
 
GB
, et al.  
Reduced risk of medical morbidity and mortality in patients selected for laparoscopic colorectal resection in England: a population-based study
.
Arch Surg
 
2012
;
147
:
219
227
.

32

Cirocchi
 
R
,
Farinella
 
E
,
Trastulli
 
S
,
Sciannameo
 
F
,
Audisio
 
RA
.
Elective sigmoid colectomy for diverticular disease. Laparoscopic vs open surgery: a systematic review
.
Colorectal Dis
 
2012
;
14
:
671
683
.

33

Murr
 
MM
,
Martin
 
T
,
Haines
 
K
,
Torrella
 
T
,
Dragotti
 
R
,
Kandil
 
A
, et al.  
A state-wide review of contemporary outcomes of gastric bypass in Florida: does provider volume impact outcomes?
 
Ann Surg
 
2007
;
245
:
699
706
.

34

Grailey
 
K
,
Markar
 
SR
,
Karthikesalingam
 
A
,
Aboud
 
R
,
Ziprin
 
P
,
Faiz
 
O
.
Laparoscopic versus open colorectal resection in the elderly population
.
Surg Endosc
 
2013
;
27
:
19
30
.

35

Wanhainen
 
A
,
Verzini
 
F
,
Van Herzeele
 
I
,
Allaire
 
E
,
Bown
 
M
,
Cohnert
 
T
, et al.  
Editor’s choice – European Society for Vascular Surgery (ESVS) 2019 Clinical Practice Guidelines on the management of abdominal aorto-iliac artery aneurysms
.
Eur J Vasc Endovasc Surg
 
2019
;
57
:
8
93
.

36

Powell
 
JT
,
Sweeting
 
MJ
,
Ulug
 
P
,
Blankensteijn
 
JD
,
Lederle
 
FA
,
Becquemin
 
JP
, et al.  
Meta-analysis of individual-patient data from EVAR-1, DREAM, OVER and ACE trials comparing outcomes of endovascular or open repair for abdominal aortic aneurysm over 5 years
.
Br J Surg
 
2017
;
104
:
166
178
.

37

Lederle
 
FA
,
Kyriakides
 
TC
,
Stroupe
 
KT
,
Freischlag
 
JA
,
Padberg
 
FT
, Jr.
,
Matsumura
 
JS
, et al.  
Open versus endovascular repair of abdominal aortic aneurysm
.
N Engl J Med
 
2019
;
380
:
2126
2135
.

38

Bauer
 
SM
,
Cayne
 
NS
,
Veith
 
FJ
.
New developments in the preoperative evaluation and perioperative management of coronary artery disease in patients undergoing vascular surgery
.
J Vasc Surg
 
2010
;
51
:
242
251
.

39

Antoniou
 
GA
,
Chalmers
 
N
,
Georgiadis
 
GS
,
Lazarides
 
MK
,
Antoniou
 
SA
,
Serracino-Inglott
 
F
, et al.  
A meta-analysis of endovascular versus surgical reconstruction of femoropopliteal arterial disease
.
J Vasc Surg
 
2013
;
57
:
242
253
.

40

Visseren
 
FLJ
,
Mach
 
F
,
Smulders
 
YM
,
Carballo
 
D
,
Koskinas
 
KC
,
Back
 
M
, et al.  
2021 ESC Guidelines on cardiovascular disease prevention in clinical practice
.
Eur Heart J
 
2021
;
42
:
3227
3337
.

41

Botto
 
F
,
Alonso-Coello
 
P
,
Chan
 
MT
,
Villar
 
JC
,
Xavier
 
D
,
Srinathan
 
S
, et al.  
Myocardial injury after noncardiac surgery: a large, international, prospective cohort study establishing diagnostic criteria, characteristics, predictors, and 30-day outcomes
.
Anesthesiology
 
2014
;
120
:
564
578
.

42

Noordzij
 
PG
,
Boersma
 
E
,
Bax
 
JJ
,
Feringa
 
HH
,
Schreiner
 
F
,
Schouten
 
O
, et al.  
Prognostic value of routine preoperative electrocardiography in patients undergoing noncardiac surgery
.
Am J Cardiol
 
2006
;
97
:
1103
1106
.

43

Chivulescu
 
M
,
Lie
 
Ø
,
Popescu
 
BA
,
Skulstad
 
H
,
Edvardsen
 
T
,
Jurcut
 
RO
, et al.  
High penetrance and similar disease progression in probands and in family members with arrhythmogenic cardiomyopathy
.
Eur Heart J
 
2020
;
41
:
1401
1410
.

44

Hallqvist
 
L
,
Granath
 
F
,
Bell
 
M
.
Myocardial infarction after noncardiac surgery in Sweden: a national, retrospective observational cohort study
.
Br J Anaesth
 
2020
;
125
:
47
54
.

45

Chaudhry
 
W
,
Cohen
 
MC
.
Cardiac screening in the noncardiac surgery patient
.
Surg Clin North Am
 
2017
;
97
:
717
732
.

46

Ford
 
MK
,
Beattie
 
WS
,
Wijeysundera
 
DN
.
Systematic review: prediction of perioperative cardiac complications and mortality by the Revised Cardiac Risk Index
.
Ann Intern Med
 
2010
;
152
:
26
35
.

47

Duceppe
 
E
,
Parlow
 
J
,
MacDonald
 
P
,
Lyons
 
K
,
McMullen
 
M
,
Srinathan
 
S
, et al.  
Canadian Cardiovascular Society guidelines on perioperative cardiac risk assessment and management for patients who undergo noncardiac surgery
.
Can J Cardiol
 
2017
;
33
:
17
32
.

48

Yap
 
MKC
,
Ang
 
KF
,
Gonzales-Porciuncula
 
LA
,
Esposo
 
E
.
Validation of the American College of Surgeons Risk Calculator for preoperative risk stratification
.
Heart Asia
 
2018
;
10
:
e010993
.

49

Dakik
 
HA
,
Chehab
 
O
,
Eldirani
 
M
,
Sbeity
 
E
,
Karam
 
C
,
Abou Hassan
 
O
, et al.  
A new index for pre-operative cardiovascular evaluation
.
J Am Coll Cardiol
 
2019
;
73
:
3067
3078
.

50

Dakik
 
HA
,
Sbaity
 
E
,
Msheik
 
A
,
Kaspar
 
C
,
Eldirani
 
M
,
Chehab
 
O
, et al.  
AUB-HAS2 Cardiovascular Risk Index: performance in surgical subpopulations and comparison to the Revised Cardiac Risk Index
.
J Am Heart Assoc
 
2020
;
9
:
e016228
.

51

Dakik
 
HA
,
Eldirani
 
M
,
Kaspar
 
C
,
Chehab
 
O
,
Msheik
 
A
,
Makki
 
M
, et al.  
Prospective validation of the AUB-HAS2 Cardiovascular Risk Index
.
Eur Heart J Qual Care Clin Outcomes
 
2022
;
8
:
96
97
.

52

Duceppe
 
E
,
Patel
 
A
,
Chan
 
MTV
,
Berwanger
 
O
,
Ackland
 
G
,
Kavsak
 
PA
, et al.  
Preoperative N-terminal pro-B-type natriuretic peptide and cardiovascular events after noncardiac surgery: a cohort study
.
Ann Intern Med
 
2020
;
172
:
96
104
.

53

Weber
 
M
,
Luchner
 
A
,
Seeberger
 
M
,
Mueller
 
C
,
Liebetrau
 
C
,
Schlitt
 
A
, et al.  
Incremental value of high-sensitive troponin T in addition to the revised cardiac index for peri-operative risk stratification in non-cardiac surgery
.
Eur Heart J
 
2013
;
34
:
853
862
.

54

Kueh
 
SH
,
Pasley
 
T
,
Wheeler
 
M
,
Pemberton
 
J
.
The not so innocent heart murmur: a 5-year experience
.
Intern Med J
 
2017
;
47
:
199
205
.

55

Douglas
 
PS
,
Garcia
 
MJ
,
Haines
 
DE
,
Lai
 
WW
,
Manning
 
WJ
,
Patel
 
AR
, et al.  
ACCF/ASE/AHA/ASNC/HFSA/HRS/SCAI/SCCM/SCCT/SCMR 2011 Appropriate Use Criteria for Echocardiography. A Report of the American College of Cardiology Foundation Appropriate Use Criteria Task Force, American Society of Echocardiography, American Heart Association, American Society of Nuclear Cardiology, Heart Failure Society of America, Heart Rhythm Society, Society for Cardiovascular Angiography and Interventions, Society of Critical Care Medicine, Society of Cardiovascular Computed Tomography, Society for Cardiovascular Magnetic Resonance American College of Chest Physicians
.
J Am Soc Echocardiogr
 
2011
;
24
:
229
267
.

56

Friedman
 
JM
,
Couso
 
R
,
Kitchens
 
M
,
Vakhshori
 
V
,
Hillin
 
CD
,
Wu
 
CH
, et al.  
Benign heart murmurs as a predictor for complications following total joint arthroplasty
.
J Orthop
 
2017
;
14
:
470
474
.

57

Hip Attack Investigators
.
Accelerated surgery versus standard care in hip fracture (HIP ATTACK): an international, randomised, controlled trial
.
Lancet
 
2020
;
395
:
698
708
.

58

Cowie
 
B
.
The preoperative patient with a systolic murmur
.
Anesth Pain Med
 
2015
;
5
:
e32105
.

59

Attenhofer Jost
 
CH
,
Turina
 
J
,
Mayer
 
K
,
Seifert
 
B
,
Amann
 
FW
,
Buechi
 
M
, et al.  
Echocardiography in the evaluation of systolic murmurs of unknown cause
.
Am J Med
 
2000
;
108
:
614
620
.

60

Kobal
 
SL
,
Trento
 
L
,
Baharami
 
S
,
Tolstrup
 
K
,
Naqvi
 
TZ
,
Cercek
 
B
, et al.  
Comparison of effectiveness of hand-carried ultrasound to bedside cardiovascular physical examination
.
Am J Cardiol
 
2005
;
96
:
1002
1006
.

61

Abidov
 
A
,
Rozanski
 
A
,
Hachamovitch
 
R
,
Hayes
 
SW
,
Aboul-Enein
 
F
,
Cohen
 
I
, et al.  
Prognostic significance of dyspnea in patients referred for cardiac stress testing
.
N Engl J Med
 
2005
;
353
:
1889
1898
.

62

Cajita
 
MI
,
Cajita
 
TR
,
Han
 
HR
.
Health literacy and heart failure: a systematic review
.
J Cardiovasc Nurs
 
2016
;
31
:
121
130
.

63

Magnani
 
JW
,
Mujahid
 
MS
,
Aronow
 
HD
,
Cene
 
CW
,
Dickson
 
VV
,
Havranek
 
E
, et al.  
Health literacy and cardiovascular disease. Fundamental relevance to primary and secondary prevention: a scientific statement from the American Heart Association
.
Circulation
 
2018
;
138
:
e48
e74
.

64

Shinkunas
 
LA
,
Klipowicz
 
CJ
,
Carlisle
 
EM
.
Shared decision making in surgery: a scoping review of patient and surgeon preferences
.
BMC Med Inform Decis Mak
 
2020
;
20
:
190
.

65

de Mik
 
SML
,
Stubenrouch
 
FE
,
Balm
 
R
,
Ubbink
 
DT
.
Systematic review of shared decision-making in surgery
.
Br J Surg
 
2018
;
105
:
1721
1730
.

66

Pham
 
C
,
Lizarondo
 
L
,
Karnon
 
J
,
Aromataris
 
E
,
Munn
 
Z
,
Gibb
 
C
, et al.  
Strategies for implementing shared decision making in elective surgery by health care practitioners: a systematic review
.
J Eval Clin Pract
 
2020
;
26
:
582
601
.

67

Niburski
 
K
,
Guadagno
 
E
,
Abbasgholizadeh-Rahimi
 
S
,
Poenaru
 
D
.
Shared decision making in surgery: a meta-analysis of existing literature
.
Patient
 
2020
;
13
:
667
681
.

68

Abate
 
SM
,
Cheko
 
YA
,
Basu
 
B
.
Global prevalence and determinants of preoperative anxiety among surgical patients: a systematic review and meta-analysis
.
Int J Surg Open
 
2020
;
25
:
6
16
.

69

Takagi
 
H
,
Ando
 
T
,
Umemoto
 
T
,
Group
 
ALICE
.
Perioperative depression or anxiety and postoperative mortality in cardiac surgery: a systematic review and meta-analysis
.
Heart Vessels
 
2017
;
32
:
1458
1468
.

70

Tully
 
PJ
,
Baker
 
RA
.
Depression, anxiety, and cardiac morbidity outcomes after coronary artery bypass surgery: a contemporary and practical review
.
J Geriatr Cardiol
 
2012
;
9
:
197
208
.

71

Rosenberger
 
PH
,
Jokl
 
P
,
Ickovics
 
J
.
Psychosocial factors and surgical outcomes: an evidence-based literature review
.
J Am Acad Orthop Surg
 
2006
;
14
:
397
405
.

72

Theunissen
 
M
,
Peters
 
ML
,
Bruce
 
J
,
Gramke
 
HF
,
Marcus
 
MA
.
Preoperative anxiety and catastrophizing: a systematic review and meta-analysis of the association with chronic postsurgical pain
.
Clin J Pain
 
2012
;
28
:
819
841
.

73

Szeverenyi
 
C
,
Kekecs
 
Z
,
Johnson
 
A
,
Elkins
 
G
,
Csernatony
 
Z
,
Varga
 
K
.
The use of adjunct psychosocial interventions can decrease postoperative pain and improve the quality of clinical care in orthopedic surgery: a systematic review and meta-analysis of randomized controlled trials
.
J Pain
 
2018
;
19
:
1231
1252
.

74

Villa
 
G
,
Lanini
 
I
,
Amass
 
T
,
Bocciero
 
V
,
Scire Calabrisotto
 
C
,
Chelazzi
 
C
, et al.  
Effects of psychological interventions on anxiety and pain in patients undergoing major elective abdominal surgery: a systematic review
.
Perioper Med (Lond)
 
2020
;
9
:
38
.

75

Richards
 
SH
,
Anderson
 
L
,
Jenkinson
 
CE
,
Whalley
 
B
,
Rees
 
K
,
Davies
 
P
, et al.  
Psychological interventions for coronary heart disease
.
Cochrane Database Syst Rev
 
2017
;
4
:
CD002902
.

76

Bilimoria
 
KY
,
Liu
 
Y
,
Paruch
 
JL
,
Zhou
 
L
,
Kmiecik
 
TE
,
Ko
 
CY
, et al.  
Development and evaluation of the universal ACS NSQIP surgical risk calculator: a decision aid and informed consent tool for patients and surgeons
.
J Am Coll Surg
 
2013
;
217
:
833
842.e831–833
.

76a

Fronczek
 
J
,
Polok
 
K
,
Devereaux
 
PJ
, et al.  
External validation of the Revised Cardiac Risk Index and National Surgical Quality Improvement Program Myocardial Infarction and Cardiac Arrest calculator in noncardiac vascular surgery
.
Br J Anaesth
.
2019
;
123
(
4
):
421
429
. doi:.

77

Bertges
 
DJ
,
Goodney
 
PP
,
Zhao
 
Y
,
Schanzer
 
A
,
Nolan
 
BW
,
Likosky
 
DS
, et al.  
The Vascular Study Group of New England Cardiac Risk Index (VSG-CRI) predicts cardiac complications more accurately than the Revised Cardiac Risk Index in vascular surgery patients
.
J Vasc Surg
 
2010
;
52
:
674
683.683.e1–683.e3
.

78

Protopapa
 
KL
,
Simpson
 
JC
,
Smith
 
NC
,
Moonesinghe
 
SR
.
Development and validation of the Surgical Outcome Risk Tool (SORT)
.
Br J Surg
 
2014
;
101
:
1774
1783
.

79

Wong
 
DJN
,
Harris
 
S
,
Sahni
 
A
,
Bedford
 
JR
,
Cortes
 
L
,
Shawyer
 
R
, et al.  
Developing and validating subjective and objective risk-assessment measures for predicting mortality after major surgery: an international prospective cohort study
.
PLoS Med
 
2020
;
17
:
e1003253
.

80

Gupta
 
PK
,
Gupta
 
H
,
Sundaram
 
A
,
Kaushik
 
M
,
Fang
 
X
,
Miller
 
WJ
, et al.  
Development and validation of a risk calculator for prediction of cardiac risk after surgery
.
Circulation
 
2011
;
124
:
381
387
.

81

Msheik
 
A
,
Kaspar
 
C
,
Mailhac
 
A
,
Hoballah
 
JJ
,
Tamim
 
H
,
Dakik
 
HA
.
Performance of the AUB-HAS2 Cardiovascular Risk Index in vascular surgery patients
.
Vasc Med
 
2021
;
26
:
535
541
.

82

Glance
 
LG
,
Faden
 
E
,
Dutton
 
RP
,
Lustik
 
SJ
,
Li
 
Y
,
Eaton
 
MP
, et al.  
Impact of the choice of risk model for identifying low-risk patients using the 2014 American College of Cardiology/American Heart Association Perioperative Guidelines
.
Anesthesiology
 
2018
;
129
:
889
900
.

83

Robinson
 
TN
,
Walston
 
JD
,
Brummel
 
NE
,
Deiner
 
S
,
Brown
 
C
,
Kennedy
 
M
, et al.  
Frailty for surgeons: review of a National Institute on Aging Conference on frailty for specialists
.
J Am Coll Surg
 
2015
;
221
:
1083
1092
.

84

Richter
 
D
,
Guasti
 
L
,
Walker
 
D
,
Lambrinou
 
E
,
Lionis
 
C
,
Abreu
 
A
, et al.  
Frailty in cardiology: definition, assessment and clinical implications for general cardiology. A consensus document of the Council for Cardiology Practice (CCP), Association for Acute Cardio Vascular Care (ACVC), Association of Cardiovascular Nursing and Allied Professions (ACNAP), European Association of Preventive Cardiology (EAPC), European Heart Rhythm Association (EHRA), Council on Valvular Heart Diseases (VHD), Council on Hypertension (CHT), Council of Cardio-Oncology (CCO), Working Group (WG) Aorta and Peripheral Vascular Diseases, WG e-Cardiology, WG Thrombosis, of the European Society of Cardiology, European Primary Care Cardiology Society (EPCCS)
.
Eur J Prev Cardiol
 
2022
;
29
:
216
227
.

85

Hornor
 
MA
,
Ma
 
M
,
Zhou
 
L
,
Cohen
 
ME
,
Rosenthal
 
RA
,
Russell
 
MM
, et al.  
Enhancing the American College of Surgeons NSQIP Surgical Risk Calculator to predict geriatric outcomes
.
J Am Coll Surg
 
2020
;
230
:
88
100 e101
.

86

Tjeertes
 
EKM
,
van Fessem
 
JMK
,
Mattace-Raso
 
FUS
,
Hoofwijk
 
AGM
,
Stolker
 
RJ
.
Hoeks SE. Influence of frailty on outcome in older patients undergoing non-cardiac surgery – a systematic review and meta-analysis
.
Aging Dis
 
2020
;
11
:
1276
1290
.

87

Mosquera
 
C
,
Spaniolas
 
K
,
Fitzgerald
 
TL
.
Impact of frailty on surgical outcomes: the right patient for the right procedure
.
Surgery
 
2016
;
160
:
272
280
.

88

Rockwood
 
K
,
Song
 
X
,
MacKnight
 
C
,
Bergman
 
H
,
Hogan
 
DB
,
McDowell
 
I
, et al.  
A global clinical measure of fitness and frailty in elderly people
.
CMAJ
 
2005
;
173
:
489
495
.

89

Fried
 
LP
,
Tangen
 
CM
,
Walston
 
J
,
Newman
 
AB
,
Hirsch
 
C
,
Gottdiener
 
J
, et al.  
Frailty in older adults: evidence for a phenotype
.
J Gerontol A Biol Sci Med Sci
 
2001
;
56
:
M146
M156
.

90

Alvarez-Nebreda
 
ML
,
Bentov
 
N
,
Urman
 
RD
,
Setia
 
S
,
Huang
 
JC
,
Pfeifer
 
K
, et al.  
Recommendations for preoperative management of frailty from the Society for Perioperative Assessment and Quality Improvement (SPAQI)
.
J Clin Anesth
 
2018
;
47
:
33
42
.

91

Grigoryan
 
KV
,
Javedan
 
H
,
Rudolph
 
JL
.
Orthogeriatric care models and outcomes in hip fracture patients: a systematic review and meta-analysis
.
J Orthop Trauma
 
2014
;
28
:
e49
e55
.

92

Borson
 
S
,
Scanlan
 
JM
,
Chen
 
P
,
Ganguli
 
M
.
The Mini-Cog as a screen for dementia: validation in a population-based sample
.
J Am Geriatr Soc
 
2003
;
51
:
1451
1454
.

93

Biccard
 
BM
.
Relationship between the inability to climb two flights of stairs and outcome after major non-cardiac surgery: implications for the pre-operative assessment of functional capacity
.
Anaesthesia
 
2005
;
60
:
588
593
.

94

Lurati Buse
 
GAL
,
Puelacher
 
C
,
Menosi Gualandro
 
D
,
Genini
 
AS
,
Hidvegi
 
R
,
Bolliger
 
D
, et al.  
Association between self-reported functional capacity and major adverse cardiac events in patients at elevated risk undergoing noncardiac surgery: a prospective diagnostic cohort study
.
Br J Anaesth
 
2020
;
126
:
102
110
.

95

Wijeysundera
 
DN
,
Pearse
 
RM
,
Shulman
 
MA
,
Abbott
 
TEF
,
Torres
 
E
,
Ambosta
 
A
, et al.  
Assessment of functional capacity before major non-cardiac surgery: an international, prospective cohort study
.
Lancet
 
2018
;
391
:
2631
2640
.

96

Wijeysundera
 
DN
,
Beattie
 
WS
,
Hillis
 
GS
,
Abbott
 
TEF
,
Shulman
 
MA
,
Ackland
 
GL
, et al.  
Integration of the Duke Activity Status Index into preoperative risk evaluation: a multicentre prospective cohort study
.
Br J Anaesth
 
2020
;
124
:
261
270
.

97

Jeger
 
RV
,
Probst
 
C
,
Arsenic
 
R
,
Lippuner
 
T
,
Pfisterer
 
ME
,
Seeberger
 
MD
, et al.  
Long-term prognostic value of the preoperative 12-lead electrocardiogram before major noncardiac surgery in coronary artery disease
.
Am Heart J
 
2006
;
151
:
508
513
.

98

Collet
 
JP
,
Thiele
 
H
,
Barbato
 
E
,
Barthelemy
 
O
,
Bauersachs
 
J
,
Bhatt
 
DL
, et al.  
2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation
.
Eur Heart J
 
2021
;
42
:
1289
1367
.

99

Hindricks
 
G
,
Potpara
 
T
,
Dagres
 
N
,
Arbelo
 
E
,
Bax
 
JJ
,
Blomstrom-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)
.
Eur Heart J
 
2021
;
42
:
373
498
.

100

Ganguli
 
I
,
Lupo
 
C
,
Mainor
 
AJ
,
Raymond
 
S
,
Wang
 
Q
,
Orav
 
EJ
, et al.  
Prevalence and cost of care cascades after low-value preoperative electrocardiogram for cataract surgery in fee-for-service medicare beneficiaries
.
JAMA Intern Med
 
2019
;
179
:
1211
1219
.

101

Gualandro
 
DM
,
Puelacher
 
C
,
LuratiBuse
 
G
,
Lampart
 
A
,
Strunz
 
C
,
Cardozo
 
FA
, et al.  
Comparison of high-sensitivity cardiac troponin I and T for the prediction of cardiac complications after non-cardiac surgery
.
Am Heart J
 
2018
;
203
:
67
73
.

102

Humble
 
CAS
,
Huang
 
S
,
Jammer
 
I
,
Bjork
 
J
,
Chew
 
MS
.
Prognostic performance of preoperative cardiac troponin and perioperative changes in cardiac troponin for the prediction of major adverse cardiac events and mortality in noncardiac surgery: a systematic review and meta-analysis
.
PLoS One
 
2019
;
14
:
e0215094
.

103

Park
 
SJ
,
Choi
 
JH
,
Cho
 
SJ
,
Chang
 
SA
,
Choi
 
JO
,
Lee
 
SC
, et al.  
Comparison of transthoracic echocardiography with N-terminal pro-brain natriuretic peptide as a tool for risk stratification of patients undergoing major noncardiac surgery
.
Korean Circ J
 
2011
;
41
:
505
511
.

104

Lee
 
G
,
Sou
 
SM
,
Twerenbold
 
R
,
Reichlin
 
T
,
Oshima
 
S
,
Hochgruber
 
T
, et al.  
B-type natriuretic peptide and clinical judgment in the detection of exercise-induced myocardial ischemia
.
Am J Med
 
2014
;
127
:
427
435
.

105

Walter
 
J
,
de Lavallaz J
 
dF
,
Koechlin
 
L
,
Zimmermann
 
T
,
Boeddinghaus
 
J
,
Honegger
 
U
, et al.  
Using high-sensitivity cardiac troponin for the exclusion of inducible myocardial ischemia in symptomatic patients: a cohort study
.
Ann Intern Med
 
2020
;
172
:
175
185
.

106

Walter
 
JE
,
Honegger
 
U
,
Puelacher
 
C
,
Mueller
 
D
,
Wagener
 
M
,
Schaerli
 
N
, et al.  
Prospective validation of a biomarker-based rule out strategy for functionally relevant coronary artery disease
.
Clin Chem
 
2018
;
64
:
386
395
.

107

Mueller
 
D
,
Puelacher
 
C
,
Honegger
 
U
,
Walter
 
JE
,
Badertscher
 
P
,
Schaerli
 
N
, et al.  
Direct comparison of cardiac troponin T and I using a uniform and a sex-specific approach in the detection of functionally relevant coronary artery disease
.
Clin Chem
 
2018
;
64
:
1596
1606
.

108

Ackland
 
GL
,
Abbott
 
TEF
,
Cain
 
D
,
Edwards
 
MR
,
Sultan
 
P
,
Karmali
 
SN
, et al.  
Preoperative systemic inflammation and perioperative myocardial injury: prospective observational multicentre cohort study of patients undergoing non-cardiac surgery
.
Br J Anaesth
 
2019
;
122
:
180
187
.

109

Writing Committee for the Vision Study Investigators
,
Devereaux
 
PJ
,
Biccard
 
BM
,
Sigamani
 
A
,
Xavier
 
D
,
Chan
 
MTV
, et al.  
Association of postoperative high-sensitivity troponin levels with myocardial injury and 30-day mortality among patients undergoing noncardiac surgery
.
JAMA
 
2017
;
317
:
1642
1651
.

110

Puelacher
 
C
,
Gualandro
 
DM
,
Lurati Buse
 
G
,
Bolliger
 
D
,
Marbot
 
S
,
Kindler
 
C
, et al.  
Etiology of peri-operative myocardial infarction/injury after noncardiac surgery and associated outcome
.
J Am Coll Cardiol
 
2020
;
76
:
1910
1912
.

111

Puelacher
 
C
,
Lurati Buse
 
G
,
Seeberger
 
D
,
Sazgary
 
L
,
Marbot
 
S
,
Lampart
 
A
, et al.  
Perioperative myocardial injury after noncardiac surgery: incidence, mortality, and characterization
.
Circulation
 
2018
;
137
:
1221
1232
.

112

Huelsmann
 
M
,
Neuhold
 
S
,
Resl
 
M
,
Strunk
 
G
,
Brath
 
H
,
Francesconi
 
C
, et al.  
PONTIAC (NT-proBNP selected prevention of cardiac events in a population of diabetic patients without a history of cardiac disease): a prospective randomized controlled trial
.
J Am Coll Cardiol
 
2013
;
62
:
1365
1372
.

113

Ledwidge
 
M
,
Gallagher
 
J
,
Conlon
 
C
,
Tallon
 
E
,
O’Connell
 
E
,
Dawkins
 
I
, et al.  
Natriuretic peptide-based screening and collaborative care for heart failure: the STOP-HF randomized trial
.
JAMA
 
2013
;
310
:
66
74
.

114

Mueller
 
C
,
McDonald
 
K
,
de Boer
 
RA
,
Maisel
 
A
,
Cleland
 
JGF
,
Kozhuharov
 
N
, et al.  
Heart Failure Association of the European Society of Cardiology practical guidance on the use of natriuretic peptide concentrations
.
Eur J Heart Fail
 
2019
;
21
:
715
731
.

