Next Article in Journal
Echocardiography and Cardiac Magnetic Resonance in the Assessment of Left-Ventricle Remodeling: Differences Implying Clinical Decision
Previous Article in Journal
Evaluating GlicoPro Tear Substitute Derived from Helix aspersa Snail Mucus in Alleviating Severe Dry Eye Disease: A First-in-Human Study on Corneal Esthesiometry Recovery and Ocular Pain Relief
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Implementation of the Care Bundle for the Management of Chronic Obstructive Pulmonary Disease with/without Heart Failure

by
Andrea Bianco
1,2,
Marco Canepa
3,4,
Giosuè Angelo Catapano
5,
Maurizio Marvisi
6,
Fabrizio Oliva
7,
Andrea Passantino
8,
Riccardo Sarzani
9,10,*,
Paolo Tarsia
11,12 and
Antonio Giovanni Versace
13
1
Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy
2
U.O.C. Pneumology Clinic “L. Vanvitelli”, A.O. dei Colli, Ospedale Monaldi, 80131 Naples, Italy
3
Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
4
Department of Internal Medicine, University of Genova, 16132 Genoa, Italy
5
G. Monasterio Tuscany Foundation, 56124 Pisa, Italy
6
Department of Internal Medicine, Cardiology and Pneumology, Istituto Figlie di S. Camillo, 26100 Cremona, Italy
7
Cardiology 1, A. De Gasperis Cardicocenter, ASST Niguarda Hospital, 20162 Milan, Italy
8
Division of Cardiology and Cardiac Rehabilitation, Scientific Clinical Institutes Maugeri, IRCCS Institute of Bari, 70124 Bari, Italy
9
Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60126 Ancona, Italy
10
Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60020 Ancona, Italy
11
Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
12
Internal Medicine Department, Metropolitan Hospital Niguarda, 20162 Milan, Italy
13
Department of Clinical and Experimental Medicine, Policlinic “Gaetano Martino”, University of Messina, 98100 Messina, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(6), 1621; https://doi.org/10.3390/jcm13061621
Submission received: 1 February 2024 / Revised: 29 February 2024 / Accepted: 2 March 2024 / Published: 12 March 2024
(This article belongs to the Section Pulmonology)

Abstract

:
Chronic obstructive pulmonary disease (COPD) is often part of a more complex cardiopulmonary disease, especially in older patients. The differential diagnosis of the acute exacerbation of COPD and/or heart failure (HF) in emergency settings is challenging due to their frequent coexistence and symptom overlap. Both conditions have a detrimental impact on each other’s prognosis, leading to increased mortality rates. The timely diagnosis and treatment of COPD and coexisting factors like left ventricular overload or HF in inpatient and outpatient care can improve prognosis, quality of life, and long-term outcomes, helping to avoid exacerbations and hospitalization, which increase future exacerbation risk. This work aims to address existing gaps, providing management recommendations for COPD with/without HF, particularly when both conditions coexist. During virtual meetings, a panel of experts (the authors) discussed and reached a consensus on the differential and paired diagnosis of COPD and HF, providing suggestions for risk stratification, accurate diagnosis, and appropriate therapy for inpatients and outpatients. They emphasize that when COPD and HF are concomitant, both conditions should receive adequate treatment and that recommended HF treatments are not contraindicated in COPD and have favorable effects. Accurate diagnosis and therapy is crucial for effective treatment, reducing hospital readmissions and associated costs. The management considerations discussed in this study can potentially be extended to address other cardiopulmonary challenges frequently encountered by COPD patients.