115

De Hert
 
SG
,
Lurati Buse
 
GA
.
Cardiac biomarkers for the prediction and detection of adverse cardiac events after noncardiac surgery: a narrative review
.
Anesth Analg
 
2020
;
131
:
187
195
.

116

Mauermann
 
E
,
Bolliger
 
D
,
Seeberger
 
E
,
Puelacher
 
C
,
Corbiere
 
S
,
Filipovic
 
M
, et al.  
Incremental value of preoperative copeptin for predicting myocardial injury
.
Anesth Analg
 
2016
;
123
:
1363
1371
.

117

Buse
 
GL
,
Manns
 
B
,
Lamy
 
A
,
Guyatt
 
G
,
Polanczyk
 
CA
,
Chan
 
MTV
, et al.  
Troponin T monitoring to detect myocardial injury after noncardiac surgery: a cost-consequence analysis
.
Can J Surg
 
2018
;
61
:
185
194
.

118

Vascular Events In Noncardiac Surgery Patients Cohort Evaluation Study Investigators
,
Devereaux
 
PJ
,
Chan
 
MT
,
Alonso-Coello
 
P
,
Walsh
 
M
,
Berwanger
 
O
, et al.  
Association between postoperative troponin levels and 30-day mortality among patients undergoing noncardiac surgery
.
JAMA
 
2012
;
307
:
2295
2304
.

119

Mantha
 
S
,
Foss
 
J
,
Ellis
 
JE
,
Roizen
 
MF
.
Intense cardiac troponin surveillance for long-term benefits is cost-effective in patients undergoing open abdominal aortic surgery: a decision analysis model
.
Anesth Analg
 
2007
;
105
:
1346
1356
.

120

Levitan
 
EB
,
Graham
 
LA
,
Valle
 
JA
,
Richman
 
JS
,
Hollis
 
R
,
Holcomb
 
CN
, et al.  
Pre-operative echocardiography among patients with coronary artery disease in the United States Veterans Affairs healthcare system: a retrospective cohort study
.
BMC Cardiovasc Disord
 
2016
;
16
:
173
.

121

Chang
 
HY
,
Chang
 
WT
,
Liu
 
YW
.
Application of transthoracic echocardiography in patients receiving intermediate- or high-risk noncardiac surgery
.
PLoS One
 
2019
;
14
:
e0215854
.

122

Wijeysundera
 
DN
,
Beattie
 
WS
,
Karkouti
 
K
,
Neuman
 
MD
,
Austin
 
PC
,
Laupacis
 
A
.
Association of echocardiography before major elective non-cardiac surgery with postoperative survival and length of hospital stay: population based cohort study
.
BMJ
 
2011
;
342
:
d3695
.

123

Faris
 
JG
,
Hartley
 
K
,
Fuller
 
CM
,
Langston
 
RB
,
Royse
 
CF
,
Veltman
 
MG
.
Audit of cardiac pathology detection using a criteria-based perioperative echocardiography service
.
Anaesth Intensive Care
 
2012
;
40
:
702
709
.

124

Steeds
 
RP
,
Garbi
 
M
,
Cardim
 
N
,
Kasprzak
 
JD
,
Sade
 
E
,
Nihoyannopoulos
 
P
, et al.  
EACVI appropriateness criteria for the use of transthoracic echocardiography in adults: a report of literature and current practice review
.
Eur Heart J Cardiovasc Imaging
 
2017
;
18
:
1191
1204
.

125

Kertai
 
MD
,
Boersma
 
E
,
Bax
 
JJ
,
Heijenbrok-Kal
 
MH
,
Hunink
 
MG
,
L'Talien G
 
J
, et al.  
A meta-analysis comparing the prognostic accuracy of six diagnostic tests for predicting perioperative cardiac risk in patients undergoing major vascular surgery
.
Heart
 
2003
;
89
:
1327
1334
.

126

Halm
 
EA
,
Browner
 
WS
,
Tubau
 
JF
,
Tateo
 
IM
,
Mangano
 
DT
.
Echocardiography for assessing cardiac risk in patients having noncardiac surgery. Study of Perioperative Ischemia Research Group
.
Ann Intern Med
 
1996
;
125
:
433
441
.

127

Rohde
 
LE
,
Polanczyk
 
CA
,
Goldman
 
L
,
Cook
 
EF
,
Lee
 
RT
,
Lee
 
TH
.
Usefulness of transthoracic echocardiography as a tool for risk stratification of patients undergoing major noncardiac surgery
.
Am J Cardiol
 
2001
;
87
:
505
509
.

128

Galasko
 
GI
,
Barnes
 
SC
,
Collinson
 
P
,
Lahiri
 
A
,
Senior
 
R
.
What is the most cost-effective strategy to screen for left ventricular systolic dysfunction: natriuretic peptides, the electrocardiogram, hand-held echocardiography, traditional echocardiography, or their combination?
 
Eur Heart J
 
2006
;
27
:
193
200
.

129

Cardim
 
N
,
Dalen
 
H
,
Voigt
 
JU
,
Ionescu
 
A
,
Price
 
S
,
Neskovic
 
AN
, et al.  
The use of handheld ultrasound devices: a position statement of the European Association of Cardiovascular Imaging (2018 update)
.
Eur Heart J Cardiovasc Imaging
 
2019
;
20
:
245
252
.

130

Canty
 
DJ
,
Royse
 
CF
,
Kilpatrick
 
D
,
Bowman
 
L
,
Royse
 
AG
.
The impact of focused transthoracic echocardiography in the pre-operative clinic
.
Anaesthesia
 
2012
;
67
:
618
625
.

131

Heiberg
 
J
,
El-Ansary
 
D
,
Canty
 
DJ
,
Royse
 
AG
,
Royse
 
CF
.
Focused echocardiography: a systematic review of diagnostic and clinical decision-making in anaesthesia and critical care
.
Anaesthesia
 
2016
;
71
:
1091
1100
.

132

Canty
 
DJ
,
Royse
 
CF
.
Audit of anaesthetist-performed echocardiography on perioperative management decisions for non-cardiac surgery
.
Br J Anaesth
 
2009
;
103
:
352
358
.

133

Canty
 
DJ
,
Heiberg
 
J
,
Yang
 
Y
,
Royse
 
AG
,
Margale
 
S
,
Nanjappa
 
N
, et al.  
One-year results of the pilot multicentre randomised trial of preoperative focused cardiac ultrasound in hip fracture surgery
.
Anaesth Intensive Care
 
2019
;
47
:
207
208
.

134

Matyal
 
R
,
Hess
 
PE
,
Subramaniam
 
B
,
Mitchell
 
J
,
Panzica
 
PJ
,
Pomposelli
 
F
, et al.  
Perioperative diastolic dysfunction during vascular surgery and its association with postoperative outcome
.
J Vasc Surg
 
2009
;
50
:
70
76
.

135

Saito
 
S
,
Takagi
 
A
,
Kurokawa
 
F
,
Ashihara
 
K
,
Hagiwara
 
N
.
Usefulness of tissue Doppler echocardiography to predict perioperative cardiac events in patients undergoing noncardiac surgery
.
Heart Vessels
 
2012
;
27
:
594
602
.

136

Higashi
 
M
,
Yamaura
 
K
,
Ikeda
 
M
,
Shimauchi
 
T
,
Saiki
 
H
,
Hoka
 
S
.
Diastolic dysfunction of the left ventricle is associated with pulmonary edema after renal transplantation
.
Acta Anaesthesiol Scand
 
2013
;
57
:
1154
1160
.

137

Brecher
 
O
,
Gulati
 
H
,
Roistacher
 
N
,
Zhang
 
H
,
Shi
 
W
,
Thaler
 
HT
, et al.  
Preoperative echocardiographic indices of diastolic dysfunction and brain natriuretic peptide in predicting postoperative atrial fibrillation after noncardiac surgery
.
Anesth Analg
 
2017
;
124
:
1099
1104
.

138

Zhou
 
Y
,
Liu
 
L
,
Cheng
 
T
,
Wang
 
DX
,
Yang
 
HY
,
Zhang
 
BW
, et al.  
Grade 3 echocardiographic diastolic dysfunction is associated with increased risk of major adverse cardiovascular events after surgery: a retrospective cohort study
.
Anesth Analg
 
2019
;
129
:
651
658
.

139

Fayad
 
A
,
Ansari
 
MT
,
Yang
 
H
,
Ruddy
 
T
,
Wells
 
GA
.
Perioperative diastolic dysfunction in patients undergoing noncardiac surgery is an independent risk factor for cardiovascular events: a systematic review and meta-analysis
.
Anesthesiology
 
2016
;
125
:
72
91
.

140

Willingham
 
M
,
Ayoubi
 
SA
,
Doan
 
M
,
Wingert
 
T
,
Scovotti
 
J
,
Howard-Quijano
 
K
, et al.  
Preoperative diastolic dysfunction and postoperative outcomes after noncardiac surgery
.
J Cardiothorac Vasc Anesth
 
2020
;
34
:
679
686
.

141

Sougawa
 
H
,
Ino
 
Y
,
Kitabata
 
H
,
Tanimoto
 
T
,
Takahata
 
M
,
Shimamura
 
K
, et al.  
Impact of left ventricular ejection fraction and preoperative hemoglobin level on perioperative adverse cardiovascular events in noncardiac surgery
.
Heart Vessels
 
2021
;
36
:
1317
1326
.

142

Hreybe
 
H
,
Zahid
 
M
,
Sonel
 
A
,
Good
 
CB
,
Shaver
 
J
,
Saba
 
S
.
Noncardiac surgery and the risk of death and other cardiovascular events in patients with hypertrophic cardiomyopathy
.
Clin Cardiol
 
2006
;
29
:
65
68
.

143

Kertai
 
MD
,
Bountioukos
 
M
,
Boersma
 
E
,
Bax
 
JJ
,
Thomson
 
IR
,
Sozzi
 
F
, et al.  
Aortic stenosis: an underestimated risk factor for perioperative complications in patients undergoing noncardiac surgery
.
Am J Med
 
2004
;
116
:
8
13
.

144

Marbach
 
JA
,
Almufleh
 
A
,
Di Santo
 
P
,
Jung
 
R
,
Simard
 
T
,
McInnes
 
M
, et al.  
Comparative accuracy of focused cardiac ultrasonography and clinical examination for left ventricular dysfunction and valvular heart disease: a systematic review and meta-analysis
.
Ann Intern Med
 
2019
;
171
:
264
272
.

145

Lurati Buse
 
GAL
,
Puelacher
 
C
,
Gualandro
 
DM
,
Kilinc
 
D
,
Glarner
 
N
,
Hidvegi
 
R
, et al.  
Adherence to the European Society of Cardiology/European Society of Anaesthesiology recommendations on preoperative cardiac testing and association with positive results and cardiac events: a cohort study
.
Br J Anaesth
 
2021
;
127
:
376
385
.

146

Knuuti
 
J
,
Wijns
 
W
,
Saraste
 
A
,
Capodanno
 
D
,
Barbato
 
E
,
Funck-Brentano
 
C
, et al.  
2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes
.
Eur Heart J
 
2020
;
41
:
407
477
.

147

Wolk
 
MJ
,
Bailey
 
SR
,
Doherty
 
JU
,
Douglas
 
PS
,
Hendel
 
RC
,
Kramer
 
CM
, et al.  
ACCF/AHA/ASE/ASNC/HFSA/HRS/SCAI/SCCT/SCMR/STS 2013 multimodality appropriate use criteria for the detection and risk assessment of stable ischemic heart disease: a report of the American College of Cardiology Foundation Appropriate Use Criteria Task Force, American Heart Association, American Society of Echocardiography, American Society of Nuclear Cardiology, Heart Failure Society of America, Heart Rhythm Society, Society for Cardiovascular Angiography and Interventions, Society of Cardiovascular Computed Tomography, Society for Cardiovascular Magnetic Resonance, and Society of Thoracic Surgeons
.
J Am Coll Cardiol
 
2014
;
63
:
380
406
.

148

Pellikka
 
PA
,
Arruda-Olson
 
A
,
Chaudhry
 
FA
,
Chen
 
MH
,
Marshall
 
JE
,
Porter
 
TR
, et al.  
Guidelines for performance, interpretation, and application of stress echocardiography in ischemic heart disease: from the American Society of Echocardiography
.
J Am Soc Echocardiogr
 
2020
;
33
:
1
41.e8
.

149

Koh
 
AS
,
Flores
 
JL
,
Keng
 
FY
,
Tan
 
RS
,
Chua
 
TS
.
Correlation between clinical outcomes and appropriateness grading for referral to myocardial perfusion imaging for preoperative evaluation prior to non-cardiac surgery
.
J Nucl Cardiol
 
2012
;
19
:
277
284
.

150

Nguyen
 
P
,
Plotkin
 
J
,
Fishbein
 
TM
,
Laurin
 
JM
,
Satoskar
 
R
,
Shetty
 
K
, et al.  
Dobutamine stress echocardiography in patients undergoing orthotopic liver transplantation: a pooled analysis of accuracy, perioperative and long term cardiovascular prognosis
.
Int J Cardiovasc Imaging
 
2013
;
29
:
1741
1748
.

151

Shaw
 
LJ
,
Eagle
 
KA
,
Gersh
 
BJ
,
Miller
 
DD
.
Meta-analysis of intravenous dipyridamole-thallium-201 imaging (1985 to 1994) and dobutamine echocardiography (1991 to 1994) for risk stratification before vascular surgery
.
J Am Coll Cardiol
 
1996
;
27
:
787
798
.

152

Beattie
 
WS
,
Abdelnaem
 
E
,
Wijeysundera
 
DN
,
Buckley
 
DN
.
A meta-analytic comparison of preoperative stress echocardiography and nuclear scintigraphy imaging
.
Anesth Analg
 
2006
;
102
:
8
16
.

153

Bach
 
DS
,
Eagle
 
KA
.
Dobutamine stress echocardiography. Stressing the indications for preoperative testing
.
Circulation
 
1997
;
95
:
8
10
.

154

Etchells
 
E
,
Meade
 
M
,
Tomlinson
 
G
,
Cook
 
D
.
Semiquantitative dipyridamole myocardial stress perfusion imaging for cardiac risk assessment before noncardiac vascular surgery: a meta-analysis
.
J Vasc Surg
 
2002
;
36
:
534
540
.

155

Ballal
 
RS
,
Kapadia
 
S
,
Secknus
 
MA
,
Rubin
 
D
,
Arheart
 
K
,
Marwick
 
TH
.
Prognosis of patients with vascular disease after clinical evaluation and dobutamine stress echocardiography
.
Am Heart J
 
1999
;
137
:
469
475
.

156

Boersma
 
E
,
Poldermans
 
D
,
Bax
 
JJ
,
Steyerberg
 
EW
,
Thomson
 
IR
,
Banga
 
JD
, et al.  
Predictors of cardiac events after major vascular surgery: role of clinical characteristics, dobutamine echocardiography, and beta-blocker therapy
.
JAMA
 
2001
;
285
:
1865
1873
.

157

Cullen
 
MW
,
McCully
 
RB
,
Widmer
 
RJ
,
Schroeder
 
DR
,
Salonen
 
BR
,
Raslau
 
D
, et al.  
Preoperative dobutamine stress echocardiography and clinical factors for assessment of cardiac risk after noncardiac surgery
.
J Am Soc Echocardiogr
 
2020
;
33
:
423
432
.

158

Van Damme
 
H
,
Pierard
 
L
,
Gillain
 
D
,
Benoit
 
T
,
Rigo
 
P
,
Limet
 
R
.
Cardiac risk assessment before vascular surgery: a prospective study comparing clinical evaluation, dobutamine stress echocardiography, and dobutamine Tc-99m sestamibi tomoscintigraphy
.
Cardiovasc Surg
 
1997
;
5
:
54
64
.

159

Metz
 
LD
,
Beattie
 
M
,
Hom
 
R
,
Redberg
 
RF
,
Grady
 
D
,
Fleischmann
 
KE
.
The prognostic value of normal exercise myocardial perfusion imaging and exercise echocardiography: a meta-analysis
.
J Am Coll Cardiol
 
2007
;
49
:
227
237
.

160

Cohen
 
MC
,
Siewers
 
AE
,
Dickens
 
JD
, Jr.
,
Hill
 
T
,
Muller
 
JE
.
Perioperative and long-term prognostic value of dipyridamole Tc-99m sestamibi myocardial tomography in patients evaluated for elective vascular surgery
.
J Nucl Cardiol
 
2003
;
10
:
464
472
.

161

Dowsley
 
TF
,
Sheth
 
T
,
Chow
 
BJW
.
Complementary pre-operative risk assessment using coronary computed tomography angiography and nuclear myocardial perfusion imaging in non-cardiac surgery: a VISION-CTA sub-study
.
J Nucl Cardiol
 
2020
;
27
:
1331
1337
.

162

Das
 
MK
,
Pellikka
 
PA
,
Mahoney
 
DW
,
Roger
 
VL
,
Oh
 
JK
,
McCully
 
RB
, et al.  
Assessment of cardiac risk before nonvascular surgery: dobutamine stress echocardiography in 530 patients
.
J Am Coll Cardiol
 
2000
;
35
:
1647
1653
.

163

Torres
 
MR
,
Short
 
L
,
Baglin
 
T
,
Case
 
C
,
Gibbs
 
H
,
Marwick
 
TH
.
Usefulness of clinical risk markers and ischemic threshold to stratify risk in patients undergoing major noncardiac surgery
.
Am J Cardiol
 
2002
;
90
:
238
242
.

164

Labib
 
SB
,
Goldstein
 
M
,
Kinnunen
 
PM
,
Schick
 
EC
.
Cardiac events in patients with negative maximal versus negative submaximal dobutamine echocardiograms undergoing noncardiac surgery: importance of resting wall motion abnormalities
.
J Am Coll Cardiol
 
2004
;
44
:
82
87
.

165

Lancellotti
 
P
,
Pellikka
 
PA
,
Budts
 
W
,
Chaudhry
 
FA
,
Donal
 
E
,
Dulgheru
 
R
, et al.  
The clinical use of stress echocardiography in non-ischaemic heart disease: recommendations from the European Association of Cardiovascular Imaging and the American Society of Echocardiography
.
Eur Heart J Cardiovasc Imaging
 
2016
;
17
:
1191
1229
.

166

Nandalur
 
KR
,
Dwamena
 
BA
,
Choudhri
 
AF
,
Nandalur
 
MR
,
Carlos
 
RC
.
Diagnostic performance of stress cardiac magnetic resonance imaging in the detection of coronary artery disease: a meta-analysis
.
J Am Coll Cardiol
 
2007
;
50
:
1343
1353
.

167

Vavere
 
AL
,
Arbab-Zadeh
 
A
,
Rochitte
 
CE
,
Dewey
 
M
,
Niinuma
 
H
,
Gottlieb
 
I
, et al.  
Coronary artery stenoses: accuracy of 64-detector row CT angiography in segments with mild, moderate, or severe calcification – a subanalysis of the CORE-64 trial
.
Radiology
 
2011
;
261
:
100
108
.

168

Sheth
 
T
,
Chan
 
M
,
Butler
 
C
,
Chow
 
B
,
Tandon
 
V
,
Nagele
 
P
, et al.  
Prognostic capabilities of coronary computed tomographic angiography before non-cardiac surgery: prospective cohort study
.
BMJ
 
2015
;
350
:
h1907
.

169

Krievins
 
D
,
Zellans
 
E
,
Latkovskis
 
G
,
Jegere
 
S
,
Kumsars
 
I
,
Kaufmanis
 
K
, et al.  
Diagnosis and management of silent coronary ischemia in patients undergoing carotid endarterectomy
.
J Vasc Surg
 
2021
;
73
:
533
541
.

170

Krievins
 
D
,
Zellans
 
E
,
Latkovskis
 
G
,
Erglis
 
A
,
Zvaigzne
 
L
,
Kumsars
 
I
, et al.  
Pre-operative diagnosis of silent coronary ischaemia may reduce post-operative death and myocardial infarction and improve survival of patients undergoing lower extremity surgical revascularisation
.
Eur J Vasc Endovasc Surg
 
2020
;
60
:
411
420
.

171

Ibanez
 
B
,
James
 
S
,
Agewall
 
S
,
Antunes
 
MJ
,
Bucciarelli-Ducci
 
C
,
Bueno
 
H
, et al.  
2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: the Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC)
.
Eur Heart J
 
2018
;
39
:
119
177
.

172

Illuminati
 
G
,
Ricco
 
JB
,
Greco
 
C
,
Mangieri
 
E
,
Calio
 
F
,
Ceccanei
 
G
, et al.  
Systematic preoperative coronary angiography and stenting improves postoperative results of carotid endarterectomy in patients with asymptomatic coronary artery disease: a randomised controlled trial
.
Eur J Vasc Endovasc Surg
 
2010
;
39
:
139
145
.

173

Sorensen
 
LT
.
Wound healing and infection in surgery. The clinical impact of smoking and smoking cessation: a systematic review and meta-analysis
.
Arch Surg
 
2012
;
147
:
373
383
.

174

Gourgiotis
 
S
,
Aloizos
 
S
,
Aravosita
 
P
,
Mystakelli
 
C
,
Isaia
 
EC
,
Gakis
 
C
, et al.  
The effects of tobacco smoking on the incidence and risk of intraoperative and postoperative complications in adults
.
Surgeon
 
2011
;
9
:
225
232
.

175

World Health Organization
.
WHO Tobacco and postsurgical outcomes
. https://apps.who.int/iris/rest/bitstreams/1265388/retrieve.

176

Mills
 
E
,
Eyawo
 
O
,
Lockhart
 
I
,
Kelly
 
S
,
Wu
 
P
,
Ebbert
 
JO
.
Smoking cessation reduces postoperative complications: a systematic review and meta-analysis
.
Am J Med
 
2011
;
124
:
144
154.e8
.

177

Lee
 
SM
,
Landry
 
J
,
Jones
 
PM
,
Buhrmann
 
O
,
Morley-Forster
 
P
.
Long-term quit rates after a perioperative smoking cessation randomized controlled trial
.
Anesth Analg
 
2015
;
120
:
582
587
.

178

Wong
 
J
,
Abrishami
 
A
,
Riazi
 
S
,
Siddiqui
 
N
,
You-Ten
 
E
,
Korman
 
J
, et al.  
A perioperative smoking cessation intervention with varenicline, counseling, and fax referral to a telephone quitline versus a brief intervention: a randomized controlled trial
.
Anesth Analg
 
2017
;
125
:
571
579
.

179

Cavalheri
 
V
,
Granger
 
C
.
Preoperative exercise training for patients with non-small cell lung cancer
.
Cochrane Database Syst Rev
 
2017
;
6
:
CD012020
.

180

Tew
 
GA
,
Ayyash
 
R
,
Durrand
 
J
,
Danjoux
 
GR
.
Clinical guideline and recommendations on pre-operative exercise training in patients awaiting major non-cardiac surgery
.
Anaesthesia
 
2018
;
73
:
750
768
.

181

Musallam
 
KM
,
Rosendaal
 
FR
,
Zaatari
 
G
,
Soweid
 
A
,
Hoballah
 
JJ
,
Sfeir
 
PM
, et al.  
Smoking and the risk of mortality and vascular and respiratory events in patients undergoing major surgery
.
JAMA Surg
 
2013
;
148
:
755
762
.

182

Turan
 
A
,
Mascha
 
EJ
,
Roberman
 
D
,
Turner
 
PL
,
You
 
J
,
Kurz
 
A
, et al.  
Smoking and perioperative outcomes
.
Anesthesiology
 
2011
;
114
:
837
846
.

183

Im
 
C
,
Oh
 
TK
,
Song
 
IA
.
Association between use of preoperative antihypertensive medication and 90-day mortality after noncardiac surgery: a retrospective cohort study.
 
Am J Hypertens
 
2020
;
33
:
534
542
.

184

Clemente-Moragon
 
A
,
Gomez
 
M
,
Villena-Gutierrez
 
R
,
Lalama
 
DV
,
Garcia-Prieto
 
J
,
Martinez
 
F
, et al.  
Metoprolol exerts a non-class effect against ischaemia-reperfusion injury by abrogating exacerbated inflammation
.
Eur Heart J
 
2020
;
41
:
4425
4440
.

185

Poise Study Group
,
Devereaux
 
PJ
,
Yang
 
H
,
Yusuf
 
S
,
Guyatt
 
G
,
Leslie
 
K
, et al.  
Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomised controlled trial
.
Lancet
 
2008
;
371
:
1839
1847
.

186

Alonso-Coello
 
P
,
Paniagua
 
P
,
Mizera
 
R
,
Devereaux
 
PJ
.
Should physicians initiate beta-blocker therapy in patients undergoing non-cardiac surgery? Insights from the POISE trial
.
Pol Arch Med Wewn
 
2008
;
118
:
616
618
.

187

Blessberger
 
H
,
Lewis
 
SR
,
Pritchard
 
MW
,
Fawcett
 
LJ
,
Domanovits
 
H
,
Schlager
 
O
, et al.  
Perioperative beta-blockers for preventing surgery-related mortality and morbidity in adults undergoing non-cardiac surgery
.
Cochrane Database Syst Rev
 
2019
;
9
:
CD013438
.

188

Friedell
 
ML
,
Van Way
 
CW
, 3rd
,
Freyberg
 
RW
,
Almenoff
 
PL
.
Beta-blockade and operative mortality in noncardiac surgery: harmful or helpful?
 
JAMA Surg
 
2015
;
150
:
658
663
.

189

Wijeysundera
 
DN
,
Duncan
 
D
,
Nkonde-Price
 
C
,
Virani
 
SS
,
Washam
 
JB
,
Fleischmann
 
KE
, et al.  
Perioperative beta blockade in noncardiac surgery: a systematic review for the 2014 ACC/AHA guideline on perioperative cardiovascular evaluation and management of patients undergoing noncardiac surgery: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines
.
J Am Coll Cardiol
 
2014
;
64
:
2406
2425
.

190

London
 
MJ
,
Hur
 
K
,
Schwartz
 
GG
,
Henderson
 
WG
.
Association of perioperative beta-blockade with mortality and cardiovascular morbidity following major noncardiac surgery
.
JAMA
 
2013
;
309
:
1704
1713
.

191

Angeli
 
F
,
Verdecchia
 
P
,
Karthikeyan
 
G
,
Mazzotta
 
G
,
Gentile
 
G
,
Reboldi
 
G
.
ß-Blockers reduce mortality in patients undergoing high-risk non-cardiac surgery
.
Am J Cardiovasc Drugs
 
2010
;
10
:
247
259
.

192

Lindenauer
 
PK
,
Pekow
 
P
,
Wang
 
K
,
Mamidi
 
DK
,
Gutierrez
 
B
,
Benjamin
 
EM
.
Perioperative beta-blocker therapy and mortality after major noncardiac surgery
.
N Engl J Med
 
2005
;
353
:
349
361
.

193

Wallace
 
AW
,
Au
 
S
,
Cason
 
BA
.
Perioperative beta-blockade: atenolol is associated with reduced mortality when compared to metoprolol
.
Anesthesiology
 
2011
;
114
:
824
836
.

194

Redelmeier
 
D
,
Scales
 
D
,
Kopp
 
A
.
Beta blockers for elective surgery in elderly patients: population based, retrospective cohort study
.
BMJ
 
2005
;
331
:
932
.

195

Ashes
 
C
,
Judelman
 
S
,
Wijeysundera
 
DN
,
Tait
 
G
,
Mazer
 
CD
,
Hare
 
GM
, et al.  
Selective beta1-antagonism with bisoprolol is associated with fewer postoperative strokes than atenolol or metoprolol: a single-center cohort study of 44,092 consecutive patients
.
Anesthesiology
 
2013
;
119
:
777
787
.

196

Shammash
 
JB
,
Trost
 
JC
,
Gold
 
JM
,
Berlin
 
JA
,
Golden
 
MA
,
Kimmel
 
SE
.
Perioperative beta-blocker withdrawal and mortality in vascular surgical patients
.
Am Heart J
 
2001
;
141
:
148
153
.

197

Wallace
 
AW
,
Au
 
S
,
Cason
 
BA
.
Association of the pattern of use of perioperative beta-blockade and postoperative mortality
.
Anesthesiology
 
2010
;
113
:
794
805
.

198

Kwon
 
S
,
Thompson
 
R
,
Florence
 
M
,
Maier
 
R
,
McIntyre
 
L
,
Rogers
 
T
, et al.  
Beta-blocker continuation after noncardiac surgery: a report from the surgical care and outcomes assessment program
.
Arch Surg
 
2012
;
147
:
467
473
.

199

Kertai
 
MD
,
Cooter
 
M
,
Pollard
 
RJ
,
Buhrman
 
W
,
Aronson
 
S
,
Mathew
 
JP
, et al.  
Is Compliance With Surgical Care Improvement Project Cardiac (SCIP-Card-2) measures for perioperative beta-blockers associated with reduced incidence of mortality and cardiovascular-related critical quality indicators after noncardiac surgery?
 
Anesth Analg
 
2018
;
126
:
1829
1838
.

200

Longrois
 
D
,
Couffignal
 
C
,
Ait-Hamou
 
N
,
Amour
 
J
,
Cholley
 
B
,
Dilly
 
M
, et al.  
Indispensable optimisation de la réintroduction des bêtabloquants après chirurgie cardiaque. Résultats de l’étude Bêtabloquants Avant et Après Chirurgie Cardiaque (BBAACC)
.
Anesthésie & Réanimation
 
2015
;
1
:
A248
.

201

Oesterle
 
A
,
Weber
 
B
,
Tung
 
R
,
Choudhry
 
NK
,
Singh
 
JP
,
Upadhyay
 
GA
.
Preventing postoperative atrial fibrillation after noncardiac surgery: a meta-analysis
.
Am J Med
 
2018
;
131
:
795
804 e795
.

202

Aoki
 
Y
,
Kawasaki
 
Y
,
Ide
 
K
,
Shimizu
 
Y
,
Sato
 
S
,
Yokoyama
 
J
.
Landiolol hydrochloride for prevention of atrial fibrillation during esophagectomy: a randomized controlled trial
.
JA Clin Rep
 
2020
;
6
:
34
.

203

Aoyama
 
H
,
Otsuka
 
Y
,
Aoyama
 
Y
.
Landiolol infusion during general anesthesia does not prevent postoperative atrial fibrillation in patients undergoing lung resection
.
Gen Thorac Cardiovasc Surg
 
2016
;
64
:
735
741
.

204

Ojima
 
T
,
Nakamori
 
M
,
Nakamura
 
M
,
Katsuda
 
M
,
Hayata
 
K
,
Kato
 
T
, et al.  
Randomized clinical trial of landiolol hydrochloride for the prevention of atrial fibrillation and postoperative complications after oesophagectomy for cancer
.
Br J Surg
 
2017
;
104
:
1003
1009
.

205

Horikoshi
 
Y
,
Goyagi
 
T
,
Kudo
 
R
,
Kodama
 
S
,
Horiguchi
 
T
,
Nishikawa
 
T
.
The suppressive effects of landiolol administration on the occurrence of postoperative atrial fibrillation and tachycardia, and plasma IL-6 elevation in patients undergoing esophageal surgery: a randomized controlled clinical trial
.
J Clin Anesth
 
2017
;
38
:
111
116
.

206

O'Brien
 
B
,
Burrage
 
PS
,
Ngai
 
JY
,
Prutkin
 
JM
,
Huang
 
CC
,
Xu
 
X
, et al.  
Society of Cardiovascular Anesthesiologists/European Association of Cardiothoracic Anaesthetists Practice Advisory for the management of perioperative atrial fibrillation in patients undergoing cardiac surgery
.
J Cardiothorac Vasc Anesth
 
2019
;
33
:
12
26
.

207

Zhu
 
J
,
Wang
 
C
,
Gao
 
D
,
Zhang
 
C
,
Zhang
 
Y
,
Lu
 
Y
, et al.  
Meta-analysis of amiodarone versus beta-blocker as a prophylactic therapy against atrial fibrillation following cardiac surgery
.
Intern Med J
 
2012
;
42
:
1078
1087
.

208

Auer
 
J
,
Weber
 
T
,
Berent
 
R
,
Puschmann
 
R
,
Hartl
 
P
,
Ng
 
CK
, et al.  
A comparison between oral antiarrhythmic drugs in the prevention of atrial fibrillation after cardiac surgery: the pilot Study of Prevention of Postoperative Atrial Fibrillation (SPPAF), a randomized, placebo-controlled trial
.
Am Heart J
 
2004
;
147
:
636
643
.

209

London
 
MJ
,
Schwartz
 
GG
,
Hur
 
K
,
Henderson
 
WG
.
Association of perioperative statin use with mortality and morbidity after major noncardiac surgery
.
JAMA Intern Med
 
2017
;
177
:
231
242
.

210

Lindenauer
 
PK
,
Pekow
 
P
,
Wang
 
K
,
Gutierrez
 
B
,
Benjamin
 
EM
.
Lipid-lowering therapy and in-hospital mortality following major noncardiac surgery
.
JAMA
 
2004
;
291
:
2092
2099
.

211

Berwanger
 
O
,
de Barros
 
ESPG
,
Barbosa
 
RR
,
Precoma
 
DB
,
Figueiredo
 
EL
,
Hajjar
 
LA
, et al.  
Atorvastatin for high-risk statin-naive patients undergoing noncardiac surgery: the Lowering the Risk of Operative Complications Using Atorvastatin Loading Dose (LOAD) randomized trial
.
Am Heart J
 
2017
;
184
:
88
96
.

212

Sanders
 
RD
,
Nicholson
 
A
,
Lewis
 
SR
,
Smith
 
AF
,
Alderson
 
P
.
Perioperative statin therapy for improving outcomes during and after noncardiac vascular surgery
.
Cochrane Database Syst Rev
 
2013
;
7
:
CD009971
.

213

Putzu
 
A
,
de Carvalho
 
ESC
,
de Almeida
 
JP
,
Belletti
 
A
,
Cassina
 
T
,
Landoni
 
G
, et al.  
Perioperative statin therapy in cardiac and non-cardiac surgery: a systematic review and meta-analysis of randomized controlled trials
.
Ann Intensive Care
 
2018
;
8
:
95
.

214

Wesselink
 
EM
,
Kappen
 
TH
,
Torn
 
HM
,
Slooter
 
AJC
,
van Klei
 
WA
.
Intraoperative hypotension and the risk of postoperative adverse outcomes: a systematic review
.
Br J Anaesth
 
2018
;
121
:
706
721
.

215

Shiffermiller
 
JF
,
Monson
 
BJ
,
Vokoun
 
CW
,
Beachy
 
MW
,
Smith
 
MP
,
Sullivan
 
JN
, et al.  
Prospective Randomized Evaluation of Preoperative Angiotensin-Converting Enzyme Inhibition (PREOP-ACEI)
.
J Hosp Med
 
2018
;
13
:
661
667
.

216

Roshanov
 
PS
,
Rochwerg
 
B
,
Patel
 
A
,
Salehian
 
O
,
Duceppe
 
E
,
Belley-Cote
 
EP
, et al.  
Withholding versus continuing angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers before noncardiac surgery: an analysis of the Vascular events In noncardiac Surgery patIents cOhort evaluatioN Prospective Cohort
.
Anesthesiology
 
2017
;
126
:
16
27
.

217

Hollmann
 
C
,
Fernandes
 
NL
,
Biccard
 
BM
.
A systematic review of outcomes associated with withholding or continuing angiotensin-converting enzyme inhibitors and angiotensin receptor blockers before noncardiac surgery
.
Anesth Analg
 
2018
;
127
:
678
687
.

218

McMurray
 
JJ
,
Packer
 
M
,
Desai
 
AS
,
Gong
 
J
,
Lefkowitz
 
MP
,
Rizkala
 
AR
, et al.  
Angiotensin-neprilysin inhibition versus enalapril in heart failure
.
N Engl J Med
 
2014
;
371
:
993
1004
.

219

Legrand
 
M
,
Futier
 
E
,
Leone
 
M
,
Deniau
 
B
,
Mebazaa
 
A
,
Plaud
 
B
, et al.  
Impact of renin-angiotensin system inhibitors continuation versus discontinuation on outcome after major surgery: protocol of a multicenter randomized, controlled trial (STOP-or-NOT trial)
.
Trials
 
2019
;
20
:
1
7
.

220

Wijeysundera
 
DN
,
Beattie
 
WS
.
Calcium channel blockers for reducing cardiac morbidity after noncardiac surgery: a meta-analysis
.
Anesth Analg
 
2003
;
97
:
634
641
.

221

Kertai
 
MD
,
Westerhout
 
CM
,
Varga
 
KS
,
Acsady
 
G
,
Gal
 
J
.
Dihydropiridine calcium-channel blockers and perioperative mortality in aortic aneurysm surgery
.
Br J Anaesth
 
2008
;
101
:
458
465
.

222

Oliver
 
MF
,
Goldman
 
L
,
Julian
 
DG
,
Holme
 
I
.
Effect of mivazerol on perioperative cardiac complications during non-cardiac surgery in patients with coronary heart disease: the European Mivazerol Trial (EMIT)
.
Anesthesiology
 
1999
;
91
:
951
961
.

223

Devereaux
 
PJ
,
Sessler
 
DI
,
Leslie
 
K
,
Kurz
 
A
,
Mrkobrada
 
M
,
Alonso-Coello
 
P
, et al.  
Clonidine in patients undergoing noncardiac surgery
.
N Engl J Med
 
2014
;
370
:
1504
1513
.

224

Hager
 
B
,
Betschart
 
M
,
Krapf
 
R
.
Effect of postoperative intravenous loop diuretic on renal function after major surgery
.
Schweiz Med Wochenschr
 
1996
;
126
:
666
673
.

225

Arora
 
P
,
Pourafkari
 
L
,
Visnjevac
 
O
,
Anand
 
EJ
,
Porhomayon
 
J
,
Nader
 
ND
.
Preoperative serum potassium predicts the clinical outcome after non-cardiac surgery
.
Clin Chem Lab Med
 
2017
;
55
:
145
153
.

226

Tagawa
 
M
,
Ogata
 
A
,
Hamano
 
T
.
Pre- and/or intra-operative prescription of diuretics, but not renin-angiotensin-system inhibitors, is significantly associated with acute kidney injury after non-cardiac surgery: a retrospective cohort study
.
PLoS One
 
2015
;
10
:
e0132507
.

227

Lo Sapio
 
P
,
Gensini
 
GF
,
Bevilacqua
 
S
,
Chiti
 
E
,
Paperetti
 
L
,
Pratesi
 
C
, et al.  
The role of ivabradine in the incidence of perioperative coronary complications in patients undergoing vascular surgery
.
Int J Cardiol
 
2013
;
168
:
4352
4353
.

228

Thiruvenkatarajan
 
V
,
Meyer
 
EJ
,
Nanjappa
 
N
,
Van Wijk
 
RM
,
Jesudason
 
D
.
Perioperative diabetic ketoacidosis associated with sodium-glucose co-transporter-2 inhibitors: a systematic review
.
Br J Anaesth
 
2019
;
123
:
27
36
.

229

Patoulias
 
D
,
Manafis
 
A
,
Mitas
 
C
,
Avranas
 
K
,
Lales
 
G
,
Zografou
 
I
, et al.  
Sodium-glucose cotransporter 2 inhibitors and the risk of diabetic ketoacidosis; from pathophysiology to clinical practice
.
Cardiovasc Hematol Disord Drug Targets
 
2018
;
18
:
139
146
.

230

Mangano
 
DT
,
Layug
 
EL
,
Wallace
 
A
,
Tateo
 
I
.
Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. Multicenter Study of Perioperative Ischemia Research Group
.
N Engl J Med
 
1996
;
335
:
1713
1720
.

231

McGory
 
ML
,
Maggard
 
MA
,
Ko
 
CY
.
A meta-analysis of perioperative beta blockade: what is the actual risk reduction?
 
Surgery
 
2005
;
138
:
171
179
.

232

Wallace
 
A
,
Layug
 
B
,
Tateo
 
I
,
Li
 
J
,
Hollenberg
 
M
,
Browner
 
W
, et al.  
Prophylactic atenolol reduces postoperative myocardial ischemia. McSPI Research Group
.
Anesthesiology
 
1998
;
88
:
7
17
.

233

Bouri
 
S
,
Shun-Shin
 
MJ
,
Cole
 
GD
,
Mayet
 
J
,
Francis
 
DP
.
Meta-analysis of secure randomised controlled trials of beta-blockade to prevent perioperative death in non-cardiac surgery
.
Heart
 
2014
;
100
:
456
464
.

234

Hajibandeh
 
S
,
Hajibandeh
 
S
,
Antoniou
 
SA
,
Torella
 
F
,
Antoniou
 
GA
.
Effect of beta-blockers on perioperative outcomes in vascular and endovascular surgery: a systematic review and meta-analysis
.
Br J Anaesth
 
2017
;
118
:
11
21
.

235

Xia
 
J
,
Qu
 
Y
,
Yin
 
C
,
Xu
 
D
.
Preoperative rosuvastatin protects patients with coronary artery disease undergoing noncardiac surgery
.
Cardiology
 
2015
;
131
:
30
37
.

236

McEvoy
 
MD
,
Gupta
 
R
,
Koepke
 
EJ
,
Feldheiser
 
A
,
Michard
 
F
,
Levett
 
D
, et al.  
Perioperative Quality Initiative Consensus Statement on postoperative blood pressure, risk and outcomes for elective surgery
.
Br J Anaesth
 
2019
;
122
:
575
586
.

237

Ellenberger
 
C
,
Tait
 
G
,
Beattie
 
WS
.
Chronic beta blockade is associated with a better outcome after elective noncardiac surgery than acute beta blockade: a single-center propensity-matched cohort study
.
Anesthesiology
 
2011
;
114
:
817
823
.

238

Zaugg
 
M
,
Bestmann
 
L
,
Wacker
 
J
,
Lucchinetti
 
E
,
Boltres
 
A
,
Schulz
 
C
, et al.  
Adrenergic receptor genotype but not perioperative bisoprolol therapy may determine cardiovascular outcome in at-risk patients undergoing surgery with spinal block: the Swiss Beta Blocker in Spinal Anesthesia (BBSA) study: a double-blinded, placebo-controlled, multicenter trial with 1-year follow-up
.
Anesthesiology
 
2007
;
107
:
33
44
.

239

Faloye
 
AO
,
Gebre
 
MA
,
Bechtel
 
AJ
.
Predicting cardiac risk in noncardiac surgery: a narrative review
.
J Anesth
 
2021
;
35
:
122
129
.

240

Steffel
 
J
,
Collins
 
R
,
Antz
 
M
,
Cornu
 
P
,
Desteghe
 
L
,
Haeusler
 
KG
, et al.  
2021 European Heart Rhythm Association Practical Guide on the use of non-vitamin k antagonist oral anticoagulants in patients with atrial fibrillation
.
Europace
 
2021
;
23
:
1612
1676
.

241

Zheng
 
SL
,
Roddick
 
AJ
.
Association of aspirin use for primary prevention with cardiovascular events and bleeding events: a systematic review and meta-analysis
.
JAMA
 
2019
;
321
:
277
287
.

242

Piepoli
 
MF
,
Hoes
 
AW
,
Agewall
 
S
,
Albus
 
C
,
Brotons
 
C
,
Catapano
 
AL
, et al.  
2016 European Guidelines on cardiovascular disease prevention in clinical practice: the Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts) developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR)
.
Eur Heart J
 
2016
;
37
:
2315
2381
.

243

Devereaux
 
PJ
,
Mrkobrada
 
M
,
Sessler
 
DI
,
Leslie
 
K
,
Alonso-Coello
 
P
,
Kurz
 
A
, et al.  
Aspirin in patients undergoing noncardiac surgery
.
N Engl J Med
 
2014
;
370
:
1494
1503
.

244

Graham
 
MM
,
Sessler
 
DI
,
Parlow
 
JL
,
Biccard
 
BM
,
Guyatt
 
G
,
Leslie
 
K
, et al.  
Aspirin in patients with previous percutaneous coronary intervention undergoing noncardiac surgery
.
Ann Intern Med
 
2018
;
168
:
237
244
.

245

Vahanian
 
A
,
Beyersdorf
 
F
,
Praz
 
F
,
Milojevic
 
M
,
Baldus
 
S
,
Bauersachs
 
J
, et al.  
2021 ESC/EACTS Guidelines for the management of valvular heart disease
.
Eur Heart J
 
2022
;
43
:
561
632
.

246

Brouwer
 
J
,
Nijenhuis
 
VJ
,
Delewi
 
R
,
Hermanides
 
RS
,
Holvoet
 
W
,
Dubois
 
CLF
, et al.  
Aspirin with or without clopidogrel after transcatheter aortic-valve implantation
.
N Engl J Med
 
2020
;
383
:
1447
1457
.

247

Koo
 
BK
,
Kang
 
J
,
Park
 
KW
,
Rhee
 
TM
,
Yang
 
HM
,
Won
 
KB
, et al.  
Aspirin versus clopidogrel for chronic maintenance monotherapy after percutaneous coronary intervention (HOST-EXAM): an investigator-initiated, prospective, randomised, open-label, multicentre trial
.
Lancet
 
2021
;
397
:
2487
2496
.

248

Mehran
 
R
,
Baber
 
U
,
Sharma
 
SK
,
Cohen
 
DJ
,
Angiolillo
 
DJ
,
Briguori
 
C
, et al.  
Ticagrelor with or without aspirin in high-risk patients after PCI
.
N Engl J Med
 
2019
;
381
:
2032
2042
.

249

Vranckx
 
P
,
Valgimigli
 
M
,
Juni
 
P
,
Hamm
 
C
,
Steg
 
PG
,
Heg
 
D
, et al.  
Ticagrelor plus aspirin for 1 month, followed by ticagrelor monotherapy for 23 months vs aspirin plus clopidogrel or ticagrelor for 12 months, followed by aspirin monotherapy for 12 months after implantation of a drug-eluting stent: a multicentre, open-label, randomised superiority trial
.
Lancet
 
2018
;
392
:
940
949
.

250

Watanabe
 
H
,
Domei
 
T
,
Morimoto
 
T
,
Natsuaki
 
M
,
Shiomi
 
H
,
Toyota
 
T
, et al.  
Effect of 1-month dual antiplatelet therapy followed by clopidogrel vs 12-month dual antiplatelet therapy on cardiovascular and bleeding events in patients receiving PCI: the STOPDAPT-2 randomized clinical trial
.
JAMA
 
2019
;
321
:
2414
2427
.

251

Berger
 
PB
,
Kleiman
 
NS
,
Pencina
 
MJ
,
Hsieh
 
WH
,
Steinhubl
 
SR
,
Jeremias
 
A
, et al.  
Frequency of major noncardiac surgery and subsequent adverse events in the year after drug-eluting stent placement results from the EVENT (Evaluation of Drug-Eluting Stents and Ischemic Events) Registry
.
JACC Cardiovasc Interv
 
2010
;
3
:
920
927
.

252

Saia
 
F
,
Belotti
 
LMB
,
Guastaroba
 
P
,
Berardini
 
A
,
Rossini
 
R
,
Musumeci
 
G
, et al.  
Risk of adverse cardiac and bleeding events following cardiac and noncardiac surgery in patients with coronary stent: how important is the interplay between stent type and time from stenting to surgery?
 
Circ Cardiovasc Qual Outcomes
 
2016
;
9
:
39
47
.

253

Huber
 
KC
,
Evans
 
MA
,
Bresnahan
 
JF
,
Gibbons
 
RJ
,
Holmes
 
DR
, Jr
.
Outcome of noncardiac operations in patients with severe coronary artery disease successfully treated preoperatively with coronary angioplasty
.
Mayo Clin Proc
 
1992
;
67
:
15
21
.

254

Egholm
 
G
,
Kristensen
 
SD
,
Thim
 
T
,
Olesen
 
KK
,
Madsen
 
M
,
Jensen
 
SE
, et al.  
Risk associated with surgery within 12 months after coronary drug-eluting stent implantation
.
J Am Coll Cardiol
 
2016
;
68
:
2622
2632
.

255

Holcomb
 
CN
,
Graham
 
LA
,
Richman
 
JS
,
Itani
 
KMF
,
Maddox
 
TM
,
Hawn
 
MT
.
The incremental risk of coronary stents on postoperative adverse events: a matched cohort study
.
Ann Surg
 
2016
;
263
:
924
930
.

256

Mahmoud
 
KD
,
Sanon
 
S
,
Habermann
 
EB
,
Lennon
 
RJ
,
Thomsen
 
KM
,
Wood
 
DL
, et al.  
Perioperative cardiovascular risk of prior coronary stent implantation among patients undergoing noncardiac surgery
.
J Am Coll Cardiol
 
2016
;
67
:
1038
1049
.

257

Holcomb
 
CN
,
Hollis
 
RH
,
Graham
 
LA
,
Richman
 
JS
,
Valle
 
JA
,
Itani
 
KM
, et al.  
Association of coronary stent indication with postoperative outcomes following noncardiac surgery
.
JAMA Surg
 
2016
;
151
:
462
.

258

Rodriguez
 
A
,
Guilera
 
N
,
Mases
 
A
,
Sierra
 
P
,
Oliva
 
JC
,
Colilles
 
C
, et al.  
Management of antiplatelet therapy in patients with coronary stents undergoing noncardiac surgery: association with adverse events
.
Br J Anaesth
 
2018
;
120
:
67
76
.

259

Armstrong
 
EJ
,
Graham
 
L
,
Waldo
 
SW
,
Valle
 
JA
,
Maddox
 
TM
,
Hawn
 
MT
.
Patient and lesion-specific characteristics predict risk of major adverse cardiovascular events among patients with previous percutaneous coronary intervention undergoing noncardiac surgery
.
Catheter Cardiovasc Interv
 
2017
;
89
:
617
627
.

260

Siller-Matula
 
JM
,
Petre
 
A
,
Delle-Karth
 
G
,
Huber
 
K
,
Ay
 
C
,
Lordkipanidze
 
M
, et al.  
Impact of preoperative use of P2Y12 receptor inhibitors on clinical outcomes in cardiac and non-cardiac surgery: a systematic review and meta-analysis
.
Eur Heart J Acute Cardiovasc Care
 
2017
;
6
:
753
770
.

261

Mehran
 
R
,
Baber
 
U
,
Steg
 
PG
,
Ariti
 
C
,
Weisz
 
G
,
Witzenbichler
 
B
, et al.  
Cessation of dual antiplatelet treatment and cardiac events after percutaneous coronary intervention (PARIS): 2 year results from a prospective observational study
.
Lancet
 
2013
;
382
:
1714
1722
.

262

So
 
DYF
,
Wells
 
GA
,
Lordkipanidze
 
M
,
Ruel
 
M
,
Perrault
 
LP
,
Yeong Chong
 
A
, et al.  
A randomized study of early vs. delayed coronary artery bypass surgery among patients with acute coronary syndromes treated with ticagrelor: the RAPID CABG study
. http://clinicaltrialresults.org/wp-content/uploads/2021/11/So_RAPID_CABG.pdf.

263

Gurbel
 
PA
,
Bliden
 
KP
,
Butler
 
K
,
Tantry
 
US
,
Gesheff
 
T
,
Wei
 
C
, et al.  
Randomized double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET study
.
Circulation
 
2009
;
120
:
2577
2585
.

264

Valgimigli
 
M
,
Bueno
 
H
,
Byrne
 
RA
,
Collet
 
JP
,
Costa
 
F
,
Jeppsson
 
A
, et al.  
2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS: the Task Force for dual antiplatelet therapy in coronary artery disease of the European Society of Cardiology (ESC) and of the European Association for Cardio-Thoracic Surgery (EACTS)
.
Eur Heart J
 
2018
;
39
:
213
260
.

265

Valgimigli
 
M
,
Frigoli
 
E
,
Heg
 
D
,
Tijssen
 
J
,
Juni
 
P
,
Vranckx
 
P
, et al.  
Dual antiplatelet therapy after PCI in patients at high bleeding risk
.
N Engl J Med
 
2021
;
385
:
1643
1655
.

266

Urban
 
P
,
Meredith
 
IT
,
Abizaid
 
A
,
Pocock
 
SJ
,
Carrie
 
D
,
Naber
 
C
, et al.  
Polymer-free drug-coated coronary stents in patients at high bleeding risk
.
N Engl J Med
 
2015
;
373
:
2038
2047
.

267

Windecker
 
S
,
Latib
 
A
,
Kedhi
 
E
,
Kirtane
 
AJ
,
Kandzari
 
DE
,
Mehran
 
R
, et al.  
Polymer-based or polymer-free stents in patients at high bleeding risk
.
N Engl J Med
 
2020
;
382
:
1208
1218
.

268

Neumann
 
FJ
,
Sousa-Uva
 
M
,
Ahlsson
 
A
,
Alfonso
 
F
,
Banning
 
AP
,
Benedetto
 
U
, et al.  
2018 ESC/EACTS Guidelines on myocardial revascularization
.
Eur Heart J
 
2019
;
40
:
87
165
.

269

Watanabe
 
H
,
Morimoto
 
T
,
Natsuaki
 
M
,
Yamamoto
 
K
,
Obayashi
 
Y
,
Ogita
 
M
, et al.  
Comparison of Clopidogrel Monotherapy After 1 to 2 Months of Dual Antiplatelet Therapy with 12 Months of Dual Antiplatelet Therapy in Patients With Acute Coronary Syndrome: The STOPDAPT-2 ACS Randomized Clinical Trial
.
JAMA Cardiol
 
2022
;
7
:
407
417
.

270

Kim
 
BK
,
Hong
 
MK
,
Shin
 
DH
,
Nam
 
CM
,
Kim
 
JS
,
Ko
 
YG
, et al.  
A new strategy for discontinuation of dual antiplatelet therapy: the RESET Trial (REal Safety and Efficacy of 3-month dual antiplatelet Therapy following Endeavor zotarolimus-eluting stent implantation)
.
J Am Coll Cardiol
 
2012
;
60
:
1340
1348
.

271

Feres
 
F
,
Costa
 
RA
,
Abizaid
 
A
,
Leon
 
MB
,
Marin-Neto
 
JA
,
Botelho
 
RV
, et al.  
Three vs twelve months of dual antiplatelet therapy after zotarolimus-eluting stents: the OPTIMIZE randomized trial
.
JAMA
 
2013
;
310
:
2510
2522
.

272

Schulz-Schupke
 
S
,
Byrne
 
RA
,
Ten Berg
 
JM
,
Neumann
 
FJ
,
Han
 
Y
,
Adriaenssens
 
T
, et al.  
ISAR-SAFE: a randomized, double-blind, placebo-controlled trial of 6 vs. 12 months of clopidogrel therapy after drug-eluting stenting
.
Eur Heart J
 
2015
;
36
:
1252
1263
.

273

Gwon
 
HC
,
Hahn
 
JY
,
Park
 
KW
,
Song
 
YB
,
Chae
 
IH
,
Lim
 
DS
, et al.  
Six-month versus 12-month dual antiplatelet therapy after implantation of drug-eluting stents: the Efficacy of Xience/Promus Versus Cypher to Reduce Late Loss After Stenting (EXCELLENT) randomized, multicenter study
.
Circulation
 
2012
;
125
:
505
513
.

274

Valgimigli
 
M
,
Campo
 
G
,
Monti
 
M
,
Vranckx
 
P
,
Percoco
 
G
,
Tumscitz
 
C
, et al.  
Short- versus long-term duration of dual-antiplatelet therapy after coronary stenting: a randomized multicenter trial
.
Circulation
 
2012
;
125
:
2015
2026
.

275

Han
 
Y
,
Xu
 
B
,
Xu
 
K
,
Guan
 
C
,
Jing
 
Q
,
Zheng
 
Q
, et al.  
Six versus 12 months of dual antiplatelet therapy after implantation of biodegradable polymer sirolimus-eluting stent: randomized substudy of the I-LOVE-IT 2 Trial
.
Circ Cardiovasc Interv
 
2016
;
9
:
e003145
.

276

Hong
 
SJ
,
Shin
 
DH
,
Kim
 
JS
,
Kim
 
BK
,
Ko
 
YG
,
Choi
 
D
, et al.  
6-Month versus 12-month dual-antiplatelet therapy following long everolimus-eluting stent implantation: the IVUS-XPL randomized clinical trial
.
JACC Cardiovasc Interv
 
2016
;
9
:
1438
1446
.

277

Hahn
 
JY
,
Song
 
YB
,
Oh
 
JH
,
Cho
 
DK
,
Lee
 
JB
,
Doh
 
JH
, et al.  
6-month versus 12-month or longer dual antiplatelet therapy after percutaneous coronary intervention in patients with acute coronary syndrome (SMART-DATE): a randomised, open-label, non-inferiority trial
.
Lancet
 
2018
;
391
:
1274
1284
.

278

Kedhi
 
E
,
Fabris
 
E
,
van der Ent
 
M
,
Buszman
 
P
,
von Birgelen
 
C
,
Roolvink
 
V
, et al.  
Six months versus 12 months dual antiplatelet therapy after drug-eluting stent implantation in ST-elevation myocardial infarction (DAPT-STEMI): randomised, multicentre, non-inferiority trial
.
BMJ
 
2018
;
363
:
k3793
.