1. Introduction

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide and has the highest rate of hospital admissions among major diseases [1,2]. The high rates of hospitalization due to the acute exacerbation of COPD (AECOPD) represent a substantial economic and social burden, accounting for 50% of the costs of this disease and they are associated with a reduced quality of life and a poor prognosis [2,3,4,5,6,7,8,9,10,11].
AECOPD can be triggered by viral or bacterial respiratory infections [12,13,14,15,16], but environmental factors such as pollution and ambient temperature can increase symptoms [12,17,18,19,20,21,22,23,24,25,26,27,28,29].
Although smoking is a major risk factor for COPD, up to 30% of cases occur in non-smokers [30,31], predominantly women with symptoms of chronic bronchitis [32]. Non-smokers with COPD may be exposed to passive smoke, environmental pollutants, or occupational hazards or have a history of asthma [11]. A recent study showed a link between dysanapsis (a mismatch of the airway tree caliber to lung size) and COPD in older adults, where a lower airway tree caliber relative to lung size increases COPD risk [33]. Non-smokers with COPD exhibit different clinical features [34,35], with predominant airway involvement [34,35] but milder symptoms [31,36,37] and airflow limitation with normal DLCO values [31,36,38,39,40] compared to smokers with COPD. They tend to have a younger age at onset [31] and a poor prognosis with an increased risk of exacerbation [41]. Biomass-exposure-related COPD shows distinct radiological and pathological differences [42], including significant bronchitis and fibrosis [32,43,44]. While pharmacological management is similar for smokers and non-smokers with COPD, individual responses to treatment may vary. Despite limited data, non-smoker COPD is considered a distinct clinical phenotype resembling the chronic bronchitis phenotype.
Several studies have reported that more than 60% of patients with COPD are readmitted to the hospital within 1 year of a previous exacerbation [45,46,47]; recurrent exacerbations and hospitalizations are associated with a poor prognosis because they facilitate a decline in lung function [48] and increase the risk of future admissions [45,46,49,50,51].
The Global Initiative for Chronic Obstructive Lung Disease (GOLD) document classifies COPD based on measured values of FEV1 (forced expiratory volume in 1 s) [11]. The 2024 version of the document emphasizes the role of exacerbations in categorizing COPD patients, using the recently introduced ABE assessment tool, which represents a departure from the earlier ABCD approach [11].
It is important to manage the patient’s comorbidities and identify predictors of hospital readmission [46,52,53] because some of them are not related to COPD. On the contrary, COPD is often part of a more complex cardiopulmonary disease, especially in older patients.
Cardiovascular comorbidities are commonly associated with COPD, making the differential diagnosis challenging [11]. All patients with COPD should have ischemic heart disease taken into consideration and treated, regardless of the presence of concomitant COPD [11]. COPD patients with concomitant ischemic heart disease face a high risk of cardiovascular events within 90 days following an exacerbation [11,54].
Additionally, COPD patients frequently experience cardiac arrhythmias [11,55], and atrial fibrillation is often observed during AECOPD [11,56]. Furthermore, the prevalence of peripheral artery disease is higher in COPD patients compared to the general population and is commonly associated with atherosclerotic heart disease [11,57].
COPD is a common comorbidity in patients with heart failure (HF); the prevalence of COPD in patients with HF ranges from 20% to 30% [58,59,60,61,62] and a similar prevalence of HF has been reported in large populations of patients with COPD [60,61]. The reasons underlying the high prevalence of COPD among patients with HF and vice versa have long been debated; many studies have shown a contribution of tobacco smoking, because it is a risk factor shared by the two conditions [63,64,65], as well as those of an unhealthy lifestyle, arterial hypertension, aging, dyslipidemia, and the chronic inflammatory burden associated with COPD [60,66,67,68,69,70]. Among the several processes associated with COPD, lung hyperinflation, hypoxemia and systemic inflammation can increase the risk of HF [71]. An overview of the common risk factors for COPD and HF is provided in Figure 1.
COPD worsens the clinical presentation and course of HF and has been associated with a 20–30% increase in the risk of hospitalization of patients with HF and a 1.61× increased risk of mortality [60,72,73,74,75,76,77]. Furthermore, COPD increases the prevalence of comorbidities such as atrial fibrillation, renal dysfunction, diabetes mellitus, higher heart rate and systolic blood pressure, and infection, which can trigger HF decompensation [60,72,75,78]. The 5-year survival rate of patients with HF and concomitant COPD is 31% versus 71% in the absence of COPD [79].
The prevalence of COPD is higher in hospitalized patients with HF [76,80,81] compared with those with chronic HF [60,82,83], probably due to the overdiagnosis of COPD in acute HF settings when symptoms are easily misinterpreted [84]. The differential diagnosis of AECOPD and HF in clinical settings is extremely challenging because they often share a similar clinical presentation with symptoms such as impaired exercise tolerance with exertional breathlessness, fatigue, sleep disturbances such as nocturnal cough and paroxysmal nocturnal dyspnea with no distinctive feature that could help differentiate between COPD and HF [85], muscle weakness, cachexia, and anxiety [72]. AECOPD symptoms such as tachycardia and hypoxemia can precipitate HF decompensation, and pulmonary fluid retention due to HF can worsen airflow restriction [77].
A systematic review found that HF increases hospitalization and mortality in patients with COPD [86], which was corroborated recently in a study of the cause of death among hospitalized patients with AECOPD [74]. The increased risk of death with cardiovascular dysfunction also affects patients with milder or moderate COPD. This was clearly demonstrated in a study by Mannino et al. [87] showing that 27.6% of GOLD 2 patients, 24.8% of GOLD 1 patients, and 39.3% of patients with airway restriction (FEV1/forced vital capacity [FVC] ≥ 0.70 and FVC < 80% predicted) died due to cardiovascular dysfunction, which predominated as a cause of death over lung cancer and respiratory failure.
The GOLD document fails to provide clear indications on the care of patients with COPD/HF, advising to use selective β1-blockers for cardiovascular indications and referring to the HF guidelines [11,88,89,90], which may not be specific enough for these challenging patients [91]. Consequently, patients with known multiple comorbidities such as COPD and HF need a comprehensive health care approach with integrated care provided by pulmonologists and cardiologists [77] rather than the separate treatment of the individual diseases.
A decrease in hospital readmissions needs to be achieved as an outcome in order to improve patients quality of life and to reduce the economic burden related to the disease. Multiple studies dealing with this topic have been published in the literature recently [92,93,94,95,96,97]; in particular, a study by Kalhan et al. [98] proposed a flowchart to simplify and improve the efficacy of the journey for the patient with COPD and HF. A recent review published by Celli et al. [99] provided clear and comprehensive algorithms for the differential diagnosis of a number of clinical conditions that may resemble AECOPD in the acute care setting, including HF.
The aim of the present work is to propose a care bundle to guide clinicians, based on clinical guidelines, on the best practices in the identification, diagnosis, and management of patients with COPD, with or without concomitant HF, with a focus on the optimal practice to distinguish COPD and HF in emergency care settings. As defined by the Institute for Healthcare Improvement, a care bundle is “a small set of evidence based interventions for a defined patient segment/population and care setting that, when implemented together, will result in significantly better outcomes than when implemented individually” [100]. Care bundles have been demonstrated to improve the outcomes of COPD, especially when a respiratory specialist is involved [101,102].
A panel of nine COPD and HF Italian experts, including cardiologists, pneumologists, and internists, participated in a virtual advisory board in October 2022 to provide recommendations on the differential diagnosis of a patient with COPD and risk of HF. The experts provided recommendations for acute care treatment, risk stratification, correct allocation, definitive diagnosis, post-discharge care, and the following up of patients admitted to emergency care settings with dyspnea, with or without a previous diagnosis of COPD and exacerbations. In particular, they highlighted the role of a multidisciplinary approach in the evaluation and management of these patients. They also gave indications on the treatment and following up of patients in outpatient care after an AECOPD event that did not require hospitalization. The recommendations provided by these clinicians to obtain a differential diagnosis of COPD and HF are summarized in a flowchart (Figure 2) and in Supplemental Table S1.
While these experts and their experiences largely reflect the nuances of the Italian health system, the insights could be applicable in part or whole to many other systems managing these patients.