279

Wallentin
 
L
,
Becker
 
RC
,
Budaj
 
A
,
Cannon
 
CP
,
Emanuelsson
 
H
,
Held
 
C
, et al.  
Ticagrelor versus clopidogrel in patients with acute coronary syndromes
.
N Engl J Med
 
2009
;
361
:
1045
1057
.

280

Wiviott
 
SD
,
Braunwald
 
E
,
McCabe
 
CH
,
Montalescot
 
G
,
Ruzyllo
 
W
,
Gottlieb
 
S
, et al.  
Prasugrel versus clopidogrel in patients with acute coronary syndromes
.
N Engl J Med
 
2007
;
357
:
2001
2015
.

281

Yusuf
 
S
,
Zhao
 
F
,
Mehta
 
SR
,
Chrolavicius
 
S
,
Tognoni
 
G
,
Fox
 
KK
, et al.  
Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation
.
N Engl J Med
 
2001
;
345
:
494
502
.

282

Sullivan
 
AE
,
Nanna
 
MG
,
Wang
 
TY
,
Bhatt
 
DL
,
Angiolillo
 
DJ
,
Mehran
 
R
, et al.  
Bridging antiplatelet therapy after percutaneous coronary intervention: JACC review topic of the week
.
J Am Coll Cardiol
 
2021
;
78
:
1550
1563
.

283

Angheloiu
 
GO
,
Gugiu
 
GB
,
Ruse
 
C
,
Pandey
 
R
,
Dasari
 
RR
,
Whatling
 
C
.
Ticagrelor removal from human blood
.
JACC Basic Transl Sci
 
2017
;
2
:
135
145
.

284

Bhatt
 
DL
,
Pollack
 
CV
,
Weitz
 
JI
,
Jennings
 
LK
,
Xu
 
S
,
Arnold
 
SE
, et al.  
Antibody-based ticagrelor reversal agent in healthy volunteers
.
N Engl J Med
 
2019
;
380
:
1825
1833
.

285

Mahla
 
E
,
Metzler
 
H
,
Bornemann-Cimenti
 
H
,
Prueller
 
F
,
Raggam
 
R
,
Pregartner
 
G
, et al.  
Platelet inhibition and bleeding in patients undergoing non-cardiac surgery—the BIANCA Observational Study
.
Thromb Haemost
 
2018
;
118
:
864
872
.

286

Mahla
 
E
,
Tantry
 
US
,
Pruller
 
F
,
Gurbel
 
PA
.
Is there a role for preoperative platelet function testing in patients undergoing cardiac surgery during antiplatelet therapy?
 
Circulation
 
2018
;
138
:
2145
2159
.

287

Mahla
 
E
,
Suarez
 
TA
,
Bliden
 
KP
,
Rehak
 
P
,
Metzler
 
H
,
Sequeira
 
AJ
, et al.  
Platelet function measurement-based strategy to reduce bleeding and waiting time in clopidogrel-treated patients undergoing coronary artery bypass graft surgery: the timing based on platelet function strategy to reduce clopidogrel-associated bleeding related to CABG (TARGET-CABG) study
.
Circ Cardiovasc Interv
 
2012
;
5
:
261
269
.

288

Windecker
 
S
,
Lopes
 
RD
,
Massaro
 
T
,
Jones-Burton
 
C
,
Granger
 
CB
,
Aronson
 
R
, et al.  
Antithrombotic therapy in patients with atrial fibrillation and acute coronary syndrome treated medically or with percutaneous coronary intervention or undergoing elective percutaneous coronary intervention: insights from the AUGUSTUS trial
.
Circulation
 
2019
;
140
:
1921
1932
.

289

Genereux
 
P
,
Rutledge
 
DR
,
Palmerini
 
T
,
Caixeta
 
A
,
Kedhi
 
E
,
Hermiller
 
JB
, et al.  
Stent thrombosis and dual antiplatelet therapy interruption with everolimus-eluting stents: insights from the Xience V Coronary Stent System Trials
.
Circ Cardiovasc Interv
 
2015
;
8
:
e001362
.

290

Beyer-Westendorf
 
J
,
Gelbricht
 
V
,
Forster
 
K
,
Ebertz
 
F
,
Kohler
 
C
,
Werth
 
S
, et al.  
Peri-interventional management of novel oral anticoagulants in daily care: results from the prospective Dresden NOAC registry
.
Eur Heart J
 
2014
;
35
:
1888
1896
.

291

Otto
 
CM
,
Nishimura
 
RA
,
Bonow
 
RO
,
Carabello
 
BA
,
Erwin
 
JP
, 3rd
,
Gentile
 
F
, et al.  
2020 ACC/AHA Guideline for the management of patients with valvular heart disease. Executive summary: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines
.
Circulation
 
2021
;
143
:
e35
e71
.

292

Douketis
 
JD
,
Spyropoulos
 
AC
,
Kaatz
 
S
,
Becker
 
RC
,
Caprini
 
JA
,
Dunn
 
AS
, et al.  
Perioperative bridging anticoagulation in patients with atrial fibrillation
.
N Engl J Med
 
2015
;
373
:
823
833
.

293

Kuo
 
HC
,
Liu
 
FL
,
Chen
 
JT
,
Cherng
 
YG
,
Tam
 
KW
,
Tai
 
YH
.
Thromboembolic and bleeding risk of periprocedural bridging anticoagulation: a systematic review and meta-analysis
.
Clin Cardiol
 
2020
;
43
:
441
449
.

294

Kovacs
 
MJ
,
Wells
 
PS
,
Anderson
 
DR
,
Lazo-Langner
 
A
,
Kearon
 
C
,
Bates
 
SM
, et al.  
Postoperative low molecular weight heparin bridging treatment for patients at high risk of arterial thromboembolism (PERIOP2): double blind randomised controlled trial
.
BMJ
 
2021
;
373
:
n1205
.

295

Caldeira
 
D
,
David
 
C
,
Santos
 
AT
,
Costa
 
J
,
Pinto
 
FJ
,
Ferreira
 
JJ
.
Efficacy and safety of low molecular weight heparin in patients with mechanical heart valves: systematic review and meta-analysis
.
J Thromb Haemost
 
2014
;
12
:
650
659
.

296

Birnie
 
DH
,
Healey
 
JS
,
Wells
 
GA
,
Verma
 
A
,
Tang
 
AS
,
Krahn
 
AD
, et al.  
Pacemaker or defibrillator surgery without interruption of anticoagulation
.
N Engl J Med
 
2013
;
368
:
2084
2093
.

297

Sant'anna
 
RT
,
Leiria
 
TL
,
Nascimento
 
T
,
Sant'anna
 
JR
,
Kalil
 
RA
,
Lima
 
GG
, et al.  
Meta-analysis of continuous oral anticoagulants versus heparin bridging in patients undergoing CIED surgery: reappraisal after the BRUISE study
.
Pacing Clin Electrophysiol
 
2015
;
38
:
417
423
.

298

Eichhorn
 
W
,
Barsukov
 
E
,
Al-Dam
 
A
,
Gröbe
 
A
,
Smeets
 
R
,
Eichhorn
 
M
, et al.  
Postoperative bleeding risk for cutaneous surgery in the head and neck region with continued phenprocoumon therapy
.
J Cranio-Maxillofac Surg
 
2014
;
42
:
608
611
.

299

Nandoe Tewarie
 
RDS
,
Bartels
 
RHMA
.
The perioperative use of oral anticoagulants during surgical procedures for carpal tunnel syndrome. A preliminary study
.
Acta Neurochirurgica
 
2010
;
152
:
1211
1213
.

300

Doherty
 
JU
,
Gluckman
 
TJ
,
Hucker
 
WJ
,
Januzzi
 
JL
, Jr.
,
Ortel
 
TL
,
Saxonhouse
 
SJ
, et al.  
2017 ACC Expert Consensus Decision Pathway for periprocedural management of anticoagulation in patients with nonvalvular atrial fibrillation: a report of the American College of Cardiology Clinical Expert Consensus Document Task Force
.
J Am Coll Cardiol
 
2017
;
69
:
871
898
.

301

Chai-Adisaksopha
 
C
,
Hillis
 
C
,
Siegal
 
DM
,
Movilla
 
R
,
Heddle
 
N
,
Iorio
 
A
, et al.  
Prothrombin complex concentrates versus fresh frozen plasma for warfarin reversal: a systematic review and meta-analysis
.
Thromb Haemost
 
2016
;
116
:
879
890
.

302

Levy
 
JH
,
Ageno
 
W
,
Chan
 
NC
,
Crowther
 
M
,
Verhamme
 
P
,
Weitz
 
JI
, et al.  
When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH
.
J Thromb Haemost
 
2016
;
14
:
623
627
.

303

Pollack
 
CV
, Jr.,
Reilly
 
PA
,
van Ryn
 
J
,
Eikelboom
 
JW
,
Glund
 
S
,
Bernstein
 
RA
, et al.  
Idarucizumab for dabigatran reversal - full cohort analysis
.
N Engl J Med
 
2017
;
377
:
431
441
.

304

Connolly
 
SJ
,
Crowther
 
M
,
Eikelboom
 
JW
,
Gibson
 
CM
,
Curnutte
 
JT
,
Lawrence
 
JH
, et al.  
Full study report of andexanet alfa for bleeding associated with factor Xa inhibitors
.
N Engl J Med
 
2019
;
380
:
1326
1335
.

305

Majeed
 
A
,
Ågren
 
A
,
Holmström
 
M
,
Bruzelius
 
M
,
Chaireti
 
R
,
Odeberg
 
J
, et al.  
Management of rivaroxaban- or apixaban-associated major bleeding with prothrombin complex concentrates: a cohort study
.
Blood
 
2017
;
130
:
1706
1712
.

306

Healey
 
JS
,
Eikelboom
 
J
,
Douketis
 
J
,
Wallentin
 
L
,
Oldgren
 
J
,
Yang
 
S
, et al.  
Periprocedural bleeding and thromboembolic events with dabigatran compared with warfarin: results from the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) Randomized Trial
.
Circulation
 
2012
;
126
:
343
348
.

307

Garcia
 
D
,
Alexander
 
JH
,
Wallentin
 
L
,
Wojdyla
 
DM
,
Thomas
 
L
,
Hanna
 
M
, et al.  
Management and clinical outcomes in patients treated with apixaban vs warfarin undergoing procedures
.
Blood
 
2014
;
124
:
3692
3698
.

308

Sherwood
 
MW
,
Douketis
 
JD
,
Patel
 
MR
,
Piccini
 
JP
,
Hellkamp
 
AS
,
Lokhnygina
 
Y
, et al.  
Outcomes of temporary interruption of rivaroxaban compared with warfarin in patients with nonvalvular atrial fibrillation: results from the rivaroxaban once daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation (ROCKET AF)
.
Circulation
 
2014
;
129
:
1850
1859
.

309

Shaw
 
JR
,
Li
 
N
,
Vanassche
 
T
,
Coppens
 
M
,
Spyropoulos
 
AC
,
Syed
 
S
, et al.  
Predictors of preprocedural direct oral anticoagulant levels in patients having an elective surgery or procedure
.
Blood Advances
 
2020
;
4
:
3520
3527
.

310

Godier
 
A
,
Dincq
 
AS
,
Martin
 
AC
,
Radu
 
A
,
Leblanc
 
I
,
Antona
 
M
, et al.  
Predictors of pre-procedural concentrations of direct oral anticoagulants: a prospective multicentre study
.
Eur Heart J
 
2017
;
38
:
2431
2439
.

311

Douketis
 
JD
,
Spyropoulos
 
AC
,
Duncan
 
J
,
Carrier
 
M
,
Le Gal
 
G
,
Tafur
 
AJ
, et al.  
Perioperative management of patients with atrial fibrillation receiving a direct oral anticoagulant
.
JAMA Intern Med
 
2019
;
179
:
1469
1478
.

312

Colonna
 
P
,
von Heymann
 
C
,
Santamaria
 
A
,
Saxena
 
M
,
Vanassche
 
T
,
Wolpert
 
D
, et al.  
Routine clinical practice in the periprocedural management of edoxaban therapy is associated with low risk of bleeding and thromboembolic complications: the prospective, observational, and multinational EMIT-AF/VTE study
.
Clin Cardiol
 
2020
;
43
:
769
780
.

313

Albaladejo
 
P
,
Pernod
 
G
,
Godier
 
A
,
de Maistre
 
E
,
Rosencher
 
N
,
Mas
 
JL
, et al.  
Management of bleeding and emergency invasive procedures in patients on dabigatran: updated guidelines from the French Working Group on Perioperative Haemostasis (GIHP) – September 2016
.
Anaesth Crit Care Pain Med
 
2018
;
37
:
391
399
.

314

Narouze
 
S
,
Benzon
 
HT
,
Provenzano
 
DA
,
Buvanendran
 
A
,
De Andres
 
J
,
Deer
 
TR
, et al.  
Interventional spine and pain procedures in patients on antiplatelet and anticoagulant medications: guidelines from the American Society of Regional Anesthesia and Pain Medicine, the European Society of Regional Anaesthesia and Pain Therapy, the American Academy of Pain Medicine, the International Neuromodulation Society, the North American Neuromodulation Society, and the World Institute of Pain
.
Reg Anesth Pain Med
 
2015
;
40
:
182
212
.

315

Johnston
 
S
.
An evidence summary of the management of patients taking direct oral anticoagulants (DOACs) undergoing dental surgery
.
Int J Oral Maxillofac Surg
 
2016
;
45
:
618
630
.

316

Sivolella
 
S
,
De Biagi
 
M
,
Brunello
 
G
,
Berengo
 
M
,
Pengo
 
V
.
Managing dentoalveolar surgical procedures in patients taking new oral anticoagulants
.
Odontology
 
2015
;
103
:
258
263
.

317

Ockerman
 
A
,
Vanhaverbeke
 
M
,
Miclotte
 
I
,
Belmans
 
A
,
Vanassche
 
T
,
Politis
 
C
, et al.  
Tranexamic acid to reduce bleeding after dental extraction in patients treated with non-vitamin K oral anticoagulants: design and rationale of the EXTRACT-NOAC trial
.
Br J Oral Maxillofac Surg
 
2019
;
57
:
1107
1112
.

318

Nijenhuis
 
VJ
,
Brouwer
 
J
,
Delewi
 
R
,
Hermanides
 
RS
,
Holvoet
 
W
,
Dubois
 
CLF
, et al.  
Anticoagulation with or without clopidogrel after transcatheter aortic-valve implantation
.
N Engl J Med
 
2020
;
382
:
1696
1707
.

319

Xu
 
K
,
Chan
 
NC
,
Ibrahim
 
Q
,
Kruger
 
P
,
Sinha
 
S
,
Bhagirath
 
V
, et al.  
Reduction in mortality following elective major hip and knee surgery: a systematic review and meta-analysis
.
Thromb Haemost
 
2019
;
119
:
668
674
.

320

Smilowitz
 
NR
,
Gupta
 
N
,
Guo
 
Y
,
Maldonado
 
TS
,
Eikelboom
 
JW
,
Goldhaber
 
SZ
, et al.  
Trends in perioperative venous thromboembolism associated with major noncardiac surgery
.
TH Open
 
2017
;
1
:
e82
e91
.

321

Klemen
 
ND
,
Feingold
 
PL
,
Hashimoto
 
B
,
Wang
 
M
,
Kleyman
 
S
,
Brackett
 
A
, et al.  
Mortality risk associated with venous thromboembolism: a systematic review and Bayesian meta-analysis
.
Lancet Haematol
 
2020
;
7
:
e583
e593
.

322

Gangireddy
 
C
,
Rectenwald
 
JR
,
Upchurch
 
GR
,
Wakefield
 
TW
,
Khuri
 
S
,
Henderson
 
WG
, et al.  
Risk factors and clinical impact of postoperative symptomatic venous thromboembolism
.
J Vasc Surg
 
2007
;
45
:
335
341
;
discussion 341–332
.

323

Caprini
 
JA
.
Risk assessment as a guide for the prevention of the many faces of venous thromboembolism
.
Am J Surg
 
2010
;
199
:
S3
S10
.

324

Obi
 
AT
,
Pannucci
 
CJ
,
Nackashi
 
A
,
Abdullah
 
N
,
Alvarez
 
R
,
Bahl
 
V
, et al.  
Validation of the Caprini venous thromboembolism risk assessment model in critically ill surgical patients
.
JAMA Surg
 
2015
;
150
:
941
948
.

325

Lobastov
 
K
,
Barinov
 
V
,
Schastlivtsev
 
I
,
Laberko
 
L
,
Rodoman
 
G
,
Boyarintsev
 
V
.
Validation of the Caprini risk assessment model for venous thromboembolism in high-risk surgical patients in the background of standard prophylaxis
.
J Vasc Surg Venous Lymphat Disord
 
2016
;
4
:
153
160
.

326

Hachey
 
KJ
,
Hewes
 
PD
,
Porter
 
LP
,
Ridyard
 
DG
,
Rosenkranz
 
P
,
McAneny
 
D
, et al.  
Caprini venous thromboembolism risk assessment permits selection for postdischarge prophylactic anticoagulation in patients with resectable lung cancer
.
J Thorac Cardiovasc Surg
 
2016
;
151
:
37
44 e31
.

327

Sterbling
 
HM
,
Rosen
 
AK
,
Hachey
 
KJ
,
Vellanki
 
NS
,
Hewes
 
PD
,
Rao
 
SR
, et al.  
Caprini risk model decreases venous thromboembolism rates in thoracic surgery cancer patients
.
Ann Thorac Surg
 
2018
;
105
:
879
885
.

328

Falck-Ytter
 
Y
,
Francis
 
CW
,
Johanson
 
NA
,
Curley
 
C
,
Dahl
 
OE
,
Schulman
 
S
, et al.  
Prevention of VTE in orthopedic surgery patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines
.
Chest
 
2012
;
141
:
e278S
e325S
.

329

Faraoni
 
D
,
Comes
 
RF
,
Geerts
 
W
,
Wiles
 
MD
,
ESA VTE Guidelines Task Force
.
European guidelines on perioperative venous thromboembolism prophylaxis: neurosurgery
.
Eur J Anaesthesiol
 
2018
;
35
:
90
95
.

330

Kozek-Langenecker
 
S
,
Fenger-Eriksen
 
C
,
Thienpont
 
E
,
Barauskas
 
G
,
ESA VTE Guidelines Task Force
.
European guidelines on perioperative venous thromboembolism prophylaxis: surgery in the elderly
.
Eur J Anaesthesiol
 
2018
;
35
:
116
122
.

331

Venclauskas
 
L
,
Maleckas
 
A
,
Arcelus
 
JI
,
ESA VTE Guidelines Task Force
.
European guidelines on perioperative venous thromboembolism prophylaxis: surgery in the obese patient
.
Eur J Anaesthesiol
 
2018
;
35
:
147
153
.

332

Afshari
 
A
,
Ageno
 
W
,
Ahmed
 
A
,
Duranteau
 
J
,
Faraoni
 
D
,
Kozek-Langenecker
 
S
, et al.  
European guidelines on perioperative venous thromboembolism prophylaxis: executive summary
.
Eur J Anaesthesiol
 
2018
;
35
:
77
83
.

333

Cimminiello
 
C
,
Prandoni
 
P
,
Agnelli
 
G
,
Di Minno
 
G
,
Polo Friz
 
H
,
Scaglione
 
F
, et al.  
Thromboprophylaxis with enoxaparin and direct oral anticoagulants in major orthopedic surgery and acutely ill medical patients: a meta-analysis
.
Intern Emerg Med
 
2017
;
12
:
1291
1305
.

334

Lassen
 
MR
,
Gallus
 
A
,
Raskob
 
GE
,
Pineo
 
G
,
Chen
 
D
,
Ramirez
 
LM
, et al.  
Apixaban versus enoxaparin for thromboprophylaxis after hip replacement
.
N Engl J Med
 
2010
;
363
:
2487
2498
.

335

Lassen
 
MR
,
Raskob
 
GE
,
Gallus
 
A
,
Pineo
 
G
,
Chen
 
D
,
Portman
 
RJ
.
Apixaban or enoxaparin for thromboprophylaxis after knee replacement
.
N Engl J Med
 
2009
;
361
:
594
604
.

336

Lassen
 
MR
,
Ageno
 
W
,
Borris
 
LC
,
Lieberman
 
JR
,
Rosencher
 
N
,
Bandel
 
TJ
, et al.  
Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty
.
N Engl J Med
 
2008
;
358
:
2776
2786
.

337

Eriksson
 
BI
,
Borris
 
LC
,
Friedman
 
RJ
,
Haas
 
S
,
Huisman
 
MV
,
Kakkar
 
AK
, et al.  
Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty
.
N Engl J Med
 
2008
;
358
:
2765
2775
.

338

Friedman
 
RJ
,
Dahl
 
OE
,
Rosencher
 
N
,
Caprini
 
JA
,
Kurth
 
AA
,
Francis
 
CW
, et al.  
Dabigatran versus enoxaparin for prevention of venous thromboembolism after hip or knee arthroplasty: a pooled analysis of three trials
.
Thromb Res
 
2010
;
126
:
175
182
.

339

Kawai
 
Y
,
Fuji
 
T
,
Fujita
 
S
,
Kimura
 
T
,
Ibusuki
 
K
,
Abe
 
K
, et al.  
Edoxaban versus enoxaparin for the prevention of venous thromboembolism after total knee or hip arthroplasty: pooled analysis of coagulation biomarkers and primary efficacy and safety endpoints from two phase 3 trials
.
Thromb J
 
2016
;
14
:
48
.

340

Petersen
 
PB
,
Kehlet
 
H
,
Jorgensen
 
CC
.
Lundbeck Foundation Centre for Fast-track, Hip, Group KRC. Safety of in-hospital only thromboprophylaxis after fast-track total hip and knee arthroplasty: a prospective follow-up study in 17,582 procedures
.
Thromb Haemost
 
2018
;
118
:
2152
2161
.

341

Wilson
 
DG
,
Poole
 
WE
,
Chauhan
 
SK
,
Rogers
 
BA
.
Systematic review of aspirin for thromboprophylaxis in modern elective total hip and knee arthroplasty
.
Bone Joint J
 
2016
;
98-B
:
1056
1061
.

342

Jenny
 
JY
,
Pabinger
 
I
,
Samama
 
CM
,
ESA VTE Guidelines Task Force
.
European guidelines on perioperative venous thromboembolism prophylaxis: aspirin
.
Eur J Anaesthesiol
 
2018
;
35
:
123
129
.

343

Anderson
 
DR
,
Dunbar
 
M
,
Murnaghan
 
J
,
Kahn
 
SR
,
Gross
 
P
,
Forsythe
 
M
, et al.  
Aspirin or rivaroxaban for VTE prophylaxis after hip or knee arthroplasty
.
N Engl J Med
 
2018
;
378
:
699
707
.

344

Cassidy
 
MR
,
Rosenkranz
 
P
,
McAneny
 
D
.
Reducing postoperative venous thromboembolism complications with a standardized risk-stratified prophylaxis protocol and mobilization program
.
J Am Coll Surg
 
2014
;
218
:
1095
1104
.

345

Musallam
 
KM
,
Tamim
 
HM
,
Richards
 
T
,
Spahn
 
DR
,
Rosendaal
 
FR
,
Habbal
 
A
, et al.  
Preoperative anaemia and postoperative outcomes in non-cardiac surgery: a retrospective cohort study
.
Lancet
 
2011
;
378
:
1396
1407
.

346

Baron
 
DM
,
Franchini
 
M
,
Goobie
 
SM
,
Javidroozi
 
M
,
Klein
 
AA
,
Lasocki
 
S
, et al.  
Patient blood management during the COVID-19 pandemic: a narrative review
.
Anaesthesia
 
2020
;
75
:
1105
1113
.

347

Munoz
 
M
,
Gomez-Ramirez
 
S
,
Campos
 
A
,
Ruiz
 
J
,
Liumbruno
 
GM
.
Pre-operative anaemia: prevalence, consequences and approaches to management
.
Blood Transfus
 
2015
;
13
:
370
379
.

348

von Heymann
 
C
,
Kaufner
 
L
,
Sander
 
M
,
Spies
 
C
,
Schmidt
 
K
,
Gombotz
 
H
, et al.  
Does the severity of preoperative anemia or blood transfusion have a stronger impact on long-term survival after cardiac surgery?
 
J Thorac Cardiovasc Surg
 
2016
;
152
:
1412
1420
.

349

Rossler
 
J
,
Schoenrath
 
F
,
Seifert
 
B
,
Kaserer
 
A
,
Spahn
 
GH
,
Falk
 
V
, et al.  
Iron deficiency is associated with higher mortality in patients undergoing cardiac surgery: a prospective study
.
Br J Anaesth
 
2020
;
124
:
25
34
.

350

Althoff
 
FC
,
Neb
 
H
,
Herrmann
 
E
,
Trentino
 
KM
,
Vernich
 
L
,
Fullenbach
 
C
, et al.  
Multimodal patient blood management program based on a three-pillar strategy: a systematic review and meta-analysis
.
Ann Surg
 
2019
;
269
:
794
804
.

351

Meybohm
 
P
,
Herrmann
 
E
,
Steinbicker
 
AU
,
Wittmann
 
M
,
Gruenewald
 
M
,
Fischer
 
D
, et al.  
Patient blood management is associated with a substantial reduction of red blood cell utilization and safe for patient's outcome: a prospective, multicenter cohort study with a noninferiority design
.
Ann Surg
 
2016
;
264
:
203
211
.

352

Frew
 
N
,
Alexander
 
D
,
Hood
 
J
,
Acornley
 
A
.
Impact of a blood management protocol on transfusion rates and outcomes following total hip and knee arthroplasty
.
Ann R Coll Surg Engl
 
2016
;
98
:
380
386
.

353

Leahy
 
MF
,
Hofmann
 
A
,
Towler
 
S
,
Trentino
 
KM
,
Burrows
 
SA
,
Swain
 
SG
, et al.  
Improved outcomes and reduced costs associated with a health-system-wide patient blood management program: a retrospective observational study in four major adult tertiary-care hospitals
.
Transfusion
 
2017
;
57
:
1347
1358
.

354

So-Osman
 
C
,
Nelissen
 
R
,
Koopman-van
 
GA
,
Kluyver
 
E
,
Pöll
 
R
,
Onstenk
 
R
, et al.  
Patient blood management in elective total hip- and knee-replacement surgery (Part 1): a randomized controlled trial on erythropoietin and blood salvage as transfusion alternatives using a restrictive transfusion policy in erythropoietin-eligible patients
.
Anesthesiology
 
2014
;
120
:
839
851
.

355

Theusinger
 
OM
,
Kind
 
SL
,
Seifert
 
B
,
Borgeat
 
l
,
Gerber
 
C
,
Spahn
 
DR
.
Patient blood management in orthopaedic surgery: a four-year follow-up of transfusion requirements and blood loss from 2008 to 2011 at the Balgrist University Hospital in Zurich, Switzerland
.
Blood Transfus
 
2014
;
12
:
195
203
.

356

Froessler
 
B
,
Palm
 
P
,
Weber
 
I
,
Hodyl
 
NA
,
Singh
 
R
,
Murphy
 
EM
.
The important role for intravenous iron in perioperative patient blood management in major abdominal surgery: a randomized controlled trial
.
Ann Surg
 
2016
;
264
:
41
46
.

357

Triphaus
 
C
,
Judd
 
L
,
Glaser
 
P
,
Goehring
 
MH
,
Schmitt
 
E
,
Westphal
 
S
, et al.  
Effectiveness of preoperative iron supplementation in major surgical patients with iron deficiency: a prospective observational study
.
Ann Surg
 
2021
;
274
:
e212
e219
.

358

Cappellini
 
MD
,
Comin-Colet
 
J
,
de Francisco
 
A
,
Dignass
 
A
,
Doehner
 
W
,
Lam
 
CS
, et al.  
Iron deficiency across chronic inflammatory conditions: international expert opinion on definition, diagnosis, and management
.
Am J Hematol
 
2017
;
92
:
1068
1078
.

359

Auerbach
 
M
,
Macdougall
 
IC
.
Safety of intravenous iron formulations: facts and folklore
.
Blood Transfus
 
2014
;
12
:
296
300
.

360

Richards
 
T
,
Baikady
 
RR
,
Clevenger
 
B
,
Butcher
 
A
,
Abeysiri
 
S
,
Chau
 
M
, et al.  
Preoperative intravenous iron to treat anaemia before major abdominal surgery (PREVENTT): a randomised, double-blind, controlled trial
.
Lancet
 
2020
;
396
:
1353
1361
.