2. Differential Diagnosis and Paired Diagnosis

Differential diagnosis has a pivotal role in discriminating between AECOPD and HF in a patient admitted to the hospital due to dyspnea. It guides appropriate acute and chronic therapy and the appropriate location for the patient in the hospital setting. A proper diagnosis reduces the need for unnecessary tests and improves the prognosis, reducing hospital admissions and decreasing costs.
A correct definition of the disease is the fundamental cornerstone of differential diagnosis. The current definition of AECOPD provided by the European Respiratory Society/American Thoracic Society reads as follows: “In a patient with underlying COPD, exacerbations are episodes of increasing respiratory symptoms, particularly dyspnea, cough and sputum production, and increased sputum purulence” [103]. It is similar to the original definition published by René Laennec over 200 years ago [104]. However, both these as well as recent GOLD definitions [11] do not relate the symptoms to measurable variables, and the severity of the event is often related to the medicine used for treatment rather than to the physiologic alterations [105,106]. Accordingly, the underlying triggers of AECOPD are rarely investigated, and the therapeutic options available have not been modified in more than 30 years.

2.1. The Importance of a Medical Interview in the Differential Diagnosis

An extensive medical interview that encompasses the known risk factors for COPD plays a fundamental role in the differential diagnosis. A recently published Lancet Commission report on COPD by Stolz et al. [107] highlighted the importance of risk factors, suggesting a classification of COPD into five types based on the risk factors responsible for the etiopathogenesis:
  • genetics (i.e., serpin family A member 1 [SERPINA1] gene mutation leading to α-1 antitrypsin deficiency, telomerase reverse transcriptase mutations, or other epigenetic causes still remaining to be defined);
  • early life events (i.e., prematurity, low weight at birth, childhood asthma);
  • infections (i.e., childhood infections such as with pneumonia and respiratory syncytial virus or adulthood infections such as with tuberculosis and human immunodeficiency virus [HIV]);
  • inhalation of tobacco, drugs, and other combustible substances;
  • environmental exposures (i.e., indoor pollutants, ambient air pollution, occupational exposures).
The classification suggested in the Lancet Commission has been implemented into the GOLD 2024 report, which proposes a classification into different “etiotypes” reflecting the different pathogenic mechanisms underlying COPD [11].
Accordingly, the clinicians recommend that the medical interview includes information on the risk factors cited above, when available, especially on smoking habits, because tobacco smoke is a leading risk factor for COPD [108]. Additionally, the clinicians advise gathering information on previous spirometry results if available, exacerbation history, and comorbidities (particularly those increasing the overall cardiopulmonary risk, such as hypertension, dyslipidemia, and type 2 diabetes), as well as ongoing pharmacologic and non-pharmacologic therapies.

2.2. Differential Diagnosis in the Acute Care Setting: AECOPD and/or HF?

The differential diagnosis can be extremely challenging in acute care settings, where patients with a previous diagnosis of COPD and patients with symptoms common to COPD and HF who do not have a previous diagnosis are admitted. Accurate clinical history recording and clinical evaluation with proper diagnostic and laboratory tests are the best means of discriminating between COPD and/or cardiovascular dysfunction (Figure 3).
During the evaluation of symptoms and the clinical examination, the authors recommend determining arterial blood pressure, heart and respiratory rate, dyspnea, and sputum production. The respiratory rate is an extremely informative albeit often-neglected measurement, and specific thresholds should be set to increase its clinical significance. A cardiovascular risk score needs to be carefully evaluated if the patient has been hospitalized in an internal medicine setting or in a respiratory ward because COPD must be managed while considering possible underlying cardiovascular dysfunction (i.e., arterial hypertension, ischemic heart disease, and HF). The presence of bilateral crackles at the basis in the upright position (initial pulmonary transudate and pulmonary edema), pulmonary congestion (even before edema) with B-lines, or white lung assessed through lung sonography, leg edema, increased jugular venous pressure, and a third heart sound should guide the diagnosis toward HF as a cause or a contributor to the AECOPD event [109] (Figure 3). The clinical evaluation should collect information about chest pain, signs of atherosclerotic disease, and ischemic cardiomyopathy given the high prevalence of ischemic heart disease in AECOPD events [110,111] (Figure 3). Localized crackles, crepitus at thoracic auscultation, and percussion dullness may indicate pneumonia [99,112], a suspicion that should be accurately ruled out because COPD increases the risk of pneumonia and its severity [113].
The authors recommend several diagnostic tests in an emergency care setting, which are summarized in Table 1.
Among them, blood gas analysis, pulse oximetry, electrocardiography, lung sonography, and chest X-ray should be performed as soon as possible, ideally within the first 24 h after hospital admission. Electrocardiographic monitoring has a strong negative predictive value in ruling out cardiac dysfunction and arrhythmias [114,115] but cannot exclude COPD [116], and pulse oximetry and blood gas analysis, as well as guiding the diagnosis, indicate the severity of the patient’s condition. Chest radiography (upright position) is recommended to exclude diseases such as pulmonary fibrosis, pneumothorax, pneumonia, or lung cancer and to identify signs of HF such as cardiomegaly, pulmonary edema [117], and pleural effusion (Figure 3). The panel strongly emphasizes the need to perform chest radiography in the seated position; most chest radiographs are performed with the patient in a recumbent position due to lack of time and experience, but if the patient has edema caused by congestive HF, this would prevent the proper evaluation of the lungs. Blood gas analysis and chest radiography are also recommended in patients with known COPD who are admitted to the hospital with the need to discriminate between AECOPD or/and underlying HF. Lung sonography should also include the assessment of diaphragmatic excursion; this provides information on respiratory muscle function, especially when blood gas analysis shows an increase in PaCO2 (partial pressure of carbon dioxide in arterial blood), and allows the prompt identification of patients with congestive HF (Figure 3). Small portable ultrasonography machines should be available to doctors, and they should be trained in evaluating diaphragmatic excursion. In cases of suspected HF, echocardiography is useful for the evaluation of cardiac structure and function; it should study both the left and the right chambers of the heart, and the left ventricle EF should be evaluated. It can identify pulmonary hypertension by measuring pulmonary artery systolic pressure or pleural effusion. A computed tomography (CT) scan of the chest is recommended for patients who are already diagnosed with COPD and experience multiple exacerbations; in addition, CT identifies opacities that cannot be seen on a chest radiograph [17,118], and it may help discriminate bronchiectasis from AECOPD [11].
The laboratory tests recommended at hospital admission are summarized in Table 1. Venous blood samples should be collected for the following analyses: blood count, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), platelet count, fibrinogen, D-dimer, troponin, glycemia, creatine kinase muscle–brain isoenzyme (CK-MB), and white blood cell differential, in particular eosinophilic and neutrophilic pattern. Increased CRP levels may indicate bacterial pneumonia, especially if associated with a high white blood cell count [119]. An increased troponin level can be observed in patients with COPD and can increase further during AECOPD [120], but myocardial ischemia should be strongly suspected when associated with an abnormal ECG and symptoms related to cardiac dysfunction [121].