361

Kaufner
 
L
,
von Heymann
 
C
,
Henkelmann
 
A
,
Pace
 
NL
,
Weibel
 
S
,
Kranke
 
P
, et al.  
Erythropoietin plus iron versus control treatment including placebo or iron for preoperative anaemic adults undergoing non-cardiac surgery
.
Cochrane Database Syst Rev
 
2020
;
8
:
CD012451
.

362

Munoz
 
M
,
Acheson
 
AG
,
Auerbach
 
M
,
Besser
 
M
,
Habler
 
O
,
Kehlet
 
H
, et al.  
International consensus statement on the peri-operative management of anaemia and iron deficiency
.
Anaesthesia
 
2017
;
72
:
233
247
.

363

World Health Assembly
.
Availability, safety and quality of blood products
. http://apps.who.int/gb/ebwha/pdf_files/WHA63/A63_R12-en.pdf  
(27 October 2017)
.

364

Kotze
 
A
,
Harris
 
A
,
Baker
 
C
,
Iqbal
 
T
,
Lavies
 
N
,
Richards
 
T
, et al.  
British Committee for Standards in Haematology Guidelines on the identification and management of pre-operative anaemia
.
Br J Haematol
 
2015
;
171
:
322
331
.

365

Koch
 
CG
,
Reineks
 
EZ
,
Tang
 
AS
,
Hixson
 
ED
,
Phillips
 
S
,
Sabik
 
JF
, 3rd, et al.  
Contemporary bloodletting in cardiac surgical care
.
Ann Thorac Surg
 
2015
;
99
:
779
784
.

366

Riessen
 
R
,
Behmenburg
 
M
,
Blumenstock
 
G
,
Guenon
 
D
,
Enkel
 
S
,
Schafer
 
R
, et al.  
A simple “blood-saving bundle” reduces diagnostic blood loss and the transfusion rate in mechanically ventilated patients
.
PLoS One
 
2015
;
10
:
e0138879
.

367

Shander
 
A
,
Kaplan
 
LJ
,
Harris
 
MT
,
Gross
 
I
,
Nagarsheth
 
NP
,
Nemeth
 
J
, et al.  
Topical hemostatic therapy in surgery: bridging the knowledge and practice gap
.
J Am Coll Surg
 
2014
;
219
:
570
579.e574
.

368

Menkis
 
AH
,
Martin
 
J
,
Cheng
 
DC
,
Fitzgerald
 
DC
,
Freedman
 
JJ
,
Gao
 
C
, et al.  
Drug, devices, technologies, and techniques for blood management in minimally invasive and conventional cardiothoracic surgery: a consensus statement from the International Society for Minimally Invasive Cardiothoracic Surgery (ISMICS) 2011
.
Innovations (Phila)
 
2012
;
7
:
229
241
.

369

Anastasiadis
 
K
,
Murkin
 
J
,
Antonitsis
 
P
,
Bauer
 
A
,
Ranucci
 
M
,
Gygax
 
E
, et al.  
Use of minimal invasive extracorporeal circulation in cardiac surgery: principles, definitions and potential benefits. A position paper from the Minimal invasive Extra-Corporeal Technologies international Society (MiECTiS)
.
Interact Cardiovasc Thorac Surg
 
2016
;
22
:
647
662
.

370

Weber
 
CF
,
Görlinger
 
K
,
Meininger
 
D
,
Herrmann
 
E
,
Bingold
 
T
,
Moritz
 
A
, et al.  
Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients
.
Anesthesiology
 
2012
;
117
:
531
547
.

371

Weber
 
CF
,
Zacharowski
 
K
,
Meybohm
 
P
,
Adam
 
EH
,
Hofer
 
S
,
Brün
 
K
, et al.  
Hemotherapy algorithms for coagulopathic cardiac surgery patients
.
Clin Lab
 
2014
;
60
:
1059
1063
.

372

Muñoz
 
M
,
Gómez-Ramírez
 
S
,
Kozek-Langeneker
 
S
.
Pre-operative haematological assessment in patients scheduled for major surgery
.
Anaesthesia
 
2016
;
71 Suppl 1
:
19
28
.

373

Kozek-Langenecker
 
SA
,
Afshari
 
A
,
Albaladejo
 
P
,
Santullano
 
CA
,
De Robertis
 
E
,
Filipescu
 
DC
, et al.  
Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology
.
Eur J Anaesthesiol
 
2013
;
30
:
270
382
.

374

Meybohm
 
P
,
Zacharowski
 
K
,
Weber
 
CF
.
Point-of-care coagulation management in intensive care medicine
.
Crit Care
 
2013
;
17
:
218
.

375

Ker
 
K
,
Edwards
 
P
,
Perel
 
P
,
Shakur
 
H
,
Roberts
 
I
.
Effect of tranexamic acid on surgical bleeding: systematic review and cumulative meta-analysis
.
BMJ
 
2012
;
344
:
e3054
.

376

Devereaux
 
PJ
,
Marcucci
 
M
,
Painter
 
TW
,
Conen
 
D
,
Lomivorotov
 
V
,
Sessler
 
DI
, et al.  
Tranexamic acid in patients undergoing noncardiac surgery
.
N Engl J Med
 
2022
.

377

Meybohm
 
P
,
Choorapoikayil
 
S
,
Wessels
 
A
,
Herrmann
 
E
,
Zacharowski
 
K
,
Spahn
 
DR
.
Washed cell salvage in surgical patients: a review and meta-analysis of prospective randomized trials under PRISMA
.
Medicine (Baltimore)
 
2016
;
95
:
e4490
.

378

Santos
 
AA
,
Silva
 
JP
,
Silva Lda
 
F
,
Sousa
 
AG
,
Piotto
 
RF
,
Baumgratz
 
JF
.
Therapeutic options to minimize allogeneic blood transfusions and their adverse effects in cardiac surgery: a systematic review
.
Rev Bras Cir Cardiovasc
 
2014
;
29
:
606
621
.

379

Haensig
 
M
,
Kempfert
 
J
,
Kempfert
 
PM
,
Girdauskas
 
E
,
Borger
 
MA
,
Lehmann
 
S
.
Thrombelastometry guided blood-component therapy after cardiac surgery: a randomized study
.
BMC Anesthesiol
 
2019
;
19
:
201
.

380

Dias
 
JD
,
Sauaia
 
A
,
Achneck
 
HE
,
Hartmann
 
J
,
Moore
 
EE
.
Thromboelastography-guided therapy improves patient blood management and certain clinical outcomes in elective cardiac and liver surgery and emergency resuscitation: a systematic review and analysis
.
J Thromb Haemost
 
2019
;
17
:
984
994
.

381

Chutipongtanate
 
A
,
Yasaeng
 
C
,
Virankabutra
 
T
,
Chutipongtanate
 
S
.
Systematic comparison of four point-of-care methods versus the reference laboratory measurement of hemoglobin in the surgical ICU setting: a cross-sectional method comparison study
.
BMC Anesthesiol
 
2020
;
20
:
92
.

382

Terada
 
R
,
Ikeda
 
T
,
Mori
 
Y
,
Yamazaki
 
S
,
Kashiwabara
 
K
,
Yamauchi
 
H
, et al.  
Comparison of two point of care whole blood coagulation analysis devices and conventional coagulation tests as a predicting tool of perioperative bleeding in adult cardiac surgery – a pilot prospective observational study in Japan
.
Transfusion
 
2019
;
59
:
3525
3535
.

383

Cohen
 
J
,
Scorer
 
T
,
Wright
 
Z
,
Stewart
 
IJ
,
Sosnov
 
J
,
Pidcoke
 
H
, et al.  
A prospective evaluation of thromboelastometry (ROTEM) to identify acute traumatic coagulopathy and predict massive transfusion in military trauma patients in Afghanistan
.
Transfusion
 
2019
;
59
:
1601
1607
.

384

Yates
 
J
,
Perelman
 
I
,
Khair
 
S
,
Taylor
 
J
,
Lampron
 
J
,
Tinmouth
 
A
, et al.  
Exclusion criteria and adverse events in perioperative trials of tranexamic acid: a systematic review and meta-analysis
.
Transfusion
 
2019
;
59
:
806
824
.

385

CRASH-2 trial Collaborators
.
Effects of tranexamic acid on death, disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): a randomised, placebo-controlled trial
.
Lancet
 
2019
;
394
:
1713
1723
.

386

HALT-IT Trial Collaborators
.
Effects of a high-dose 24-h infusion of tranexamic acid on death and thromboembolic events in patients with acute gastrointestinal bleeding (HALT-IT): an international randomised, double-blind, placebo-controlled trial
.
Lancet
 
2020
;
395
:
1927
1936
.

387

Mukhopadhyay
 
A
,
Yip
 
HS
,
Prabhuswamy
 
D
,
Chan
 
YH
,
Phua
 
J
,
Lim
 
TK
, et al.  
The use of a blood conservation device to reduce red blood cell transfusion requirements: a before and after study
.
Crit Care
 
2010
;
14
:
R7
.

388

Peruzzi
 
WT
,
Parker
 
MA
,
Lichtenthal
 
PR
,
Cochran-Zull
 
C
,
Toth
 
B
,
Blake
 
M
.
A clinical evaluation of a blood conservation device in medical intensive care unit patients
.
Crit Care Med
 
1993
;
21
:
501
506
.

389

Kataife
 
ED
,
Said
 
S
,
Braun
 
J
,
Roche
 
TR
,
Rossler
 
J
,
Kaserer
 
A
, et al.  
The Haemostasis Traffic Light, a user-centred coagulation management tool for acute bleeding situations: a simulation-based randomised dual-centre trial
.
Anaesthesia
 
2021
;
76
:
902
910
.

390

Goodnough
 
LT
,
Shieh
 
L
,
Hadhazy
 
E
,
Cheng
 
N
,
Khari
 
P
,
Maggio
 
P
.
Improved blood utilization using real-time clinical decision support
.
Transfusion
 
2014
;
54
:
1358
1365
.

391

Oliver
 
JC
,
Griffin
 
RL
,
Hannon
 
T
,
Marques
 
MB
.
The success of our patient blood management program depended on an institution-wide change in transfusion practices
.
Transfusion
 
2014
;
54
:
2617
2624
.

392

Kaserer
 
A
,
Rössler
 
J
,
Braun
 
J
,
Farokhzad
 
F
,
Pape
 
HC
,
Dutkowski
 
P
, et al.  
Impact of a patient blood management monitoring and feedback programme on allogeneic blood transfusions and related costs
.
Anaesthesia
 
2019
;
74
:
1534
1541
.

393

Sardar
 
M
,
Azharuddin
 
M
,
Subedi
 
A
,
Ghatage
 
P
,
Du
 
D
,
Szallasi
 
A
.
Improving blood transfusion practices in a community hospital setting: our experience with real-time clinical decision support
.
Med Sci (Basel)
 
2018
;
6
.

394

Jenkins
 
I
,
Doucet
 
JJ
,
Clay
 
B
,
Kopko
 
P
,
Fipps
 
D
,
Hemmen
 
E
, et al.  
Transfusing wisely: clinical decision support improves blood transfusion practices
.
Jt Comm J Qual Patient Saf
 
2017
;
43
:
389
395
.

395

Staples
 
S
,
Salisbury
 
RA
,
King
 
AJ
,
Polzella
 
P
,
Bakhishli
 
G
,
Staves
 
J
, et al.  
How do we use electronic clinical decision support and feedback to promote good transfusion practice
.
Transfusion
 
2020
;
60
:
1658
1665
.

396

Derzon
 
JH
,
Clarke
 
N
,
Alford
 
A
,
Gross
 
I
,
Shander
 
A
,
Thurer
 
R
.
Restrictive transfusion strategy and clinical decision support practices for reducing RBC transfusion overuse
.
Am J Clin Pathol
 
2019
;
152
:
544
557
.

397

Dawood
 
MM
,
Gutpa
 
DK
,
Southern
 
J
,
Walia
 
A
,
Atkinson
 
JB
,
Eagle
 
KA
.
Pathology of fatal perioperative myocardial infarction: implications regarding pathophysiology and prevention
.
Int J Cardiol
 
1996
;
57
:
37
44
.

398

Eagle
 
KA
,
Rihal
 
CS
,
Mickel
 
MC
,
Holmes
 
DR
,
Foster
 
ED
,
Gersh
 
BJ
.
Cardiac risk of noncardiac surgery: influence of coronary disease and type of surgery in 3368 operations. CASS Investigators and University of Michigan Heart Care Program. Coronary Artery Surgery Study
.
Circulation
 
1997
;
96
:
1882
1887
.

399

McFalls
 
EO
,
Ward
 
HB
,
Moritz
 
TE
,
Goldman
 
S
,
Krupski
 
WC
,
Littooy
 
F
, et al.  
Coronary-artery revascularization before elective major vascular surgery
.
N Engl J Med
 
2004
;
351
:
2795
2804
.

400

Wong
 
EY
,
Lawrence
 
HP
,
Wong
 
DT
.
The effects of prophylactic coronary revascularization or medical management on patient outcomes after noncardiac surgery – a meta-analysis
.
Can J Anaesth
 
2007
;
54
:
705
717
.

401

Feng
 
B
,
Lin
 
J
,
Jin
 
J
,
Qian
 
W
,
Cao
 
S
,
Weng
 
X
.
The effect of previous coronary artery revascularization on the adverse cardiac events ninety days after total joint arthroplasty
.
J Arthroplasty
 
2018
;
33
:
235
240
.

402

Maron
 
DJ
,
Hochman
 
JS
,
Reynolds
 
HR
,
Bangalore
 
S
,
O’Brien
 
SM
,
Boden
 
WE
, et al.  
Initial invasive or conservative strategy for stable coronary disease
.
N Engl J Med
 
2020
;
382
:
1395
1407
.

403

Bainey
 
KR
,
Alemayehu
 
W
,
Welsh
 
RC
,
Kumar
 
A
,
King
 
SB
, 3rd
,
Kirtane
 
AJ
.
Long-term clinical outcomes following revascularization in high-risk coronary anatomy patients with stable ischemic heart disease
.
J Am Heart Assoc
 
2021
;
10
:
e018104
.

404

Windecker
 
S
,
Neumann
 
FJ
,
Juni
 
P
,
Sousa-Uva
 
M
,
Falk
 
V
.
Considerations for the choice between coronary artery bypass grafting and percutaneous coronary intervention as revascularization strategies in major categories of patients with stable multivessel coronary artery disease: an accompanying article of the task force of the 2018 ESC/EACTS guidelines on myocardial revascularization
.
Eur Heart J
 
2019
;
40
:
204
212
.

405

Raber
 
L
,
Mintz
 
GS
,
Koskinas
 
KC
,
Johnson
 
TW
,
Holm
 
NR
,
Onuma
 
Y
, et al.  
Clinical use of intracoronary imaging. Part 1: guidance and optimization of coronary interventions. An expert consensus document of the European Association of Percutaneous Cardiovascular Interventions
.
EuroIntervention
 
2018
;
14
:
656
677
.

406

Johnson
 
TW
,
Raber
 
L
,
Di Mario
 
C
,
Bourantas
 
CV
,
Jia
 
H
,
Mattesini
 
A
, et al.  
Clinical use of intracoronary imaging. Part 2: acute coronary syndromes, ambiguous coronary angiography findings, and guiding interventional decision-making: an expert consensus document of the European Association of Percutaneous Cardiovascular Interventions
.
EuroIntervention
 
2019
;
15
:
434
451
.

407

Richardt
 
G
,
Abdelghani
 
M
,
Allali
 
A
,
Toelg
 
R
,
Abdellaoui
 
M
,
Krackhardt
 
F
, et al.  
Polymer-free drug-coated vs. bare-metal coronary stents in patients undergoing non-cardiac surgery: a subgroup analysis of the LEADERS FREE trial
.
Clin Res Cardiol
 
2021
;
110
:
162
171
.

408

Cassese
 
S
,
Belle
 
L
,
Ndrepepa
 
G
,
Bosson
 
JL
,
Fusaro
 
M
,
Lonborg
 
J
, et al.  
Deferred vs immediate stenting in primary percutaneous coronary intervention: a collaborative meta-analysis of randomized trials with cardiac magnetic resonance imaging data
.
Can J Cardiol
 
2018
;
34
:
1573
1580
.

409

Belle
 
L
,
Motreff
 
P
,
Mangin
 
L
,
Range
 
G
,
Marcaggi
 
X
,
Marie
 
A
, et al.  
Comparison of immediate with delayed stenting using the minimalist immediate mechanical intervention approach in acute ST-segment-elevation myocardial infarction: the MIMI Study
.
Circ Cardiovasc Interv
 
2016
;
9
:
e003388
.

410

Hammill
 
BG
,
Curtis
 
LH
,
Bennett-Guerrero
 
E
,
O'Connor
 
CM
,
Jollis
 
JG
,
Schulman
 
KA
, et al.  
Impact of heart failure on patients undergoing major noncardiac surgery
.
Anesthesiology
 
2008
;
108
:
559
567
.

411

Goldman
 
L
,
Caldera
 
DL
,
Nussbaum
 
SR
,
Southwick
 
FS
,
Krogstad
 
D
,
Murray
 
B
, et al.  
Multifactorial index of cardiac risk in noncardiac surgical procedures
.
N Engl J Med
 
1977
;
297
:
845
850
.

412

McDonagh
 
TA
,
Metra
 
M
,
Adamo
 
M
,
Gardner
 
RS
,
Baumbach
 
A
,
Bohm
 
M
, et al.  
2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure
.
Eur Heart J
 
2021
;
42
:
3599
3726
.

413

Lee
 
TH
,
Marcantonio
 
ER
,
Mangione
 
CM
,
Thomas
 
EJ
,
Polanczyk
 
CA
,
Cook
 
EF
, et al.  
Derivation and prospective validation of a simple index for prediction of cardiac risk of major noncardiac surgery
.
Circulation
 
1999
;
100
:
1043
1049
.

414

Flu
 
WJ
,
van Kuijk
 
JP
,
Hoeks
 
SE
,
Kuiper
 
R
,
Schouten
 
O
,
Goei
 
D
, et al.  
Prognostic implications of asymptomatic left ventricular dysfunction in patients undergoing vascular surgery
.
Anesthesiology
 
2010
;
112
:
1316
1324
.

415

Cohn
 
SL
.
Preoperative evaluation for noncardiac surgery
.
Ann Intern Med
 
2016
;
165
:
ITC81
ITC96
.

416

Smilowitz
 
NR
,
Banco
 
D
,
Katz
 
SD
,
Beckman
 
JA
,
Berger
 
JS
.
Association between heart failure and perioperative outcomes in patients undergoing non-cardiac surgery
.
Eur Heart J Qual Care Clin Outcomes
 
2021
;
7
:
68
75
.

417

Lerman
 
BJ
,
Popat
 
RA
,
Assimes
 
TL
,
Heidenreich
 
PA
,
Wren
 
SM
.
Association between heart failure and postoperative mortality among patients undergoing ambulatory noncardiac surgery
.
JAMA Surg
 
2019
;
154
:
907
914
.

418

Dhillon
 
A
,
Khanna
 
A
,
Randhawa
 
MS
,
Cywinski
 
J
,
Saager
 
L
,
Thamilarasan
 
M
, et al.  
Perioperative outcomes of patients with hypertrophic cardiomyopathy undergoing non-cardiac surgery
.
Heart
 
2016
;
102
:
1627
1632
.

419

Nagueh
 
SF
,
Bierig
 
SM
,
Budoff
 
MJ
,
Desai
 
M
,
Dilsizian
 
V
,
Eidem
 
B
, et al.  
American Society of Echocardiography clinical recommendations for multimodality cardiovascular imaging of patients with hypertrophic cardiomyopathy. Endorsed by the American Society of Nuclear Cardiology, Society for Cardiovascular Magnetic Resonance, and Society of Cardiovascular Computed Tomography
.
J Am Soc Echocardiogr
 
2011
;
24
:
473
498
.

420

Kormos
 
RL
,
Cowger
 
J
,
Pagani
 
FD
,
Teuteberg
 
JJ
,
Goldstein
 
DJ
,
Jacobs
 
JP
, et al.  
The Society of Thoracic Surgeons Intermacs database annual report: evolving indications, outcomes, and scientific partnerships
.
J Heart Lung Transplant
 
2019
;
38
:
114
126
.

421

Roberts
 
SM
,
Hovord
 
DG
,
Kodavatiganti
 
R
,
Sathishkumar
 
S
.
Ventricular assist devices and non-cardiac surgery
.
BMC Anesthesiol
 
2015
;
15
:
185
.

422

Rodseth
 
RN
,
Biccard
 
BM
,
Le Manach
 
Y
,
Sessler
 
DI
,
Lurati Buse
 
GA
,
Thabane
 
L
, et al.  
The prognostic value of pre-operative and post-operative B-type natriuretic peptides in patients undergoing noncardiac surgery: B-type natriuretic peptide and N-terminal fragment of pro-B-type natriuretic peptide: a systematic review and individual patient data meta-analysis
.
J Am Coll Cardiol
 
2014
;
63
:
170
180
.

423

Karthikeyan
 
G
,
Moncur
 
RA
,
Levine
 
O
,
Heels-Ansdell
 
D
,
Chan
 
MT
,
Alonso-Coello
 
P
, et al.  
Is a pre-operative brain natriuretic peptide or N-terminal pro-B-type natriuretic peptide measurement an independent predictor of adverse cardiovascular outcomes within 30 days of noncardiac surgery? A systematic review and meta-analysis of observational studies
.
J Am Coll Cardiol
 
2009
;
54
:
1599
1606
.

424

Agarwal
 
S
,
Rajamanickam
 
A
,
Bajaj
 
NS
,
Griffin
 
BP
,
Catacutan
 
T
,
Svensson
 
LG
, et al.  
Impact of aortic stenosis on postoperative outcomes after noncardiac surgeries
.
Circ Cardiovasc Qual Outcomes
 
2013
;
6
:
193
200
.

425

Taniguchi
 
T
,
Morimoto
 
T
,
Shiomi
 
H
,
Ando
 
K
,
Shirai
 
S
,
Kanamori
 
N
, et al.  
Elective non-cardiac surgery in patients with severe aortic stenosis – observations from the CURRENT AS Registry
.
Circ J
 
2020
;
84
:
1173
1182
.

426

Luis
 
SA
,
Dohaei
 
A
,
Chandrashekar
 
P
,
Scott
 
CG
,
Padang
 
R
,
Lokineni
 
S
, et al.  
Impact of aortic valve replacement for severe aortic stenosis on perioperative outcomes following major noncardiac surgery
.
Mayo Clin Proc
 
2020
;
95
:
727
737
.

427

Okuno
 
T
,
Yahagi
 
K
,
Horiuchi
 
Y
,
Sato
 
Y
,
Tanaka
 
T
,
Koseki
 
K
, et al.  
The role of transcatheter aortic valve replacement in the patients with severe aortic stenosis requiring major non-cardiac surgery
.
Cardiovasc Interv Ther
 
2019
;
34
:
345
351
.

428

Calleja
 
AM
,
Dommaraju
 
S
,
Gaddam
 
R
,
Cha
 
S
,
Khandheria
 
BK
,
Chaliki
 
HP
.
Cardiac risk in patients aged >75 years with asymptomatic, severe aortic stenosis undergoing noncardiac surgery
.
Am J Cardiol
 
2010
;
105
:
1159
1163
.

429

Sohrabi
 
B
,
Kazemi
 
B
,
Mehryar
 
A
,
Teimouri-Dereshki
 
A
,
Toufan
 
M
,
Aslanabadi
 
N
.
Correlation between pulmonary artery pressure measured by echocardiography and right heart catheterization in patients with rheumatic mitral valve stenosis (a prospective study)
.
Echocardiography
 
2016
;
33
:
7
13
.

430

Bajaj
 
NS
,
Agarwal
 
S
,
Rajamanickam
 
A
,
Parashar
 
A
,
Poddar
 
KL
,
Griffin
 
BP
, et al.  
Impact of severe mitral regurgitation on postoperative outcomes after noncardiac surgery
.
Am J Med
 
2013
;
126
:
529
535
.

431

Stone
 
GW
,
Lindenfeld
 
J
,
Abraham
 
WT
,
Kar
 
S
,
Lim
 
DS
,
Mishell
 
JM
, et al.  
Transcatheter mitral-valve repair in patients with heart failure
.
N Engl J Med
 
2018
;
379
:
2307
2318
.

432

Habib
 
G
,
Lancellotti
 
P
,
Antunes
 
MJ
,
Bongiorni
 
MG
,
Casalta
 
JP
,
Del Zotti
 
F
, et al.  
2015 ESC Guidelines for the management of infective endocarditis: the Task Force for the Management of Infective Endocarditis of the European Society of Cardiology (ESC). Endorsed by: European Association for Cardio-Thoracic Surgery (EACTS), the European Association of Nuclear Medicine (EANM)
.
Eur Heart J
 
2015
;
36
:
3075
3128
.

433

Polanczyk
 
CA
,
Goldman
 
L
,
Marcantonio
 
ER
,
Orav
 
EJ
,
Lee
 
TH
.
Supraventricular arrhythmia in patients having noncardiac surgery: clinical correlates and effect on length of stay
.
Ann Intern Med
 
1998
;
129
:
279
285
.

434

van Diepen
 
S
,
Bakal
 
JA
,
McAlister
 
FA
,
Ezekowitz
 
JA
.
Mortality and readmission of patients with heart failure, atrial fibrillation, or coronary artery disease undergoing noncardiac surgery: an analysis of 38 047 patients
.
Circulation
 
2011
;
124
:
289
296
.

435

Alberte
 
C
,
Zipes
 
DP
.
Use of nonantiarrhythmic drugs for prevention of sudden cardiac death
.
J Cardiovasc Electrophysiol
 
2003
;
14
:
S87
S95
.

436

Brugada
 
J
,
Katritsis
 
DG
,
Arbelo
 
E
,
Arribas
 
F
,
Bax
 
JJ
,
Blomstrom-Lundqvist
 
C
, et al.  
2019 ESC Guidelines for the management of patients with supraventricular tachycardia
.
Eur Heart J
 
2020
;
41
:
655
720
.

437

Calkins
 
H
,
Yong
 
P
,
Miller
 
JM
,
Olshansky
 
B
,
Carlson
 
M
,
Saul
 
JP
, et al.  
Catheter ablation of accessory pathways, atrioventricular nodal reentrant tachycardia, and the atrioventricular junction: final results of a prospective, multicenter clinical trial. The Atakr Multicenter Investigators Group
.
Circulation
 
1999
;
99
:
262
270
.

438

Chen
 
SA
,
Chiang
 
CE
,
Yang
 
CJ
,
Cheng
 
CC
,
Wu
 
TJ
,
Wang
 
SP
, et al.  
Sustained atrial tachycardia in adult patients. Electrophysiological characteristics, pharmacological response, possible mechanisms, and effects of radiofrequency ablation
.
Circulation
 
1994
;
90
:
1262
1278
.

439

Katritsis
 
DG
,
Zografos
 
T
,
Katritsis
 
GD
,
Giazitzoglou
 
E
,
Vachliotis
 
V
,
Paxinos
 
G
, et al.  
Catheter ablation vs. antiarrhythmic drug therapy in patients with symptomatic atrioventricular nodal re-entrant tachycardia: a randomized, controlled trial
.
Europace
 
2017
;
19
:
602
606
.

440

Pappone
 
C
,
Vicedomini
 
G
,
Manguso
 
F
,
Saviano
 
M
,
Baldi
 
M
,
Pappone
 
A
, et al.  
Wolff-Parkinson-White syndrome in the era of catheter ablation: insights from a registry study of 2169 patients
.
Circulation
 
2014
;
130
:
811
819
.

441

Spector
 
P
,
Reynolds
 
MR
,
Calkins
 
H
,
Sondhi
 
M
,
Xu
 
Y
,
Martin
 
A
, et al.  
Meta-analysis of ablation of atrial flutter and supraventricular tachycardia
.
Am J Cardiol
 
2009
;
104
:
671
677
.

442

Potpara
 
TS
,
Polovina
 
MM
,
Marinkovic
 
JM
,
Lip
 
GY
.
A comparison of clinical characteristics and long-term prognosis in asymptomatic and symptomatic patients with first-diagnosed atrial fibrillation: the Belgrade Atrial Fibrillation Study
.
Int J Cardiol
 
2013
;
168
:
4744
4749
.

443

Mujovic
 
N
,
Dobrev
 
D
,
Marinkovic
 
M
,
Russo
 
V
,
Potpara
 
TS
.
The role of amiodarone in contemporary management of complex cardiac arrhythmias
.
Pharmacol Res
 
2020
;
151
:
104521
.