COPD and its comorbidities may account for an additional chronic hemodynamic overloading of the heart. Both B-type natriuretic peptide (BNP) and N-terminal prohormone of brain natriuretic peptide (NT-proBNP) are useful markers to exclude HF in patients with AECOPD [122,123,124,125] (Figure 3), but BNP is less reliable, and the related studies have major limitations such as the underuse of echocardiography to diagnose HF and misdiagnosis of HF due to advanced lung diseases that cause increased pulmonary arterial pressure (cor pulmonale) [126,127]. Natriuretic peptides are commonly increased in patients, with COPD increasing further during AECOPD [128,129,130], so higher thresholds would probably be needed to discriminate between COPD and COPD with concomitant HF, even if abnormalities in other tests may help discriminate. For example, a large pleural effusion—detected through chest radiography, lung sonography, or CT—associated with increased natriuretic peptides guides the diagnosis toward acute HF [99]. Increased natriuretic peptides correlate with an increased risk of death in patients with COPD during AECOPD [131]. European HF guidelines recommend determining blood NT-proBNP levels in patients presenting with signs compatible with HF, including dyspnea, to identify an underlying cardiac involvement that can be further investigated using echocardiography when available [130]. Accordingly, NT-proBNP is a good diagnostic/prognostic marker of heart overload or HF with a high negative predictive value, but it also shows a good positive predictive value in older patients; it is increased in a high percentage of cases of COPD in the elderly, as demonstrated by a retrospective study by Sarzani et al. [132] that included 403 elderly patients (mean age, 88.1 ± 5.1 years) who had been hospitalized for different conditions other than HF. Of the patients with at least one symptom compatible with HF, 61% had NT-proBNP values higher than 1800 pg/mL [132], the most validated and widely used age-adjusted cutoff for a diagnosis of HF [124]. Furthermore, 32.8% of patients had values of NT-proBNP ranging between 300 pg/mL and 1799 pg/mL, which could indicate underlying HF or other types of cardiac dysfunction [132,133]. Of the patients who died during hospitalization, 56.4% had an NT-proBNP value ≥ 1800 pg/mL on admission, and accordingly, high values of NT-proBNP were positively associated with in-hospital death through a logistic regression model [132]. Considering that only 19.7% of patients had a known history of HF, these findings underline the importance of NT-proBNP as a diagnostic criterion for HF in the elderly and also as a prognostic factor for mortality; these results are in line with those obtained in previous studies [134,135,136]. Although European HF guidelines recommend echocardiography to evaluate cardiac structure and function, it is difficult to obtain promptly in emergency settings. The interpretation of its results can be complicated in the elderly due to the high prevalence of atrial fibrillation, which impedes the proper measure of several echocardiographic diastolic components and the uncertain diagnostic values in patients aged >80 years [137,138,139]. NT-proBNP is inexpensive and easy to measure in peripheral blood and should be included in the tests ideally conducted in the first 24 h after hospital admission. Its value as a diagnostic marker of cardiac illness and a prognostic factor for mortality is not influenced by multiple comorbidities in the elderly [140]. Furthermore, its increased levels correlate with a number of echocardiographic abnormalities [139] despite the potential for independently increased levels due to other conditions (e.g., atrial fibrillation, pulmonary hypertension, and right ventricular strain and failure [141,142]).
A consensus document released by the Heart Failure Association of the ESC has explored the practical applications of NT-proBNP across different clinical situations. This research has established validated NT-proBNP cut-off points for non-obese individuals without kidney failure and atrial fibrillation/flutter during the baseline electrocardiogram, stratified based on age and gender, enabling the identification of acute HF in emergency department cases and the diagnosis of de novo HF in outpatient settings [143].
The study also highlights that irrespective of age or gender, patients with NT-proBNP values higher than 2000 pg/mL should undergo echocardiography and clinical evaluation within 2 weeks of diagnosis while HF is very unlikely if NT-pro-BNP values are lower than 125 pg/mL [143].
The assessment of NT-proBNP levels guides the treatment of HF, leading to an increased number of patients treated with β-blockers and renin–angiotensin system blockers, resulting in reduced levels of NT-proBNP at discharge and heart protection [132].

3. Acute Care Therapy

If the patient is critically ill, with unstable vital signs, fast action is necessary. Blood gas analysis and electrocardiography are performed and hemodynamic parameters monitored. If acute coronary syndrome is suspected, the patient has to be referred to the cardiac intensive care unit further investigations including a high-resolution CT angiography scan. Blood thinners or thrombolytic drugs are administered in cases of pulmonary embolism.
Treatment is guided by the results of the previous tests; supplemental oxygen is administered if necessary, according to blood gas analysis, and non-invasive ventilation is provided in cases of acute hypercapnic respiratory failure, taking into consideration that oxygen therapy and non-invasive ventilation are most efficacious when started within the first 30 and 60 min, respectively. Non-invasive ventilation is highly recommended for the treatment of AECOPD with acute hypercapnic respiratory failure, as suggested also by the British Thoracic Society guidelines [144], and is frequently used by pulmonologists. During the SARS-CoV-2 pandemic, physicians from different specializations also gained experience in the use of non-invasive mechanical ventilation. Corticosteroids and antibiotics in cases of pneumonia should be administered when indicated. The patient’s health status should be monitored for 6 h and then regularly reevaluated. If HF is present, patients should receive appropriate therapy including a β1-selective blocker, diuretics, angiotensin-converting enzyme (ACE) inhibitors, or, better, angiotensin receptor blockers (ARBs), angiotensin receptor neprilysin inhibitors (ARNIs), and/or sodium–glucose co-transporter 2 inhibitors (SGLT2-is). ACE inhibitors can cause persistent cough and are contraindicated if ARNI use is necessary. If required, dual bronchodilators or triple therapies with a pressurized metered dose inhaler and spacer should be started during the hospital stay for better results as, generally, delayed therapy is related to additional exacerbations and costs [145].

4. Risk Stratification and Correct Allocation

Consultation with pulmonologists and cardiologists is fundamental for the correct risk stratification of COPD and/or HF and to avoid misdiagnosis. The patient needs to be referred to the appropriate hospital setting during the hospital stay for proper testing and to reduce the future risk of readmission. After stabilizing the patient with AECOPD, it is important to perform risk stratification for further exacerbations, considering the comorbidities through a synergic interplay between pulmonologists, cardiologists, and internists.
In this context, second-level analyses need to be carried out to provide a definitive diagnosis. Even when the patient is hospitalized in an internal medicine setting, the cardiovascular risk should be assessed. It is necessary to explore both the right and left chambers through echocardiography and to quantify the left ventricular EF to determine whether there is heart dysfunction. Biomarkers such as BNP, NT-proBNP, CRP, and ESR need to be monitored. If hypoxemia is present and its cause is not identified, a pulmonary CT angiography scan and D-dimer analysis may be necessary to exclude pulmonary embolism; when severe hypoxia is present and the patient needs therapy with high-flow oxygen, possibly with pulmonary hypertension as a complication, and COPD and HF are excluded, the scenario is suggestive of pulmonary embolism, which can be difficult to diagnose, especially in older patients with multiple comorbidities.