444

Dan
 
GA
,
Martinez-Rubio
 
A
,
Agewall
 
S
,
Boriani
 
G
,
Borggrefe
 
M
,
Gaita
 
F
, et al.  
Antiarrhythmic drugs-clinical use and clinical decision making: a consensus document from the European Heart Rhythm Association (EHRA) and European Society of Cardiology (ESC) Working Group on Cardiovascular Pharmacology, endorsed by the Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS) and International Society of Cardiovascular Pharmacotherapy (ISCP)
.
Europace
 
2018
;
20
:
731
732an
.

445

Ellenbogen
 
KA
,
Dias
 
VC
,
Plumb
 
VJ
,
Heywood
 
JT
,
Mirvis
 
DM
.
A placebo-controlled trial of continuous intravenous diltiazem infusion for 24-hour heart rate control during atrial fibrillation and atrial flutter: a multicenter study
.
J Am Coll Cardiol
 
1991
;
18
:
891
897
.

446

Phillips
 
BG
,
Gandhi
 
AJ
,
Sanoski
 
CA
,
Just
 
VL
,
Bauman
 
JL
.
Comparison of intravenous diltiazem and verapamil for the acute treatment of atrial fibrillation and atrial flutter
.
Pharmacotherapy
 
1997
;
17
:
1238
1245
.

447

Platia
 
EV
,
Michelson
 
EL
,
Porterfield
 
JK
,
Das
 
G
.
Esmolol versus verapamil in the acute treatment of atrial fibrillation or atrial flutter
.
Am J Cardiol
 
1989
;
63
:
925
929
.

448

Sethi
 
NJ
,
Nielsen
 
EE
,
Safi
 
S
,
Feinberg
 
J
,
Gluud
 
C
,
Jakobsen
 
JC
.
Digoxin for atrial fibrillation and atrial flutter: a systematic review with meta-analysis and trial sequential analysis of randomised clinical trials
.
PLoS One
 
2018
;
13
:
e0193924
.

449

Karth
 
GD
,
Geppert
 
A
,
Neunteufl
 
T
,
Priglinger
 
U
,
Haumer
 
M
,
Gschwandtner
 
M
, et al.  
Amiodarone versus diltiazem for rate control in critically ill patients with atrial tachyarrhythmias
.
Crit Care Med
 
2001
;
29
:
1149
1153
.

450

Vos
 
MA
,
Golitsyn
 
SR
,
Stangl
 
K
,
Ruda
 
MY
,
Van Wijk
 
LV
,
Harry
 
JD
, et al.  
Superiority of ibutilide (a new class III agent) over DL-sotalol in converting atrial flutter and atrial fibrillation. The Ibutilide/Sotalol Comparator Study Group
.
Heart
 
1998
;
79
:
568
575
.

451

Singh
 
S
,
Zoble
 
RG
,
Yellen
 
L
,
Brodsky
 
MA
,
Feld
 
GK
,
Berk
 
M
, et al.  
Efficacy and safety of oral dofetilide in converting to and maintaining sinus rhythm in patients with chronic atrial fibrillation or atrial flutter: the symptomatic atrial fibrillation investigative research on dofetilide (SAFIRE-D) study
.
Circulation
 
2000
;
102
:
2385
2390
.

452

Volgman
 
AS
,
Carberry
 
PA
,
Stambler
 
B
,
Lewis
 
WR
,
Dunn
 
GH
,
Perry
 
KT
, et al.  
Conversion efficacy and safety of intravenous ibutilide compared with intravenous procainamide in patients with atrial flutter or fibrillation
.
J Am Coll Cardiol
 
1998
;
31
:
1414
1419
.

453

Bianconi
 
L
,
Castro
 
A
,
Dinelli
 
M
,
Alboni
 
P
,
Pappalardo
 
A
,
Richiardi
 
E
, et al.  
Comparison of intravenously administered dofetilide versus amiodarone in the acute termination of atrial fibrillation and flutter. A multicentre, randomized, double-blind, placebo-controlled study
.
Eur Heart J
 
2000
;
21
:
1265
1273
.

454

Kerola
 
T
,
Dewland
 
TA
,
Vittinghoff
 
E
,
Heckbert
 
SR
,
Stein
 
PK
,
Marcus
 
GM
.
Modifiable predictors of ventricular ectopy in the community
.
J Am Heart Assoc
 
2018
;
7
:
e010078
.

455

Yang
 
J
,
Dudum
 
R
,
Mandyam
 
MC
,
Marcus
 
GM
.
Characteristics of unselected high-burden premature ventricular contraction patients
.
Pacing Clin Electrophysiol
 
2014
;
37
:
1671
1680
.

456

Parreira
 
L
,
Marinheiro
 
R
,
Amador
 
P
,
Mesquita
 
D
,
Farinha
 
J
,
Lopes
 
A
, et al.  
Frequent premature ventricular contractions. Association of burden and complexity with prognosis according to the presence of structural heart disease
.
Ann Noninvasive Electrocardiol
 
2021
;
26
:
e12800
.

457

Agarwal
 
V
,
Vittinghoff
 
E
,
Whitman
 
IR
,
Dewland
 
TA
,
Dukes
 
JW
,
Marcus
 
GM
.
Relation between ventricular premature complexes and incident heart failure
.
Am J Cardiol
 
2017
;
119
:
1238
1242
.

458

Dukes
 
JW
,
Dewland
 
TA
,
Vittinghoff
 
E
,
Mandyam
 
MC
,
Heckbert
 
SR
,
Siscovick
 
DS
, et al.  
Ventricular ectopy as a predictor of heart failure and death
.
J Am Coll Cardiol
 
2015
;
66
:
101
109
.

459

Marcus
 
GM
.
Evaluation and management of premature ventricular complexes
.
Circulation
 
2020
;
141
:
1404
1418
.

460

Mukharji
 
J
,
Rude
 
RE
,
Poole
 
WK
,
Gustafson
 
N
,
Thomas
 
LJ
, Jr.
,
Strauss
 
HW
, et al.  
Risk factors for sudden death after acute myocardial infarction: two-year follow-up
.
Am J Cardiol
 
1984
;
54
:
31
36
.

461

Chen
 
J
,
Johnson
 
G
,
Hellkamp
 
AS
,
Anderson
 
J
,
Mark
 
DB
,
Lee
 
KL
, et al.  
Rapid-rate nonsustained ventricular tachycardia found on implantable cardioverter-defibrillator interrogation: relationship to outcomes in the SCD-HeFT (Sudden Cardiac Death in Heart Failure Trial)
.
J Am Coll Cardiol
 
2013
;
61
:
2161
2168
.

462

Makimoto
 
H
,
Zielke
 
S
,
Clasen
 
L
,
Lin
 
T
,
Gerguri
 
S
,
Muller
 
P
, et al.  
Clinical significance of precedent asymptomatic non-sustained ventricular tachycardias on subsequent ICD interventions and heart failure hospitalization in primary prevention ICD patients
.
Eur J Med Res
 
2020
;
25
:
5
.

463

Sanchez Munoz
 
JJ
,
Garcia-Alberola
 
A
,
Martinez-Sanchez
 
J
,
Penafiel-Verdu
 
P
,
Caro-Martinez
 
C
,
Manzano-Fernandez
 
S
, et al.  
Premature ventricular complexes as a trigger for ventricular fibrillation
.
Rev Esp Cardiol
 
2010
;
63
:
798
801
.

464

Kakishita
 
M
,
Kurita
 
T
,
Matsuo
 
K
,
Taguchi
 
A
,
Suyama
 
K
,
Shimizu
 
W
, et al.  
Mode of onset of ventricular fibrillation in patients with Brugada syndrome detected by implantable cardioverter defibrillator therapy
.
J Am Coll Cardiol
 
2000
;
36
:
1646
1653
.

465

Srivathsan
 
K
,
Gami
 
AS
,
Ackerman
 
MJ
,
Asirvatham
 
SJ
.
Treatment of ventricular fibrillation in a patient with prior diagnosis of long QT syndrome: importance of precise electrophysiologic diagnosis to successfully ablate the trigger
.
Heart Rhythm
 
2007
;
4
:
1090
1093
.

466

Almendral
 
J
,
Villacastin
 
JP
,
Arenal
 
A
,
Tercedor
 
L
,
Merino
 
JL
,
Delcan
 
JL
.
Evidence favoring the hypothesis that ventricular arrhythmias have prognostic significance in left ventricular hypertrophy secondary to systemic hypertension
.
Am J Cardiol
 
1995
;
76
:
60D
63D
.

467

Priori
 
SG
,
Blomstrom-Lundqvist
 
C
,
Mazzanti
 
A
,
Blom
 
N
,
Borggrefe
 
M
,
Camm
 
J
, et al.  
2015 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: the Task Force for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death of the European Society of Cardiology (ESC). Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC)
.
Eur Heart J
 
2015
;
36
:
2793
2867
.

468

Connolly
 
SJ
,
Dorian
 
P
,
Roberts
 
RS
,
Gent
 
M
,
Bailin
 
S
,
Fain
 
ES
, et al.  
Comparison of beta-blockers, amiodarone plus beta-blockers, or sotalol for prevention of shocks from implantable cardioverter defibrillators: the OPTIC Study: a randomized trial
.
JAMA
 
2006
;
295
:
165
171
.

469

Brodine
 
WN
,
Tung
 
RT
,
Lee
 
JK
,
Hockstad
 
ES
,
Moss
 
AJ
,
Zareba
 
W
, et al.  
Effects of beta-blockers on implantable cardioverter defibrillator therapy and survival in the patients with ischemic cardiomyopathy (from the Multicenter Automatic Defibrillator Implantation Trial-II)
.
Am J Cardiol
 
2005
;
96
:
691
695
.

470

Tilz
 
RR
,
Lenarczyk
 
R
,
Scherr
 
D
,
Haugaa
 
KH
,
Iliodromitis
 
K
,
Purerfellner
 
H
, et al.  
Management of ventricular tachycardia in the ablation era: results of the European Heart Rhythm Association Survey
.
Europace
 
2018
;
20
:
209
213
.

471

Cronin
 
EM
,
Bogun
 
FM
,
Maury
 
P
,
Peichl
 
P
,
Chen
 
M
,
Namboodiri
 
N
, et al.  
2019 HRS/EHRA/APHRS/LAHRS expert consensus statement on catheter ablation of ventricular arrhythmias
.
Europace
 
2019
;
21
:
1143
1144
.

472

Scheuermeyer
 
FX
,
Grafstein
 
E
,
Stenstrom
 
R
,
Innes
 
G
,
Heslop
 
C
,
MacPhee
 
J
, et al.  
Thirty-day and 1-year outcomes of emergency department patients with atrial fibrillation and no acute underlying medical cause
.
Ann Emerg Med
 
2012
;
60
:
755
765.e752
.

473

Bonfanti
 
L
,
Annovi
 
A
,
Sanchis-Gomar
 
F
,
Saccenti
 
C
,
Meschi
 
T
,
Ticinesi
 
A
, et al.  
Effectiveness and safety of electrical cardioversion for acute-onset atrial fibrillation in the emergency department: a real-world 10-year single center experience
.
Clin Exp Emerg Med
 
2019
;
6
:
64
69
.

474

Kheiri
 
B
,
Barbarawi
 
M
,
Zayed
 
Y
,
Hicks
 
M
,
Osman
 
M
,
Rashdan
 
L
, et al.  
Antiarrhythmic drugs or catheter ablation in the management of ventricular tachyarrhythmias in patients with implantable cardioverter-defibrillators: a systematic review and meta-analysis of randomized controlled trials
.
Circ Arrhythm Electrophysiol
 
2019
;
12
:
e007600
.

475

Santangeli
 
P
,
Muser
 
D
,
Maeda
 
S
,
Filtz
 
A
,
Zado
 
ES
,
Frankel
 
DS
, et al.  
Comparative effectiveness of antiarrhythmic drugs and catheter ablation for the prevention of recurrent ventricular tachycardia in patients with implantable cardioverter-defibrillators: a systematic review and meta-analysis of randomized controlled trials
.
Heart Rhythm
 
2016
;
13
:
1552
1559
.

476

Martinez
 
BK
,
Baker
 
WL
,
Konopka
 
A
,
Giannelli
 
D
,
Coleman
 
CI
,
Kluger
 
J
, et al.  
Systematic review and meta-analysis of catheter ablation of ventricular tachycardia in ischemic heart disease
.
Heart Rhythm
 
2020
;
17
:
e206
e219
.

477

Reddy
 
VY
,
Reynolds
 
MR
,
Neuzil
 
P
,
Richardson
 
AW
,
Taborsky
 
M
,
Jongnarangsin
 
K
, et al.  
Prophylactic catheter ablation for the prevention of defibrillator therapy
.
N Engl J Med
 
2007
;
357
:
2657
2665
.

478

Sapp
 
JL
,
Wells
 
GA
,
Parkash
 
R
,
Stevenson
 
WG
,
Blier
 
L
,
Sarrazin
 
JF
, et al.  
Ventricular tachycardia ablation versus escalation of antiarrhythmic drugs
.
N Engl J Med
 
2016
;
375
:
111
121
.

479

Kuck
 
KH
,
Schaumann
 
A
,
Eckardt
 
L
,
Willems
 
S
,
Ventura
 
R
,
Delacretaz
 
E
, et al.  
Catheter ablation of stable ventricular tachycardia before defibrillator implantation in patients with coronary heart disease (VTACH): a multicentre randomised controlled trial
.
Lancet
 
2010
;
375
:
31
40
.

480

Briceno
 
DF
,
Gupta
 
T
,
Romero
 
J
,
Kolte
 
D
,
Khera
 
S
,
Villablanca
 
PA
, et al.  
Catheter ablation of ventricular tachycardia in nonischemic cardiomyopathy: a propensity score-matched analysis of in-hospital outcomes in the United States
.
J Cardiovasc Electrophysiol
 
2018
;
29
:
771
779
.

481

Glikson
 
M
,
Nielsen
 
JC
,
Kronborg
 
MB
,
Michowitz
 
Y
,
Auricchio
 
A
,
Barbash
 
IM
, et al.  
2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy
.
Eur Heart J
 
2021
;
42
:
3427
3520
.

482

Marti-Almor
 
J
,
Cladellas
 
M
,
Bazan
 
V
,
Delclos
 
J
,
Altaba
 
C
,
Guijo
 
MA
, et al.  
Novel predictors of progression of atrioventricular block in patients with chronic bifascicular block
.
Rev Esp Cardiol
 
2010
;
63
:
400
408
.

483

Santini
 
M
,
Castro
 
A
,
Giada
 
F
,
Ricci
 
R
,
Inama
 
G
,
Gaggioli
 
G
, et al.  
Prevention of syncope through permanent cardiac pacing in patients with bifascicular block and syncope of unexplained origin: the PRESS study
.
Circ Arrhythm Electrophysiol
 
2013
;
6
:
101
107
.

484

Stone
 
ME
,
Salter
 
B
,
Fischer
 
A
.
Perioperative management of patients with cardiac implantable electronic devices
.
Br J Anaesth
 
2011
;
107
:
i16
i26
.

485

Cardiac implantable electronic device management (corrected)
.
Anesthesiology
 
2020
;
132
:
225
252
.

486

Gifford
 
J
,
Larimer
 
K
,
Thomas
 
C
,
May
 
P
.
ICD-ON Registry for perioperative management of CIEDs: most require no change
.
Pacing Clin Electrophysiol
 
2017
;
40
:
128
134
.

487

Mahlow
 
WJ
,
Craft
 
RM
,
Misulia
 
NL
,
Cox
 
JW
, Jr.
,
Hirsh
 
JB
,
Snider
 
CC
, et al.  
A perioperative management algorithm for cardiac rhythm management devices: the PACED-OP protocol
.
Pacing Clin Electrophysiol
 
2013
;
36
:
238
248
.

488

Neubauer
 
H
,
Wellmann
 
M
,
Herzog-Niescery
 
J
,
Wutzler
 
A
,
Weber
 
TP
,
Mugge
 
A
, et al.  
Comparison of perioperative strategies in ICD patients: the perioperative ICD management study (PIM study)
.
Pacing Clin Electrophysiol
 
2018
;
41
:
1536
1542
.

489

Feldman
 
JB
,
Stone
 
ME
.
Anesthesia teams managing pacemakers and ICDs for the perioperative period: enhanced patient safety and improved workflows
.
Curr Opin Anaesthesiol
 
2020
;
33
:
441
447
.

490

Mickus
 
GJ
,
Soliman
 
GI
,
Reed
 
RR
,
Martin
 
AK
.
Perioperative management of a leadless pacemaker: the paucity of evidence-based guidelines
.
J Cardiothorac Vasc Anesth
 
2016
;
30
:
1594
1598
.

491

Karuppiah
 
S
,
Prielipp
 
R
,
Banik
 
RK
.
Anesthetic consideration for patients with micra leadless pacemaker
.
Ann Card Anaesth
 
2020
;
23
:
493
495
.

492

Gifford
 
J
,
Larimer
 
K
,
Thomas
 
C
,
May
 
P
,
Stanhope
 
S
,
Gami
 
A
.
Randomized controlled trial of perioperative ICD management: magnet application versus reprogramming
.
Pacing Clin Electrophysiol
 
2014
;
37
:
1219
1224
.

493

Winter
 
J
,
Kohlmeier
 
A
,
Shin
 
DI
,
O’Connor
 
S
.
Subcutaneous implantable cardioverter-defibrillators and sternal wires: a cautionary tale
.
Circ Arrhythm Electrophysiol
 
2014
;
7
:
986
987
.

494

Schulman
 
PM
,
Treggiari
 
MM
,
Yanez
 
ND
,
Henrikson
 
CA
,
Jessel
 
PM
,
Dewland
 
TA
, et al.  
Electromagnetic interference with protocolized electrosurgery dispersive electrode positioning in patients with implantable cardioverter defibrillators
.
Anesthesiology
 
2019
;
130
:
530
540
.

495

Baeg
 
MK
,
Kim
 
SW
,
Ko
 
SH
,
Lee
 
YB
,
Hwang
 
S
,
Lee
 
BW
, et al.  
Endoscopic electrosurgery in patients with cardiac implantable electronic devices
.
Clin Endosc
 
2016
;
49
:
176
181
.

496

Mangar
 
D
,
Atlas
 
GM
,
Kane
 
PB
.
Electrocautery-induced pacemaker malfunction during surgery
.
Can J Anaesth
 
1991
;
38
:
616
618
.

497

Rozner
 
MA
.
Review of electrical interference in implanted cardiac devices
.
Pacing Clin Electrophysiol
 
2003
;
26
:
923
925
.

498

de Cock
 
CC
,
Spruijt
 
HJ
,
van Campen
 
LM
,
Plu
 
AW
,
Visser
 
CA
.
Electromagnetic interference of an implantable loop recorder by commonly encountered electronic devices
.
Pacing Clin Electrophysiol
 
2000
;
23
:
1516
1518
.

499

Suarez-Fuster
 
L
,
Oh
 
C
,
Baranchuk
 
A
.
Transcutaneous electrical nerve stimulation electromagnetic interference in an implantable loop recorder
.
J Arrhythm
 
2018
;
34
:
96
97
.

500

Gilboa
 
SM
,
Salemi
 
JL
,
Nembhard
 
WN
,
Fixler
 
DE
,
Correa
 
A
.
Mortality resulting from congenital heart disease among children and adults in the United States, 1999 to 2006
.
Circulation
 
2010
;
122
:
2254
2263
.

501

Marelli
 
AJ
,
Ionescu-Ittu
 
R
,
Mackie
 
AS
,
Guo
 
L
,
Dendukuri
 
N
,
Kaouache
 
M
.
Lifetime prevalence of congenital heart disease in the general population from 2000 to 2010
.
Circulation
 
2014
;
130
:
749
756
.

502

Maxwell
 
BG
,
Wong
 
JK
,
Kin
 
C
,
Lobato
 
RL
.
Perioperative outcomes of major noncardiac surgery in adults with congenital heart disease
.
Anesthesiology
 
2013
;
119
:
762
769
.

503

Baumgartner
 
H
,
De Backer
 
J
,
Babu-Narayan
 
SV
,
Budts
 
W
,
Chessa
 
M
,
Diller
 
GP
, et al.  
2020 ESC Guidelines for the management of adult congenital heart disease
.
Eur Heart J
 
2021
;
42
:
563
645
.

504

Regitz-Zagrosek
 
V
,
Roos-Hesselink
 
JW
,
Bauersachs
 
J
,
Blomström-Lundqvist
 
C
,
Cífková
 
R
,
De Bonis
 
M
, et al.  
2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy
.
Eur Heart J
 
2018
;
39
:
3165
3241
.

505

Ammash
 
NM
,
Connolly
 
HM
,
Abel
 
MD
,
Warnes
 
CA
.
Noncardiac surgery in Eisenmenger syndrome
.
J Am Coll Cardiol
 
1999
;
33
:
222
227
.

506

Faraoni
 
D
,
Zurakowski
 
D
,
Vo
 
D
,
Goobie
 
SM
,
Yuki
 
K
,
Brown
 
ML
, et al.  
Post-operative outcomes in children with and without congenital heart disease undergoing noncardiac surgery
.
J Am Coll Cardiol
 
2016
;
67
:
793
801
.

507

Alonso-Gonzalez
 
R
,
Borgia
 
F
,
Diller
 
GP
,
Inuzuka
 
R
,
Kempny
 
A
,
Martinez-Naharro
 
A
, et al.  
Abnormal lung function in adults with congenital heart disease: prevalence, relation to cardiac anatomy, and association with survival
.
Circulation
 
2013
;
127
:
882
890
.

508

Maxwell
 
BG
,
Posner
 
KL
,
Wong
 
JK
,
Oakes
 
DA
,
Kelly
 
NE
,
Domino
 
KB
, et al.  
Factors contributing to adverse perioperative events in adults with congenital heart disease: a structured analysis of cases from the closed claims project
.
Congenit Heart Dis
 
2015
;
10
:
21
29
.

509

Lui
 
GK
,
Saidi
 
A
,
Bhatt
 
AB
,
Burchill
 
LJ
,
Deen
 
JF
,
Earing
 
MG
, et al.  
Diagnosis and management of noncardiac complications in adults with congenital heart disease: a scientific statement from the American Heart Association
.
Circulation
 
2017
;
136
:
e348
e392
.

510

Adler
 
Y
,
Charron
 
P
,
Imazio
 
M
,
Badano
 
L
,
Baron-Esquivias
 
G
,
Bogaert
 
J
, et al.  
2015 ESC Guidelines for the diagnosis and management of pericardial diseases: the Task Force for the Diagnosis and Management of Pericardial Diseases of the European Society of Cardiology (ESC). Endorsed by: the European Association for Cardio-Thoracic Surgery (EACTS)
.
Eur Heart J
 
2015
;
36
:
2921
2964
.

511

Tuck
 
BC
,
Townsley
 
MM
.
Clinical update in pericardial diseases
.
J Cardiothorac Vasc Anesth
 
2019
;
33
:
184
199
.

512

Ristic
 
AD
,
Imazio
 
M
,
Adler
 
Y
,
Anastasakis
 
A
,
Badano
 
LP
,
Brucato
 
A
, et al.  
Triage strategy for urgent management of cardiac tamponade: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases
.
Eur Heart J
 
2014
;
35
:
2279
2284
.

513

Sin
 
DD
,
Wu
 
L
,
Man
 
SF
.
The relationship between reduced lung function and cardiovascular mortality: a population-based study and a systematic review of the literature
.
Chest
 
2005
;
127
:
1952
1959
.

514

Canet
 
J
,
Gallart
 
L
,
Gomar
 
C
,
Paluzie
 
G
,
Valles
 
J
,
Castillo
 
J
, et al.  
Prediction of postoperative pulmonary complications in a population-based surgical cohort
.
Anesthesiology
 
2010
;
113
:
1338
1350
.

515

Edrich
 
T
,
Sadovnikoff
 
N
.
Anesthesia for patients with severe chronic obstructive pulmonary disease
.
Curr Opin Anaesthesiol
 
2010
;
23
:
18
24
.

516

Chau
 
EH
,
Lam
 
D
,
Wong
 
J
,
Mokhlesi
 
B
,
Chung
 
F
.
Obesity hypoventilation syndrome: a review of epidemiology, pathophysiology, and perioperative considerations
.
Anesthesiology
 
2012
;
117
:
188
205
.

517

Kaw
 
R
,
Gali
 
B
,
Collop
 
NA
.
Perioperative care of patients with obstructive sleep apnea
.
Curr Treat Options Neurol
 
2011
;
13
:
496
507
.

518

Galie
 
N
,
Humbert
 
M
,
Vachiery
 
JL
,
Gibbs
 
S
,
Lang
 
I
,
Torbicki
 
A
, et al.  
2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: the Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS). Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT)
.
Eur Heart J
 
2016
;
37
:
67
119
.

519

Bowker
 
D
,
Banks
 
D
. Pulmonary hypertension in noncardiac surgical patients. In:
Kaplan
 
JA
 
Cronin
 
B
and
Maus
 
TM
, editors.
Essentials of Cardiac Anesthesia for Noncardiac Surgery
.
Philadelphia
,
PA
:
Elsevier
;
2019
. p
138
164
.

520

Meyer
 
S
,
McLaughlin
 
VV
,
Seyfarth
 
HJ
,
Bull
 
TM
,
Vizza
 
CD
,
Gomberg-Maitland
 
M
, et al.  
Outcomes of noncardiac, nonobstetric surgery in patients with PAH: an international prospective survey
.
Eur Respir J
 
2013
;
41
:
1302
1307
.

521

Price
 
LC
,
Montani
 
D
,
Jais
 
X
,
Dick
 
JR
,
Simonneau
 
G
,
Sitbon
 
O
, et al.  
Noncardiothoracic nonobstetric surgery in mild-to-moderate pulmonary hypertension
.
Eur Respir J
 
2010
;
35
:
1294
1302
.

522

Olsson
 
KM
,
Halank
 
M
,
Egenlauf
 
B
,
Fistera
 
D
,
Gall
 
H
,
Kaehler
 
C
, et al.  
Decompensated right heart failure, intensive care and perioperative management in patients with pulmonary hypertension: updated recommendations from the Cologne Consensus Conference 2018
.
Int J Cardiol
 
2018
;
272S
:
46
52
.

523

Pilkington
 
SA
,
Taboada
 
D
,
Martinez
 
G
.
Pulmonary hypertension and its management in patients undergoing non-cardiac surgery
.
Anaesthesia
 
2015
;
70
:
56
70
.

524

ESC
.
2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension
.
Eur Heart J
 
2022
;
43
:
3618
3731
.

525

Thunberg
 
CA
,
Gaitan
 
BD
,
Grewal
 
A
,
Ramakrishna
 
H
,
Stansbury
 
LG
,
Grigore
 
AM
.
Pulmonary hypertension in patients undergoing cardiac surgery: pathophysiology, perioperative management, and outcomes
.
J Cardiothorac Vasc Anesth
 
2013
;
27
:
551
572
.

526

Hayward
 
CS
,
Kelly
 
RP
,
Macdonald
 
PS
.
Inhaled nitric oxide in cardiology practice
.
Cardiovasc Res
 
1999
;
43
:
628
638
.

527

Chow
 
CK
,
Teo
 
KK
,
Rangarajan
 
S
,
Islam
 
S
,
Gupta
 
R
,
Avezum
 
A
, et al.  
Prevalence, awareness, treatment, and control of hypertension in rural and urban communities in high-, middle-, and low-income countries
.
JAMA
 
2013
;
310
:
959
968
.

528

Futier
 
E
,
Lefrant
 
JY
,
Guinot
 
PG
,
Godet
 
T
,
Lorne
 
E
,
Cuvillon
 
P
, et al.  
Effect of individualized vs standard blood pressure management strategies on postoperative organ dysfunction among high-risk patients undergoing major surgery: a randomized clinical trial
.
JAMA
 
2017
;
318
:
1346
1357
.

529

Williams
 
B
,
Mancia
 
G
,
Spiering
 
W
,
Agabiti Rosei
 
E
,
Azizi
 
M
,
Burnier
 
M
, et al.  
2018 ESC/ESH Guidelines for the management of arterial hypertension
.
Eur Heart J
 
2018
;
39
:
3021
3104
.

530

Howell
 
SJ
,
Sear
 
JW
,
Foex
 
P
.
Hypertension, hypertensive heart disease and perioperative cardiac risk
.
Br J Anaesth
 
2004
;
92
:
570
583
.