5. Hospital Stay: Definitive Diagnosis

Spirometry evidence of bronchial obstruction is a stringent requirement and a fundamental criterion in pulmonary guidelines to confirm a diagnosis of COPD [11,90]. The authors advise the use of spirometry to discriminate between AECOPD or HF in a patient with previously diagnosed COPD; however, real-world evidence shows that this is not used routinely for COPD patients [146]. During the early 2000s, a significant number of newly diagnosed patients with COPD who met high-risk criteria did not have their diagnoses confirmed through spirometry [146]. However, the situation underwent a rapid change after 2004, with the percentage of patients having a record of spirometry reaching its peak at 72% in 2014 before declining again [146]. By 2019, only 59% of high-risk newly diagnosed patients had a recorded spirometry result within the 12-month period prior to their COPD diagnosis [146].
Spirometry measures lung volumes mobilized with inspiratory or expiratory maneuvers with the aim of determining whether lung obstruction is present. Ventilator defects are common in patients with both HF and COPD, but obstructive spirometry in patients with HF usually improves after the use of diuretics [147,148]. However, spirometry is rarely considered in patients with HF and known or suspected COPD [60]. Canepa and colleagues calculated that only 19.6% of patients hospitalized with HF and 30.6% in a population of patients with chronic HF underwent spirometry [75,91]. In a cohort study, it was found that 65% of COPD patients with concomitant HF receiving appropriate HF therapy and 39% of COPD patients receiving inadequate HF therapy underwent spirometry testing [149]. Additionally only 35% of patients with both HF and newly diagnosed COPD, who were prescribed inadequate therapy for COPD, had recorded spirometry results [149]. Conversely, among HF patients who were prescribed appropriate therapy for newly diagnosed COPD, the percentage of recorded spirometry results increased to 57% [149]. A possible explanation for the underuse of spirometry in patients with HF who have symptoms of COPD is that spirometry is not easily accessible to cardiologists and the fact that the cardiology community shows some skepticism towards the discriminatory capacity of spirometry in patients with HF [72]; accordingly, the prevalence of COPD is notably lower in patients managed by cardiologists as opposed to general physicians [150,151,152,153]. The interpretation of spirometry results during acute decompensation in patients with HF can be challenging because “wet lung HF” may present with airway restriction or obstruction [62], so the current guidelines of both the European Society of Cardiology [154] and the GOLD group [11,90] recommend that spirometry is performed when HF conditions are stable [62,75,148,155]. However, timely referral and following up to the pulmonologist or respiratory clinic, or alternatively the use of spirometry in cardiac clinics, needs to be implemented because its scarce use leads to about 30% of patients with chronic HF being mislabeled with COPD without confirmation of airflow obstruction, and a similar proportion of patients have unrecognized airflow obstruction [156]. A restrictive spirometric pattern warrants further investigation to determine whether it indicates HF decompensation, airway wall edema due to congestion, other pulmonary conditions, or neuromuscular disease [157].
The GOLD and American Thoracic Society/European Respiratory Society guidelines defined a fixed cutoff for the ratio of post-bronchodilator FEV1 to an FVC [158] of <0.70 [11,159] for the diagnosis of persistent airflow reduction and the use of the percentage of predicted FEV1 to classify the degree of obstruction [90]. The fixed cutoff value of FEV1/FVC < 0.70 may lead to the overdiagnosis of COPD in elderly patients because this ratio decreases with aging [160]. An alternative approach was proposed whereby airflow limitation is diagnosed when the FEV1/FVC is lower than or equal to the lower limit of normality (LLN), which is the fifth percentile of a healthy, nonsmoking population [160]. This latter option may be more accurate, but sometimes, patients may have a normal spirometry considering the LLN and an abnormal spirometry using the fixed cutoff; an increased risk of hospitalization in this subgroup of patients has been reported [160] and since the fixed ratio is simpler and more consistent to use than LLN, the international guidelines for COPD promote the use of the fixed ratio over the LLN [11,90].
Dyspnea is a common presentation of decompensated HF, COPD, or asthma. Differentiating AECOPD from asthma is another clinical challenge: asthma and COPD can be comorbid [161], symptoms often overlap, and even though reversibility with bronchodilators [162] and resolution with inhaled corticosteroids [90] are suggestive of asthma, there are exceptions. In addition to spirometry, other tests such as of the diffusing capacity of the lungs for carbon monoxide (DLCO) and bronchial provocation testing may help diagnose asthma. However, differences between COPD and asthma are beyond the scope of this paper; for more details, we refer the reader to extensive reviews of the literature on this topic [163,164,165].
Diagnostic tests such as spirometry, DLCO, and cardiac biomarkers such as NT-proBNP, echocardiography, and chest imaging are fundamental to diagnose HF in patients with COPD and to discriminate between AECOPD and HF in patients admitted to the hospital with dyspnea. The same synergistic approach should be used to differentiate COPD from other conditions that have overlapping symptoms, such as asthma, bronchiectasis, pneumonia, and pleural effusion, as highlighted by the recent review published by Celli and colleagues [99]. Past history and clinical evaluation should be combined with the results of imaging and laboratory tests and with spirometry. A synergistic approach that involves cardiologists, pulmonologists, and internists is fundamental to interpret the results. The British Thoracic Society recommends a specialist examination within the first 24 h of hospital admission. Although this can be difficult to obtain in most care settings, at least in Italy, a specialist visit before hospital discharge is highly advised for all patients admitted for AECOPD to provide a definitive diagnosis.