531

Messina
 
A
,
Robba
 
C
,
Calabro
 
L
,
Zambelli
 
D
,
Iannuzzi
 
F
,
Molinari
 
E
, et al.  
Association between perioperative fluid administration and postoperative outcomes: a 20-year systematic review and a meta-analysis of randomized goal-directed trials in major visceral/noncardiac surgery
.
Crit Care
 
2021
;
25
:
43
.

532

Jorgensen
 
ME
,
Hlatky
 
MA
,
Kober
 
L
,
Sanders
 
RD
,
Torp-Pedersen
 
C
,
Gislason
 
GH
, et al.  
Beta-blocker-associated risks in patients with uncomplicated hypertension undergoing noncardiac surgery
.
JAMA Intern Med
 
2015
;
175
:
1923
1931
.

533

Weksler
 
N
,
Klein
 
M
,
Szendro
 
G
,
Rozentsveig
 
V
,
Schily
 
M
,
Brill
 
S
, et al.  
The dilemma of immediate preoperative hypertension: to treat and operate, or to postpone surgery?
 
J Clin Anesth
 
2003
;
15
:
179
183
.

534

Perk
 
J
,
De Backer
 
G
,
Gohlke
 
H
,
Graham
 
I
,
Reiner
 
Z
,
Verschuren
 
M
, et al.  
European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts)
.
Eur Heart J
 
2012
;
33
:
1635
1701
.

535

Aboyans
 
V
,
Ricco
 
JB
,
Bartelink
 
MEL
,
Bjorck
 
M
,
Brodmann
 
M
,
Cohnert
 
T
, et al.  
2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS). Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteries. Endorsed by: the European Stroke Organization (ESO). The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS)
.
Eur Heart J
 
2018
;
39
:
763
816
.

536

Bonaca
 
MP
,
Nault
 
P
,
Giugliano
 
RP
,
Keech
 
AC
,
Pineda
 
AL
,
Kanevsky
 
E
, et al.  
Low-density lipoprotein cholesterol lowering with evolocumab and outcomes in patients with peripheral artery disease: insights from the FOURIER Trial (Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk)
.
Circulation
 
2018
;
137
:
338
350
.

537

Anand
 
SS
,
Bosch
 
J
,
Eikelboom
 
JW
,
Connolly
 
SJ
,
Diaz
 
R
,
Widimsky
 
P
, et al.  
Rivaroxaban with or without aspirin in patients with stable peripheral or carotid artery disease: an international, randomised, double-blind, placebo-controlled trial
.
Lancet
 
2018
;
391
:
219
229
.

538

Ashton
 
CM
,
Petersen
 
NJ
,
Wray
 
NP
,
Kiefe
 
CI
,
Dunn
 
JK
,
Wu
 
L
, et al.  
The incidence of perioperative myocardial infarction in men undergoing noncardiac surgery
.
Ann Intern Med
 
1993
;
118
:
504
510
.

539

Conte
 
MS
,
Bradbury
 
AW
,
Kolh
 
P
,
White
 
JV
,
Dick
 
F
,
Fitridge
 
R
, et al.  
Global vascular guidelines on the management of chronic limb-threatening ischemia
.
Eur J Vasc Endovasc Surg
 
2019
;
58
:
S1
S109.e33
.

540

Brown
 
LC
,
Powell
 
JT
,
Thompson
 
SG
,
Epstein
 
DM
,
Sculpher
 
MJ
,
Greenhalgh
 
RM
.
The UK EndoVascular Aneurysm Repair (EVAR) trials: randomised trials of EVAR versus standard therapy
.
Health Technol Assess
 
2012
;
16
:
1
218
.

541

Paravastu
 
SC
,
Jayarajasingam
 
R
,
Cottam
 
R
,
Palfreyman
 
SJ
,
Michaels
 
JA
,
Thomas
 
SM
.
Endovascular repair of abdominal aortic aneurysm
.
Cochrane Database Syst Rev
 
2014
:
CD004178
. doi:.

542

Stather
 
PW
,
Sidloff
 
D
,
Dattani
 
N
,
Choke
 
E
,
Bown
 
MJ
,
Sayers
 
RD
.
Systematic review and meta-analysis of the early and late outcomes of open and endovascular repair of abdominal aortic aneurysm
.
Br J Surg
 
2013
;
100
:
863
872
.

543

Eslami
 
MH
,
Rybin
 
DV
,
Doros
 
G
,
Siracuse
 
JJ
,
Farber
 
A
.
External validation of Vascular Study Group of New England risk predictive model of mortality after elective abdominal aorta aneurysm repair in the Vascular Quality Initiative and comparison against established models
.
J Vasc Surg
 
2018
;
67
:
143
150
.

544

Bonaca
 
MP
,
Bauersachs
 
RM
,
Anand
 
SS
,
Debus
 
ES
,
Nehler
 
MR
,
Patel
 
MR
, et al.  
Rivaroxaban in peripheral artery disease after revascularization
.
N Engl J Med
 
2020
;
382
:
1994
2004
.

545

Cutlip
 
DE
,
Pinto
 
DS
.
Extracranial carotid disease revascularization
.
Circulation
 
2012
;
126
:
2636
2644
.

546

Herzog
 
CA
,
Asinger
 
RW
,
Berger
 
AK
,
Charytan
 
DM
,
Diez
 
J
,
Hart
 
RG
, et al.  
Cardiovascular disease in chronic kidney disease. A clinical update from Kidney Disease: Improving Global Outcomes (KDIGO)
.
Kidney Int
 
2011
;
80
:
572
586
.

547

Section 3: Prevention and Treatment of AKI
.
Kidney Int Suppl (2011)
 
2012
;
2
:
37
68
.

548

Romagnoli
 
S
,
Ricci
 
Z
,
Ronco
 
C
.
Perioperative acute kidney injury: prevention, early recognition, and supportive measures
.
Nephron
 
2018
;
140
:
105
110
.

549

Kheterpal
 
S
,
Tremper
 
KK
,
Heung
 
M
,
Rosenberg
 
AL
,
Englesbe
 
M
,
Shanks
 
AM
, et al.  
Development and validation of an acute kidney injury risk index for patients undergoing general surgery: results from a national data set
.
Anesthesiology
 
2009
;
110
:
505
515
.

550

Rebholz
 
CM
,
Inker
 
LA
,
Chen
 
Y
,
Liang
 
M
,
Foster
 
MC
,
Eckfeldt
 
JH
, et al.  
Risk of ESRD and mortality associated with change in filtration markers
.
Am J Kidney Dis
 
2017
;
70
:
551
560
.

551

McCullough
 
PA
,
Choi
 
JP
,
Feghali
 
GA
,
Schussler
 
JM
,
Stoler
 
RM
,
Vallabahn
 
RC
, et al.  
Contrast-induced acute kidney injury
.
J Am Coll Cardiol
 
2016
;
68
:
1465
1473
.

552

Section 2: AKI Definition
.
Kidney Int Suppl (2011)
 
2012
;
2
:
19
36
.

553

Goren
 
O
,
Matot
 
I
.
Perioperative acute kidney injury
.
Br J Anaesth
 
2015
;
115
:
ii3
ii14
.

554

Zealley
 
I
,
Wang
 
H
,
Donnan
 
PT
,
Bell
 
S
.
Exposure to contrast media in the perioperative period confers no additional risk of acute kidney injury in surgical patients
.
Nephrol Dial Transplant
 
2018
;
33
:
1751
1756
.

555

STARSurg Collaborative
.
Perioperative intravenous contrast administration and the incidence of acute kidney injury after major gastrointestinal surgery: prospective, multicentre cohort study
.
Br J Surg
 
2020
;
107
:
1023
1032
.

556

Meersch
 
M
,
Schmidt
 
C
,
Zarbock
 
A
.
Patient with chronic renal failure undergoing surgery
.
Curr Opin Anaesthesiol
 
2016
;
29
:
413
420
.

557

NCD Risk Factor Collaboration
.
Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults
.
Lancet
 
2017
;
390
:
2627
2642
.

558

Lavie
 
CJ
,
Arena
 
R
,
Alpert
 
MA
,
Milani
 
RV
,
Ventura
 
HO
.
Management of cardiovascular diseases in patients with obesity
.
Nat Rev Cardiol
 
2018
;
15
:
45
56
.

559

Ortega
 
FB
,
Lavie
 
CJ
,
Blair
 
SN
.
Obesity and cardiovascular disease
.
Circ Res
 
2016
;
118
:
1752
1770
.

560

De Hert
 
S
,
Staender
 
S
,
Fritsch
 
G
,
Hinkelbein
 
J
,
Afshari
 
A
,
Bettelli
 
G
, et al.  
Pre-operative evaluation of adults undergoing elective noncardiac surgery: updated guideline from the European Society of Anaesthesiology
.
Eur J Anaesthesiol
 
2018
;
35
:
407
465
.

561

Lavie
 
CJ
,
De Schutter
 
A
,
Parto
 
P
,
Jahangir
 
E
,
Kokkinos
 
P
,
Ortega
 
FB
, et al.  
Obesity and prevalence of cardiovascular diseases and prognosis – the obesity paradox updated
.
Prog Cardiovasc Dis
 
2016
;
58
:
537
547
.

562

Elagizi
 
A
,
Kachur
 
S
,
Lavie
 
CJ
,
Carbone
 
S
,
Pandey
 
A
,
Ortega
 
FB
, et al.  
An overview and update on obesity and the obesity paradox in cardiovascular diseases
.
Prog Cardiovasc Dis
 
2018
;
61
:
142
150
.

563

Valentijn
 
TM
,
Galal
 
W
,
Tjeertes
 
EK
,
Hoeks
 
SE
,
Verhagen
 
HJ
,
Stolker
 
RJ
.
The obesity paradox in the surgical population
.
Surgeon
 
2013
;
11
:
169
176
.

564

Hidvegi
 
R
,
Puelacher
 
C
,
Gualandro
 
DM
,
Lampart
 
A
,
Lurati Buse
 
G
,
Hammerer-Lerchner
 
A
, et al.  
Obesity paradox and perioperative myocardial infarction/injury in non-cardiac surgery
.
Clin Res Cardiol
 
2020
;
109
:
1140
1147
.

565

Ross
 
R
,
Blair
 
SN
,
Arena
 
R
,
Church
 
TS
,
Despres
 
JP
,
Franklin
 
BA
, et al.  
Importance of assessing cardiorespiratory fitness in clinical practice: a case for fitness as a clinical vital sign: a scientific statement from the American Heart Association
.
Circulation
 
2016
;
134
:
e653
e699
.

566

McAuley
 
PA
,
Artero
 
EG
,
Sui
 
X
,
Lee
 
DC
,
Church
 
TS
,
Lavie
 
CJ
, et al.  
The obesity paradox, cardiorespiratory fitness, and coronary heart disease
.
Mayo Clin Proc
 
2012
;
87
:
443
451
.

567

Pedersen
 
BK
.
Body mass index-independent effect of fitness and physical activity for all-cause mortality
.
Scand J Med Sci Sports
 
2007
;
17
:
196
204
.

568

Roman
 
M
,
Monaghan
 
A
,
Serraino
 
GF
,
Miller
 
D
,
Pathak
 
S
,
Lai
 
F
, et al.  
Meta-analysis of the influence of lifestyle changes for preoperative weight loss on surgical outcomes
.
Br J Surg
 
2019
;
106
:
181
189
.

569

McCullough
 
PA
,
Gallagher
 
MJ
,
Dejong
 
AT
,
Sandberg
 
KR
,
Trivax
 
JE
,
Alexander
 
D
, et al.  
Cardiorespiratory fitness and short-term complications after bariatric surgery
.
Chest
 
2006
;
130
:
517
525
.

570

Smith
 
TB
,
Stonell
 
C
,
Purkayastha
 
S
,
Paraskevas
 
P
.
Cardiopulmonary exercise testing as a risk assessment method in non cardio-pulmonary surgery: a systematic review
.
Anaesthesia
 
2009
;
64
:
883
893
.

571

Emerging Risk Factors Collaboration
,
Sarwar
 
N
,
Gao
 
P
,
Seshasai
 
SR
,
Gobin
 
R
,
Kaptoge
 
S
, et al.  
Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies
.
Lancet
 
2010
;
375
:
2215
2222
.

572

Kadoi
 
Y
.
Anesthetic considerations in diabetic patients. Part II: intraoperative and postoperative management of patients with diabetes mellitus
.
J Anesth
 
2010
;
24
:
748
756
.

573

Cosentino
 
F
,
Grant
 
PJ
,
Aboyans
 
V
,
Bailey
 
CJ
,
Ceriello
 
A
,
Delgado
 
V
, et al.  
2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD
.
Eur Heart J
 
2020
;
41
:
255
323
.

574

American Diabetes Association
.
15. Diabetes care in the hospital: standards of medical care in diabetes - 2020
.
Diabetes Care
 
2021
;
44
:
S211
S220
.

575

Shah
 
NJ
,
Leis
 
A
,
Kheterpal
 
S
,
Englesbe
 
MJ
,
Kumar
 
SS
.
Association of intraoperative hyperglycemia and postoperative outcomes in patients undergoing non-cardiac surgery: a multicenter retrospective study
.
BMC Anesthesiol
 
2020
;
20
:
106
.

576

Aharaz
 
A
,
Pottegard
 
A
,
Henriksen
 
DP
,
Hallas
 
J
,
Beck-Nielsen
 
H
,
Lassen
 
AT
.
Risk of lactic acidosis in type 2 diabetes patients using metformin: a case control study
.
PLoS One
 
2018
;
13
:
e0196122
.

577

Vogt
 
AP
,
Bally
 
L
.
Perioperative glucose management: current status and future directions
.
Best Pract Res Clin Anaesthesiol
 
2020
;
34
:
213
224
.

578

Pasquel
 
FJ
,
Gomez-Huelgas
 
R
,
Anzola
 
I
,
Oyedokun
 
F
,
Haw
 
JS
,
Vellanki
 
P
, et al.  
Predictive value of admission hemoglobin A1c on inpatient glycemic control and response to insulin therapy in medicine and surgery patients with type 2 diabetes
.
Diabetes Care
 
2015
;
38
:
e202
e203
.

579

Carpenter
 
DL
,
Gregg
 
SR
,
Xu
 
K
,
Buchman
 
TG
,
Coopersmith
 
CM
.
Prevalence and impact of unknown diabetes in the ICU
.
Crit Care Med
 
2015
;
43
:
e541
e550
.

580

van den Boom
 
W
,
Schroeder
 
RA
,
Manning
 
MW
,
Setji
 
TL
,
Fiestan
 
GO
,
Dunson
 
DB
.
Effect of A1C and glucose on postoperative mortality in noncardiac and cardiac surgeries
.
Diabetes Care
 
2018
;
41
:
782
788
.

581

Dhatariya
 
K
,
Levy
 
N
,
Kilvert
 
A
,
Watson
 
B
,
Cousins
 
D
,
Flanagan
 
D
, et al.  
NHS Diabetes guideline for the perioperative management of the adult patient with diabetes
.
Diabet Med
 
2012
;
29
:
420
433
.

582

Bergqvist
 
D
,
Agnelli
 
G
,
Cohen
 
AT
,
Eldor
 
A
,
Nilsson
 
PE
,
Le Moigne-Amrani
 
A
, et al.  
Duration of prophylaxis against venous thromboembolism with enoxaparin after surgery for cancer
.
N Engl J Med
 
2002
;
346
:
975
980
.

583

Kirschner
 
M
,
do
 
OHN
,
Parmentier
 
S
,
Hart
 
C
,
Henze
 
L
,
Bisping
 
G
, et al.  
Primary thromboprophylaxis in patients with malignancies: Daily Practice recommendations by the Hemostasis Working Party of the German Society of Hematology and Medical Oncology (DGHO), the Society of Thrombosis and Hemostasis Research (GTH), and the Austrian Society of Hematology and Oncology (OGHO)
.
Cancers (Basel)
 
2021
;
13
:
2905
.

584

ESC
.
2022 ESC Guidelines on cardio-oncology
.
Eur Heart J
 
2022
;
XX
:
XX
XX
.

585

COVIDSurg Collaborative, GlobalSurg Collaborative
.
Timing of surgery following SARS-CoV-2 infection: an international prospective cohort study
.
Anaesthesia
 
2021
;
76
:
748
758
.

586

COVIDSurg Collaborative, GlobalSurg Collaborative
.
SARS-CoV-2 infection and venous thromboembolism after surgery: an international prospective cohort study
.
Anaesthesia
 
2022
;
77
:
28
39
.

587

Guzik
 
TJ
,
Mohiddin
 
SA
,
Dimarco
 
A
,
Patel
 
V
,
Savvatis
 
K
,
Marelli-Berg
 
FM
, et al.  
COVID-19 and the cardiovascular system: implications for risk assessment, diagnosis, and treatment options
.
Cardiovasc Res
 
2020
;
116
:
1666
1687
.

588

Puntmann
 
VO
,
Carerj
 
ML
,
Wieters
 
I
,
Fahim
 
M
,
Arendt
 
C
,
Hoffmann
 
J
, et al.  
Outcomes of cardiovascular magnetic resonance imaging in patients recently recovered from coronavirus disease 2019 (COVID-19)
.
JAMA Cardiol
 
2020
;
5
:
1265
1273
.

589

Rohatgi
 
N
,
Smilowitz
 
NR
,
Reejhsinghani
 
R
.
Perioperative cardiovascular considerations prior to elective noncardiac surgery in patients with a history of COVID-19
.
JAMA Surg
 
2022
;
157
:
187
188
.

590

Deng
 
JZ
,
Chan
 
JS
,
Potter
 
AL
,
Chen
 
YW
,
Sandhu
 
HS
,
Panda
 
N
, et al.  
The risk of postoperative complications after major elective surgery in active or resolved COVID-19 in the United States
.
Ann Surg
 
2022
;
275
:
242
246
.

591

Anesthesia Patient Safety Foundation
.
American Society of Anesthesiologists and Anesthesia Patient Safety Foundation Joint Statement on Elective Surgery and Anesthesia for Patients after COVID-19 Infection
. https://www.apsf.org/news-updates/asa-and-apsf-joint-statement-on-elective-surgery-and-anesthesia-for-patients-after-covid-19-infection/  
(30 March 2022)
.

592

Mellin-Olsen
 
J
,
Staender
 
S
,
Whitaker
 
DK
,
Smith
 
AF
.
The Helsinki Declaration on patient safety in anaesthesiology
.
Eur J Anaesthesiol
 
2010
;
27
:
592
597
.

593

De Hert
 
S
. Perioperative monitoring: anaesthesiology. In:
Camm
 
AJ
 
Lüscher
 
TF
 
Maurer
 
G
and
Serruys
 
PW
, editors.
ESC CardioMed
. 3rd ed.
Oxford
:
Oxford University Press
;
2018
. p
2683
2686
.

594

Hamilton-Davies
 
C
,
Mythen
 
MG
,
Salmon
 
JB
,
Jacobson
 
D
,
Shukla
 
A
,
Webb
 
AR
.
Comparison of commonly used clinical indicators of hypovolaemia with gastrointestinal tonometry
.
Intensive Care Med
 
1997
;
23
:
276
281
.

595

Marik
 
PE
,
Cavallazzi
 
R
.
Does the central venous pressure predict fluid responsiveness? An updated meta-analysis and a plea for some common sense
.
Crit Care Med
 
2013
;
41
:
1774
1781
.

596

Gaba
 
DM
.
Improving anesthesiologists’ performance by simulating reality
.
Anesthesiology
 
1992
;
76
:
491
494
.

597

Runciman
 
WB
.
Commentary on equipment recommendations
.
Eur J Anaesthesiol Suppl
 
1993
;
7
:
16
18
.

598

Block
 
FE
, Jr.,
Nuutinen
 
L
,
Ballast
 
B
.
Optimization of alarms: a study on alarm limits, alarm sounds, and false alarms, intended to reduce annoyance
.
J Clin Monit Comput
 
1999
;
15
:
75
83
.

599

De Hert
 
S
,
Moerman
 
A
.
Anesthetic preconditioning: have we found the holy grail of perioperative cardioprotection?
 
J Cardiothorac Vasc Anesth
 
2018
;
32
:
1135
1136
.

600

Bijker
 
JB
,
van Klei
 
WA
,
Kappen
 
TH
,
van Wolfswinkel
 
L
,
Moons
 
KG
,
Kalkman
 
CJ
.
Incidence of intraoperative hypotension as a function of the chosen definition: literature definitions applied to a retrospective cohort using automated data collection
.
Anesthesiology
 
2007
;
107
:
213
220
.

601

Vernooij
 
LM
,
van Klei
 
WA
,
Machina
 
M
,
Pasma
 
W
,
Beattie
 
WS
,
Peelen
 
LM
.
Different methods of modelling intraoperative hypotension and their association with postoperative complications in patients undergoing non-cardiac surgery
.
Br J Anaesth
 
2018
;
120
:
1080
1089
.

602

Sessler
 
DI
,
Bloomstone
 
JA
,
Aronson
 
S
,
Berry
 
C
,
Gan
 
TJ
,
Kellum
 
JA
, et al.  
Perioperative Quality Initiative consensus statement on intraoperative blood pressure, risk and outcomes for elective surgery
.
Br J Anaesth
 
2019
;
122
:
563
574
.

603

Sessler
 
DI
,
Meyhoff
 
CS
,
Zimmerman
 
NM
,
Mao
 
G
,
Leslie
 
K
,
Vasquez
 
SM
, et al.  
Period-dependent associations between hypotension during and for four days after noncardiac surgery and a composite of myocardial infarction and death: a substudy of the POISE-2 trial
.
Anesthesiology
 
2018
;
128
:
317
327
.

604

Ladha
 
KS
,
Beattie
 
WS
,
Tait
 
G
,
Wijeysundera
 
DN
.
Association between preoperative ambulatory heart rate and postoperative myocardial injury: a retrospective cohort study
.
Br J Anaesth
 
2018
;
121
:
722
729
.

605

Abbott
 
TE
,
Ackland
 
GL
,
Archbold
 
RA
,
Wragg
 
A
,
Kam
 
E
,
Ahmad
 
T
, et al.  
Preoperative heart rate and myocardial injury after non-cardiac surgery: results of a predefined secondary analysis of the VISION study
.
Br J Anaesth
 
2016
;
117
:
172
181
.

606

Abbott
 
TEF
,
Minto
 
G
,
Lee
 
AM
,
Pearse
 
RM
,
Ackland
 
GL
.,
POM-HR, POMO-O and OPTIMISE study groups
.
Elevated preoperative heart rate is associated with cardiopulmonary and autonomic impairment in high-risk surgical patients
.
Br J Anaesth
 
2017
;
119
:
87
94
.

607

Ruetzler
 
K
,
Yilmaz
 
HO
,
Turan
 
A
,
Zimmerman
 
NM
,
Mao
 
G
,
Hung
 
MH
, et al.  
Intra-operative tachycardia is not associated with a composite of myocardial injury and mortality after noncardiac surgery: a retrospective cohort analysis
.
Eur J Anaesthesiol
 
2019
;
36
:
105
113
.

608

Straarup
 
TS
,
Hausenloy
 
DJ
,
Rolighed Larsen
 
JK
.
Cardiac troponins and volatile anaesthetics in coronary artery bypass graft surgery: a systematic review, meta-analysis and trial sequential analysis
.
Eur J Anaesthesiol
 
2016
;
33
:
396
407
.

609

Landoni
 
G
,
Lomivorotov
 
VV
,
Nigro Neto
 
C
,
Monaco
 
F
,
Pasyuga
 
VV
,
Bradic
 
N
, et al.  
Volatile anesthetics versus total intravenous anesthesia for cardiac surgery
.
N Engl J Med
 
2019
;
380
:
1214
1225
.

610

Lurati Buse
 
GA
,
Schumacher
 
P
,
Seeberger
 
E
,
Studer
 
W
,
Schuman
 
RM
,
Fassl
 
J
, et al.  
Randomized comparison of sevoflurane versus propofol to reduce perioperative myocardial ischemia in patients undergoing noncardiac surgery
.
Circulation
 
2012
;
126
:
2696
2704
.

611

Rodgers
 
A
,
Walker
 
N
,
Schug
 
S
,
McKee
 
A
,
Kehlet
 
H
,
van Zundert
 
A
, et al.  
Reduction of postoperative mortality and morbidity with epidural or spinal anaesthesia: results from overview of randomised trials
.
BMJ
 
2000
;
321
:
1493
.

612

Bos
 
EME
,
Hollmann
 
MW
,
Lirk
 
P
.
Safety and efficacy of epidural analgesia
.
Curr Opin Anaesthesiol
 
2017
;
30
:
736
742
.

613

Gogarten
 
W
,
Vandermeulen
 
E
,
Van Aken
 
H
,
Kozek
 
S
,
Llau
 
JV
,
Samama
 
CM
, et al.  
Regional anaesthesia and antithrombotic agents: recommendations of the European Society of Anaesthesiology
.
Eur J Anaesthesiol
 
2010
;
27
:
999
1015
.

614

Hamilton
 
MA
,
Cecconi
 
M
,
Rhodes
 
A
.
A systematic review and meta-analysis on the use of preemptive hemodynamic intervention to improve postoperative outcomes in moderate and high-risk surgical patients
.
Anesth Analg
 
2011
;
112
:
1392
1402
.

615

Grocott
 
MP
,
Dushianthan
 
A
,
Hamilton
 
MA
,
Mythen
 
MG
,
Harrison
 
D
,
Rowan
 
K
, et al.  
Perioperative increase in global blood flow to explicit defined goals and outcomes following surgery
.
Cochrane Database Syst Rev
 
2012
;
11
:
CD004082
.

616

Cecconi
 
M
,
Corredor
 
C
,
Arulkumaran
 
N
,
Abuella
 
G
,
Ball
 
J
,
Grounds
 
RM
, et al.  
Clinical review: goal-directed therapy – what is the evidence in surgical patients? The effect on different risk groups
.
Crit Care
 
2013
;
17
:
209
.

617

Arulkumaran
 
N
,
Corredor
 
C
,
Hamilton
 
MA
,
Ball
 
J
,
Grounds
 
RM
,
Rhodes
 
A
, et al.  
Cardiac complications associated with goal-directed therapy in high-risk surgical patients: a meta-analysis
.
Br J Anaesth
 
2014
;
112
:
648
659
.

618

Nicklas
 
JY
,
Diener
 
O
,
Leistenschneider
 
M
,
Sellhorn
 
C
,
Schon
 
G
,
Winkler
 
M
, et al.  
Personalised haemodynamic management targeting baseline cardiac index in high-risk patients undergoing major abdominal surgery: a randomised single-centre clinical trial
.
Br J Anaesth
 
2020
;
125
:
122
132
.

619

Taenzer
 
AH
,
Pyke
 
JB
,
McGrath
 
SP
.
A review of current and emerging approaches to address failure-to-rescue
.
Anesthesiology
 
2011
;
115
:
421
431
.

620

Pyke
 
J
,
Taenzer
 
AH
,
Renaud
 
CE
,
McGrath
 
SP
.
Developing a continuous monitoring infrastructure for detection of inpatient deterioration
.
Jt Comm J Qual Patient Saf
 
2012
;
38
:
428
431
.

621

McGrath
 
SP
,
Taenzer
 
AH
,
Karon
 
N
,
Blike
 
G
.
Surveillance monitoring management for general care units: strategy, design, and implementation
.
Jt Comm J Qual Patient Saf
 
2016
;
42
:
293
302
.

622

Taenzer
 
AH
,
Spence
 
BC
.
The afferent limb of rapid response systems: continuous monitoring on general care units
.
Crit Care Clin
 
2018
;
34
:
189
198
.

623

Liu
 
SS
,
Wu
 
CL
.
The effect of analgesic technique on postoperative patient-reported outcomes including analgesia: a systematic review
.
Anesth Analg
 
2007
;
105
:
789
808
.

624

White
 
PF
,
Kehlet
 
H
.
Postoperative pain management and patient outcome: time to return to work!
.
Anesth Analg
 
2007
;
104
:
487
489
.

625

Turan
 
A
,
Leung
 
S
,
Bajracharya
 
GR
,
Babazade
 
R
,
Barnes
 
T
,
Schacham
 
YN
, et al.  
Acute postoperative pain is associated with myocardial injury after noncardiac surgery
.
Anesth Analg
 
2020
;
131
:
822
829
.

626

Olsen
 
AM
,
Fosbol
 
EL
,
Lindhardsen
 
J
,
Folke
 
F
,
Charlot
 
M
,
Selmer
 
C
, et al.  
Long-term cardiovascular risk of nonsteroidal anti-inflammatory drug use according to time passed after first-time myocardial infarction: a nationwide cohort study
.
Circulation
 
2012
;
126
:
1955
1963
.

627

Schmidt
 
M
,
Sorensen
 
HT
,
Pedersen
 
L
.
Diclofenac use and cardiovascular risks: series of nationwide cohort studies
.
BMJ
 
2018
;
362
:
k3426
.

628

Farkouh
 
ME
,
Greenberg
 
BP
.
An evidence-based review of the cardiovascular risks of nonsteroidal anti-inflammatory drugs
.
Am J Cardiol
 
2009
;
103
:
1227
1237
.

629

Ray
 
WA
,
Varas-Lorenzo
 
C
,
Chung
 
CP
,
Castellsague
 
J
,
Murray
 
KT
,
Stein
 
CM
, et al.  
Cardiovascular risks of nonsteroidal antiinflammatory drugs in patients after hospitalization for serious coronary heart disease
.
Circ Cardiovasc Qual Outcomes
 
2009
;
2
:
155
163
.

630

Antman
 
EM
,
Bennett
 
JS
,
Daugherty
 
A
,
Furberg
 
C
,
Roberts
 
H
,
Taubert
 
KA
, et al.  
Use of nonsteroidal antiinflammatory drugs: an update for clinicians: a scientific statement from the American Heart Association
.
Circulation
 
2007
;
115
:
1634
1642
.

631

Schug
 
SA
,
Joshi
 
GP
,
Camu
 
F
,
Pan
 
S
,
Cheung
 
R
.
Cardiovascular safety of the cyclooxygenase-2 selective inhibitors parecoxib and valdecoxib in the postoperative setting: an analysis of integrated data
.
Anesth Analg
 
2009
;
108
:
299
307
.

632

Liu
 
SS
,
Bae
 
JJ
,
Bieltz
 
M
,
Ma
 
Y
,
Memtsoudis
 
S
.
Association of perioperative use of nonsteroidal anti-inflammatory drugs with postoperative myocardial infarction after total joint replacement
.
Reg Anesth Pain Med
 
2012
;
37
:
45
50
.

633

Schmidt
 
M
,
Lamberts
 
M
,
Olsen
 
AM
,
Fosboll
 
E
,
Niessner
 
A
,
Tamargo
 
J
, et al.  
Cardiovascular safety of non-aspirin non-steroidal anti-inflammatory drugs: review and position paper by the working group for Cardiovascular Pharmacotherapy of the European Society of Cardiology
.
Eur Heart J
 
2016
;
37
:
1015
1023
.

634

Gregory
 
A
,
Stapelfeldt
 
WH
,
Khanna
 
AK
,
Smischney
 
NJ
,
Boero
 
IJ
,
Chen
 
Q
, et al.  
Intraoperative hypotension is associated with adverse clinical outcomes after noncardiac surgery
.
Anesth Analg
 
2021
;
132
:
1654
1665
.

635

Aldington
 
S
,
Shirtcliffe
 
P
,
Weatherall
 
M
,
Beasley
 
R
.
Increased risk of cardiovascular events with parecoxib/valdecoxib: a systematic review and meta-analysis
.
N Z Med J
 
2005
;
118
:
U1755
.

636

Devereaux
 
PJ
,
Sessler
 
DI
.
Cardiac complications in patients undergoing major noncardiac surgery
.
N Engl J Med
 
2015
;
373
:
2258
2269
.

637

Sabate
 
S
,
Mases
 
A
,
Guilera
 
N
,
Canet
 
J
,
Castillo
 
J
,
Orrego
 
C
, et al.  
Incidence and predictors of major perioperative adverse cardiac and cerebrovascular events in non-cardiac surgery
.
Br J Anaesth
 
2011
;
107
:
879
890
.

638

Smilowitz
 
NR
,
Redel-Traub
 
G
,
Hausvater
 
A
,
Armanious
 
A
,
Nicholson
 
J
,
Puelacher
 
C
, et al.  
Myocardial injury after noncardiac surgery: a systematic review and meta-analysis
.
Cardiol Rev
 
2019
;
27
:
267
273
.

639

Smilowitz
 
NR
,
Gupta
 
N
,
Guo
 
Y
,
Berger
 
JS
,
Bangalore
 
S
.
Perioperative acute myocardial infarction associated with non-cardiac surgery
.
Eur Heart J
 
2017
;
38
:
2409
2417
.

640

Master
 
AM
,
Dack
 
S
,
Jaffe
 
HL
.
Postoperative coronary artery occlusion
.
JAMA
 
1938
;
110
:
1415
1418
.

641

Devereaux
 
PJ
,
Xavier
 
D
,
Pogue
 
J
,
Guyatt
 
G
,
Sigamani
 
A
,
Garutti
 
I
, et al.  
Characteristics and short-term prognosis of perioperative myocardial infarction in patients undergoing noncardiac surgery: a cohort study
.
Ann Intern Med
 
2011
;
154
:
523
528
.

642

Borges
 
FK
,
Devereaux
 
PJ
.
Physicians should obtain perioperative cardiac troponin measurements in at-risk patients undergoing noncardiac surgery
.
Clin Chem
 
2021
;
67
:
50
53
.

643

Thygesen
 
K
,
Alpert
 
JS
,
Jaffe
 
AS
,
Chaitman
 
BR
,
Bax
 
JJ
,
Morrow
 
DA
, et al.  
Fourth Universal Definition of Myocardial Infarction (2018)
.
Circulation
 
2018
;
138
:
e618
e651
.

644

Toda
 
H
,
Nakamura
 
K
,
Shimizu
 
K
,
Ejiri
 
K
,
Iwano
 
T
,
Miyoshi
 
T
, et al.  
Effects of bisoprolol transdermal patches for prevention of perioperative myocardial injury in high-risk patients undergoing non-cardiac surgery: multicenter randomized controlled study
.
Circ J
 
2020
;
84
:
642
649
.

645

Devereaux
 
PJ
,
Szczeklik
 
W
.
Myocardial injury after non-cardiac surgery: diagnosis and management
.
Eur Heart J
 
2020
;
41
:
3083
3091
.

646

Ruetzler
 
K
,
Smilowitz
 
NR
,
Berger
 
JS
,
Devereaux
 
PJ
,
Maron
 
BA
,
Newby
 
LK
, et al.  
Diagnosis and management of patients with myocardial injury after noncardiac surgery: A scientific statement from the American Heart Association
.
Circulation
 
2021
;
144
:
e287
e305
.

647

Borges
 
FK
,
Sheth
 
T
,
Patel
 
A
,
Marcucci
 
M
,
Yung
 
T
,
Langer
 
T
, et al.  
Accuracy of physicians in differentiating type 1 and type 2 myocardial infarction based on clinical information
.
CJC Open
 
2020
;
2
:
577
584
.

648

Gualandro
 
DM
,
Puelacher
 
C
,
Lurati Buse
 
G
,
Glarner
 
N
,
Cardozo
 
FA
,
Vogt
 
R
, et al.  
Incidence and outcomes of perioperative myocardial infarction/injury diagnosed by high-sensitivity cardiac troponin I
.
Clin Res Cardiol
 
2021
;
110
:
1450
1463
.

649

Park
 
J
,
Oh
 
AR
,
Kwon
 
JH
,
Kim
 
S
,
Kim
 
J
,
Yang
 
K
, et al.  
Association between cardiologist evaluation and mortality in myocardial injury after non-cardiac surgery
.
Heart
 
2021
.

650

Devereaux
 
PJ
,
Duceppe
 
E
,
Guyatt
 
G
,
Tandon
 
V
,
Rodseth
 
R
,
Biccard
 
BM
, et al.  
Dabigatran in patients with myocardial injury after non-cardiac surgery (MANAGE): an international, randomised, placebo-controlled trial
.
Lancet
 
2018
;
391
:
2325
2334
.

651

Ponikowski
 
P
,
Voors
 
AA
,
Anker
 
SD
,
Bueno
 
H
,
Cleland
 
JGF
,
Coats
 
AJS
, et al.  
2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the Heart Failure Association (HFA) of the ESC
.
Eur Heart J
 
2016
;
37
:
2129
2200
.

652

Konstantinides
 
SV
,
Meyer
 
G
,
Becattini
 
C
,
Bueno
 
H
,
Geersing
 
GJ
,
Harjola
 
VP
, et al.  
2019 ESC Guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European Respiratory Society (ERS)
.
Eur Heart J
 
2020
;
41
:
543
603
.

653

Marti
 
C
,
John
 
G
,
Konstantinides
 
S
,
Combescure
 
C
,
Sanchez
 
O
,
Lankeit
 
M
, et al.  
Systemic thrombolytic therapy for acute pulmonary embolism: a systematic review and meta-analysis
.
Eur Heart J
 
2015
;
36
:
605
614
.

654

Neely
 
RC
,
Byrne
 
JG
,
Gosev
 
I
,
Cohn
 
LH
,
Javed
 
Q
,
Rawn
 
JD
, et al.  
Surgical embolectomy for acute massive and submassive pulmonary embolism in a series of 115 patients
.
Ann Thorac Surg
 
2015
;
100
:
1245
1251
;
discussion 1251–1242
.

655

Conen
 
D
,
Alonso-Coello
 
P
,
Douketis
 
J
,
Chan
 
MTV
,
Kurz
 
A
,
Sigamani
 
A
, et al.  
Risk of stroke and other adverse outcomes in patients with perioperative atrial fibrillation 1 year after non-cardiac surgery
.
Eur Heart J
 
2020
;
41
:
645
651
.

656

AlTurki
 
A
,
Marafi
 
M
,
Proietti
 
R
,
Cardinale
 
D
,
Blackwell
 
R
,
Dorian
 
P
, et al.  
Major adverse cardiovascular events associated with postoperative atrial fibrillation after noncardiac surgery: a systematic review and meta-analysis
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e007437
.

657

Chebbout
 
R
,
Heywood
 
EG
,
Drake
 
TM
,
Wild
 
JRL
,
Lee
 
J
,
Wilson
 
M
, et al.  
A systematic review of the incidence of and risk factors for postoperative atrial fibrillation following general surgery
.
Anaesthesia
 
2018
;
73
:
490
498
.

658

Albini
 
A
,
Malavasi
 
VL
,
Vitolo
 
M
,
Imberti
 
JF
,
Marietta
 
M
,
Lip
 
GYH
, et al.  
Long-term outcomes of postoperative atrial fibrillation following non cardiac surgery: a systematic review and metanalysis
.
Eur J Intern Med
 
2021
;
85
:
27
33
.

659

Hyun
 
J
,
Cho
 
MS
,
Nam
 
GB
,
Kim
 
M
,
Do
 
U
,
Kim
 
J
, et al.  
Natural course of new-onset postoperative atrial fibrillation after noncardiac surgery
.
J Am Heart Assoc
 
2021
;
10
:
e018548
.

660

Heywood
 
EG
,
Drake
 
TM
,
Bradburn
 
M
,
Lee
 
J
,
Wilson
 
MJ
,
Lee
 
MJ
.
Atrial fibrillation after gastrointestinal surgery: incidence and associated risk factors
.
J Surg Res
 
2019
;
238
:
23
28
.

661

Lin
 
MH
,
Kamel
 
H
,
Singer
 
DE
,
Wu
 
YL
,
Lee
 
M
,
Ovbiagele
 
B
.
Perioperative/postoperative atrial fibrillation and risk of subsequent stroke and/or mortality
.
Stroke
 
2019
;
50
:
1364
1371
.

662

Dobrev
 
D
,
Aguilar
 
M
,
Heijman
 
J
,
Guichard
 
JB
,
Nattel
 
S
.
Postoperative atrial fibrillation: mechanisms, manifestations and management
.
Nat Rev Cardiol
 
2019
;
16
:
417
436
.

663

Mathew
 
JP
,
Fontes
 
ML
,
Tudor
 
IC
,
Ramsay
 
J
,
Duke
 
P
,
Mazer
 
CD
, et al.  
A multicenter risk index for atrial fibrillation after cardiac surgery
.
JAMA
 
2004
;
291
:
1720
1729
.

664

Villareal
 
RP
,
Hariharan
 
R
,
Liu
 
BC
,
Kar
 
B
,
Lee
 
VV
,
Elayda
 
M
, et al.  
Postoperative atrial fibrillation and mortality after coronary artery bypass surgery
.
J Am Coll Cardiol
 
2004
;
43
:
742
748
.

665

Cardinale
 
D
,
Sandri
 
MT
,
Colombo
 
A
,
Salvatici
 
M
,
Tedeschi
 
I
,
Bacchiani
 
G
, et al.  
Prevention of atrial fibrillation in high-risk patients undergoing lung cancer surgery: the PRESAGE Trial
.
Ann Surg
 
2016
;
264
:
244
251
.

666

Arsenault
 
KA
,
Yusuf
 
AM
,
Crystal
 
E
,
Healey
 
JS
,
Morillo
 
CA
,
Nair
 
GM
, et al.  
Interventions for preventing post-operative atrial fibrillation in patients undergoing heart surgery
.
Cochrane Database Syst Rev
 
2013
;
2013
:
CD003611
.

667

Ozaydin
 
M
,
Icli
 
A
,
Yucel
 
H
,
Akcay
 
S
,
Peker
 
O
,
Erdogan
 
D
, et al.  
Metoprolol vs. carvedilol or carvedilol plus N-acetyl cysteine on post-operative atrial fibrillation: a randomized, double-blind, placebo-controlled study
.
Eur Heart J
 
2013
;
34
:
597
604
.

668

O'Neal
 
JB
,
Billings
 
F
,
Liu
 
X
,
Shotwell
 
MS
,
Liang
 
Y
,
Shah
 
AS
, et al.  
Effect of preoperative beta-blocker use on outcomes following cardiac surgery
.
Am J Cardiol
 
2017
;
120
:
1293
1297
.

669

Buckley
 
MS
,
Nolan
 
PE
, Jr.
,
Slack
 
MK
,
Tisdale
 
JE
,
Hilleman
 
DE
,
Copeland
 
JG
.
Amiodarone prophylaxis for atrial fibrillation after cardiac surgery: meta-analysis of dose response and timing of initiation
.
Pharmacotherapy
 
2007
;
27
:
360
368
.

670

Riber
 
LP
,
Christensen
 
TD
,
Jensen
 
HK
,
Hoejsgaard
 
A
,
Pilegaard
 
HK
.
Amiodarone significantly decreases atrial fibrillation in patients undergoing surgery for lung cancer
.
Ann Thorac Surg
 
2012
;
94
:
339
344
;
discussion 345–336
.

671

Tisdale
 
JE
,
Wroblewski
 
HA
,
Wall
 
DS
,
Rieger
 
KM
,
Hammoud
 
ZT
,
Young
 
JV
, et al.  
A randomized trial evaluating amiodarone for prevention of atrial fibrillation after pulmonary resection
.
Ann Thorac Surg
 
2009
;
88
:
886
893
;
discussion 894–885
.

672

Yuan
 
X
,
Du
 
J
,
Liu
 
Q
,
Zhang
 
L
.
Defining the role of perioperative statin treatment in patients after cardiac surgery: a meta-analysis and systematic review of 20 randomized controlled trials
.
Int J Cardiol
 
2017
;
228
:
958
966
.

673

Zheng
 
Z
,
Jayaram
 
R
,
Jiang
 
L
,
Emberson
 
J
,
Zhao
 
Y
,
Li
 
Q
, et al.  
Perioperative rosuvastatin in cardiac surgery
.
N Engl J Med
 
2016
;
374
:
1744
1753
.

674

Fairley
 
JL
,
Zhang
 
L
,
Glassford
 
NJ
,
Bellomo
 
R
.
Magnesium status and magnesium therapy in cardiac surgery: a systematic review and meta-analysis focusing on arrhythmia prevention
.
J Crit Care
 
2017
;
42
:
69
77
.

675

Tabbalat
 
RA
,
Hamad
 
NM
,
Alhaddad
 
IA
,
Hammoudeh
 
A
,
Akasheh
 
BF
,
Khader
 
Y
.
Effect of ColchiciNe on the InciDence of atrial fibrillation in open heart surgery patients: END-AF trial
.
Am Heart J
 
2016
;
178
:
102
107
.

676

Wang
 
W
,
Mei
 
YQ
,
Yuan
 
XH
,
Feng
 
XD
.
Clinical efficacy of epicardial application of drug-releasing hydrogels to prevent postoperative atrial fibrillation
.
J Thorac Cardiovasc Surg
 
2016
;
151
:
80
85
.

677

Dieleman
 
JM
,
Nierich
 
AP
,
Rosseel
 
PM
,
van der Maaten
 
JM
,
Hofland
 
J
,
Diephuis
 
JC
, et al.  
Intraoperative high-dose dexamethasone for cardiac surgery: a randomized controlled trial
.
JAMA
 
2012
;
308
:
1761
1767
.

678

Whitlock
 
RP
,
Devereaux
 
PJ
,
Teoh
 
KH
,
Lamy
 
A
,
Vincent
 
J
,
Pogue
 
J
, et al.  
Methylprednisolone in patients undergoing cardiopulmonary bypass (SIRS): a randomised, double-blind, placebo-controlled trial
.
Lancet
 
2015
;
386
:
1243
1253
.

679

Kowey
 
PR
,
Dorian
 
P
,
Mitchell
 
LB
,
Pratt
 
CM
,
Roy
 
D
,
Schwartz
 
PJ
, et al.  
Vernakalant hydrochloride for the rapid conversion of atrial fibrillation after cardiac surgery: a randomized, double-blind, placebo-controlled trial
.
Circ Arrhythm Electrophysiol
 
2009
;
2
:
652
659
.

680

Gillinov
 
AM
,
Bagiella
 
E
,
Moskowitz
 
AJ
,
Raiten
 
JM
,
Groh
 
MA
,
Bowdish
 
ME
, et al.  
Rate control versus rhythm control for atrial fibrillation after cardiac surgery
.
N Engl J Med
 
2016
;
374
:
1911
1921
.

681

Saxena
 
A
,
Dinh
 
DT
,
Smith
 
JA
,
Shardey
 
GC
,
Reid
 
CM
,
Newcomb
 
AE
.
Usefulness of postoperative atrial fibrillation as an independent predictor for worse early and late outcomes after isolated coronary artery bypass grafting (multicenter Australian study of 19,497 patients)
.
Am J Cardiol
 
2012
;
109
:
219
225
.

682

Gialdini
 
G
,
Nearing
 
K
,
Bhave
 
PD
,
Bonuccelli
 
U
,
Iadecola
 
C
,
Healey
 
JS
, et al.  
Perioperative atrial fibrillation and the long-term risk of ischemic stroke
.
JAMA
 
2014
;
312
:
616
622
.

683

Horwich
 
P
,
Buth
 
KJ
,
Legare
 
JF
.
New onset postoperative atrial fibrillation is associated with a long-term risk for stroke and death following cardiac surgery
.
J Card Surg
 
2013
;
28
:
8
13
.

684

Ahlsson
 
A
,
Fengsrud
 
E
,
Bodin
 
L
,
Englund
 
A
.
Postoperative atrial fibrillation in patients undergoing aortocoronary bypass surgery carries an eightfold risk of future atrial fibrillation and a doubled cardiovascular mortality
.
Eur J Cardiothorac Surg
 
2010
;
37
:
1353
1359
.

685

Butt
 
JH
,
Xian
 
Y
,
Peterson
 
ED
,
Olsen
 
PS
,
Rorth
 
R
,
Gundlund
 
A
, et al.  
Long-term thromboembolic risk in patients with postoperative atrial fibrillation after coronary artery bypass graft surgery and patients with nonvalvular atrial fibrillation
.
JAMA Cardiol
 
2018
;
3
:
417
424
.

686

Butt
 
JH
,
Olesen
 
JB
,
Havers-Borgersen
 
E
,
Gundlund
 
A
,
Andersson
 
C
,
Gislason
 
GH
, et al.  
Risk of thromboembolism associated with atrial fibrillation following noncardiac surgery
.
J Am Coll Cardiol
 
2018
;
72
:
2027
2036
.

687

Selim
 
M
.
Perioperative stroke
.
N Engl J Med
 
2007
;
356
:
706
713
.

688

Macellari
 
F
,
Paciaroni
 
M
,
Agnelli
 
G
,
Caso
 
V
.
Perioperative stroke risk in nonvascular surgery
.
Cerebrovasc Dis
 
2012
;
34
:
175
181
.

689

Mashour
 
GA
,
Shanks
 
AM
,
Kheterpal
 
S
.
Perioperative stroke and associated mortality after noncardiac, nonneurologic surgery
.
Anesthesiology
 
2011
;
114
:
1289
1296
.

690

Ng
 
JL
,
Chan
 
MT
,
Gelb
 
AW
.
Perioperative stroke in noncardiac, nonneurosurgical surgery
.
Anesthesiology
 
2011
;
115
:
879
890
.

691

Walicka
 
M
,
Tuszynska
 
A
,
Chlebus
 
M
,
Sanchak
 
Y
,
Sliwczynski
 
A
,
Brzozowska
 
M
, et al.  
Predictors of in-hospital mortality in surgical wards: a multivariable retrospective cohort analysis of 2,800,069 hospitalizations
.
World J Surg
 
2021
;
45
:
480
487
.

692

Mattingly
 
AS
,
Lerman
 
BJ
,
Popat
 
R
,
Wren
 
SM
.
Association of sex with postoperative mortality among patients with heart failure who underwent elective noncardiac operations
.
JAMA Netw Open
 
2019
;
2
:
e1914420
.

693

World Health Organization
. Prevalence of anaemia in women of reproductive age (aged 15–49) (%). In:
Global Health Observatory
.
Geneva
:
World Health Organiziation
;
2022
. https://www.who.int/data/gho/data/indicators/indicator-details/GHO/prevalence-of-anaemia-in-women-of-reproductive-age-(-)

694

Gombotz
 
H
,
Schreier
 
G
,
Neubauer
 
S
,
Kastner
 
P
,
Hofmann
 
A
.
Gender disparities in red blood cell transfusion in elective surgery: a post hoc multicentre cohort study
.
BMJ Open
 
2016
;
6
:
e012210
.

695

Riesenhuber
 
M
,
Spannbauer
 
A
,
Rauscha
 
F
,
Schmidinger
 
H
,
Boszotta
 
A
,
Pezawas
 
T
, et al.  
Sex differences and long-term outcome in patients with pacemakers
.
Front Cardiovasc Med
 
2020
;
7
:
569060
.

696

Varma
 
N
,
Mittal
 
S
,
Prillinger
 
JB
,
Snell
 
J
,
Dalal
 
N
,
Piccini
 
JP
.
Survival in women versus men following implantation of pacemakers, defibrillators, and cardiac resynchronization therapy devices in a large, nationwide cohort
.
J Am Heart Assoc
 
2017
;
6
:
e005031
.

697

Fang
 
MC
,
Singer
 
DE
,
Chang
 
Y
,
Hylek
 
EM
,
Henault
 
LE
,
Jensvold
 
NG
, et al.  
Gender differences in the risk of ischemic stroke and peripheral embolism in atrial fibrillation: the AnTicoagulation and Risk factors In Atrial fibrillation (ATRIA) study
.
Circulation
 
2005
;
112
:
1687
1691
.

698

Aktaa
 
S
,
Batra
 
G
,
Wallentin
 
L
,
Baigent
 
C
,
Erlinge
 
D
,
James
 
S
, et al.  
European Society of Cardiology methodology for the development of quality indicators for the quantification of cardiovascular care and outcomes
.
Eur Heart J Qual Care Clin Outcomes
 
2022
;
8
:
4
13
.

699

Minchin
 
M
,
Roland
 
M
,
Richardson
 
J
,
Rowark
 
S
,
Guthrie
 
B
.
Quality of care in the United Kingdom after removal of financial incentives
.
N Engl J Med
 
2018
;
379
:
948
957
.

700

Song
 
Z
,
Ji
 
Y
,
Safran
 
DG
,
Chernew
 
ME
.
Health care spending, utilization, and quality 8 years into global payment
.
N Engl J Med
 
2019
;
381
:
252
263
.

701

Arbelo
 
E
,
Aktaa
 
S
,
Bollmann
 
A
,
D’Avila
 
A
,
Drossart
 
I
,
Dwight
 
J
, et al.  
Quality indicators for the care and outcomes of adults with atrial fibrillation
.
Europace
 
2021
;
23
:
494
495
.

702

Schiele
 
F
,
Aktaa
 
S
,
Rossello
 
X
,
Ahrens
 
I
,
Claeys
 
MJ
,
Collet
 
JP
, et al.  
2020 Update of the quality indicators for acute myocardial infarction: a position paper of the Association for Acute Cardiovascular Care: the study group for quality indicators from the ACVC and the NSTE-ACS guideline group
.
Eur Heart J Acute Cardiovasc Care
 
2021
;
10
:
224
233
.

703

Aktaa
 
S
,
Abdin
 
A
,
Arbelo
 
E
,
Burri
 
H
,
Vernooy
 
K
,
Blomstrom-Lundqvist
 
C
, et al.  
European Society of Cardiology Quality Indicators for the care and outcomes of cardiac pacing: developed by the Working Group for Cardiac Pacing Quality Indicators in collaboration with the European Heart Rhythm Association of the European Society of Cardiology
.
Europace
 
2022
;
24
:
165
172
.

704

Corrigendum to: 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: the Task Force for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC)
.
Eur Heart J
 
2021
;
42
:
2298
.

705

Batra
 
G
,
Aktaa
 
S
,
Wallentin
 
L
,
Maggioni
 
AP
,
Wilkinson
 
C
,
Casadei
 
B
, et al.  
Methodology for the development of international clinical data standards for common cardiovascular conditions: European Unified Registries for Heart Care Evaluation and Randomised Trials (EuroHeart)
.
Eur Heart J Qual Care Clin Outcomes
 
2021
:
qcab052
.

Author notes

The two chairpersons contributed equally to the document and are joint corresponding authors.

The two Task Force Coordinators contributed equally to the document

Author/Task Force Member affiliations: listed in author information.

Representing the European Society of Anaesthesiology and Intensive Care (ESAIC)

ESC Clinical Practice Guidelines (CPG) Committee listed in the Appendix.

ESC subspecialty communities having participated in the development of this document:

Associations: Association for Acute CardioVascular Care (ACVC), Association of Cardiovascular Nursing & Allied Professions (ACNAP), European Association of Cardiovascular Imaging (EACVI), European Association of Percutaneous Cardiovascular Interventions (EAPCI), European Heart Rhythm Association (EHRA), and Heart Failure Association (HFA).

Councils: Council of Cardio-Oncology and Council on Valvular Heart Disease.

Working Groups: Adult Congenital Heart Disease, Aorta and Peripheral Vascular Diseases, Cardiovascular Pharmacotherapy, Cardiovascular Surgery, and Thrombosis.

Patient Forum The content of these European Society of Cardiology (ESC) Guidelines has been published for personal and educational use only. No commercial use is authorized. No part of the ESC Guidelines may be translated or reproduced in any form without written permission from the ESC. Permission can be obtained upon submission of a written request to Oxford University Press, the publisher of the European Heart Journal, and the party authorized to handle such permissions on behalf of the ESC (journals.permissions@oup.com).

Disclaimer: The ESC Guidelines represent the views of the ESC and were produced after careful consideration of the scientific and medical knowledge and the evidence available at the time of their publication. The ESC is not responsible in the event of any contradiction, discrepancy, and/or ambiguity between the ESC Guidelines and any other official recommendations or guidelines issued by the relevant public health authorities, particularly in relation to good use of health care or therapeutic strategies. Health professionals are encouraged to take the ESC Guidelines fully into account when exercising their clinical judgment, and in the determination and the implementation of preventive, diagnostic, or therapeutic medical strategies; however, the ESC Guidelines do not override, in any way whatsoever, the individual responsibility of health professionals to make appropriate and accurate decisions in consideration of each patient’s health condition and in consultation with that patient and, where appropriate and/or necessary, the patient’s caregiver. The ESC Guidelines do not exempt health professionals from taking into full and careful consideration the relevant official updated recommendations or guidelines issued by the competent public health authorities, in order to manage each patient’s case in light of the scientifically accepted data pursuant to their respective ethical and professional obligations. It is also the health professional’s responsibility to verify the applicable rules and regulations relating to drugs and medical devices at the time of prescription.

© The European Society of Cardiology 2022. All rights reserved. For permissions please e-mail: journals.permissions@oup.com

All experts involved in the development of these guidelines have submitted declarations of interest. These have been compiled in a report and simultaneously published in a supplementary document to the guidelines. The report is also available on the ESC website www.escardio.org/Guidelines

graphic See the European Heart Journal online for supplementary data that include background information and detailed discussion of the data that have provided the basis of the guidelines.

graphic Click here to access the corresponding ESC CardioMed chapters.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)