6. Hospital Discharge and Follow-Up

The correct planning of follow-ups and post-discharge care can have an enormous impact on reducing hospitalizations; several previous studies have demonstrated the beneficial effect of timely follow-up visits and disease-specific management programs [166,167,168,169]. These may consist of medical education sessions on COPD (to increase knowledge of the disease and treatment adherence); demonstrations of inhaler techniques; discharge planning of an appropriate maintenance treatment; counseling to quit tobacco smoking; and recommendations to refer to pulmonary rehabilitation programs, to adhere to influenza and pneumococcal vaccination programs, and to exercise regularly, as well as coaching by a case manager [6,170,171]. These recommendations have all been shown to be useful in hastening recovery, reducing hospitalizations and readmissions [166,167,168,169,170,172,173,174,175,176,177,178], and reducing the economic burden for each patient [179]. Recommendations for patients and caregivers are summarized in the Supplemental Table S2. The transition from hospital to home or to long-term care facilities has a fundamental role for a successful management of COPD after discharge, and it should be handled considering care coordination and a structured discharge planning.
In a recent study, COPD patients participated in a video telehealth-based pulmonary rehabilitation program comprising 36 exercise and educational sessions over 12 weeks, customized to their individual baseline characteristics [180]. The findings revealed a decrease in the 30-day all-cause readmission rate, a reduction in the 30-day readmission rate specifically due to AECOPD, and an extended time to the first readmission in the telehealth groups compared to the control group [180]. Support at home with telemonitoring may reduce mortality and readmissions after 12 months, as suggested by a study conducted by Marcos et al. [181]. A recent systematic review reported a decrease in readmission rates and/or an improved emotional state in COPD patients provided with personalized case management action plans with constant phone calls, visits, and technical training and education after discharge [182]. Additional research is necessary to underscore the advantages and the great potential linked to telehealth-based interventions [183], as the diversity in clinical trials currently hinders a comprehensive understanding of their potential to enhance COPD healthcare [184,185,186].
The main care gaps in the management of patients with COPD that are responsible for the increase in readmission rates are significant variability in inpatient care and suboptimal care transitions such as a lack of access to timely follow-ups and early disease management programs [9,10,90,172,173,187,188,189,190,191,192,193,194,195,196,197]. Care gaps post discharge are particularly significant for patients with COPD; in some studies, more than 50% of incidences of AECOPD were not reported to health care providers, with a significant impact on health status [51,174,198]. A coordinator should be in charge of managing a structured program for the discharge of COPD patients, alleviating their burden and responsibilities [182]. The CONQUEST multi-national program developed quality standards for the primary and secondary care of COPD patients [199]. These standards aim to promote early intervention in identifying, assessing, and effectively managing both diagnosed and undiagnosed COPD patients who are at a modifiable and high risk of exacerbations [199]. Furthermore, the program emphasizes the importance of follow-up care for such patients [199].
A study conducted two years later compared the management of high-risk COPD patients in the UK with national and international management recommendations and quality standards including those of the CONQUEST program [146]. The study revealed that there is a significant oversight in identifying COPD patients at high risk of exacerbations early on [146]. Both newly diagnosed and already-diagnosed patients at high risk are not promptly assessed or treated [146]. This highlights a considerable opportunity for enhancing the assessment and optimization of treatment for these patients.
COPD tends to be undervalued by primary care physicians compared with other diseases such as cardiovascular disorders, probably because of the absence of specific biomarkers. It is necessary to inform primary care physicians about the need for the punctual following up of patients with COPD; they must monitor the risk of exacerbation after hospital discharge, adherence and response to treatment, and the symptom burden, especially during the first month after discharge when the risk of cardiovascular or ischemic attacks is high, and they should ensure appropriate home assistance if needed. The authors recommend a follow-up visit scheduled up to 12 weeks after discharge if the patient was admitted in an internal medicine setting or 1 month after discharge if the reason for hospital admission was acute HF. A post-discharge follow-up visit within the first 30 days has been shown to significantly reduce mortality in patients with COPD [200]. The authors underline that scheduling visits 6 months or more after hospital discharge for the last event of AECOPD is not useful because the risk of additional exacerbations is higher in the first months.
The visit should take place in pulmonology, cardiology, or internal medicine units according to the patient’s diagnosis. During the visit, it may be useful to perform blood gas analysis, spirometry, and the 6 min walking test and to consult the pulmonologist in case of latent respiratory failure. Additional parameters such as vital signs, pulse oximetry, renal function, and blood inflammatory markers could also be assessed.

7. Post-Discharge COPD and COPD/HF Therapy

The recently updated GOLD document for the treatment of COPD [11] recommends the following:
  • Patients with COPD who present with occasional symptoms or are already under treatment with long-acting bronchodilators but need immediate relief of symptoms are treated with short-acting bronchodilators: either a short-acting muscarinic antagonist or a short-acting β2-agonist or their combination [11].
  • Patients with COPD with dyspnea and two or more moderate AECOPDs or with one or more severe AECOPDs requiring hospitalization are treated with a long-acting β2-agonist (LABA) or a long-acting muscarinic antagonist (LAMA) or their combination; triple therapy with LABA + LAMA + inhaled corticosteroids (ICSs) is recommended if the eosinophil count is above the threshold of ≥300 eosinophils/μL [11]. However, eosinophil levels should be combined with clinical assessment as certain patients with eosinophil counts below 300 eosinophils/μL could also benefit from triple therapy [11].
  • Patients with COPD with no or one moderate AECOPD and without the need for hospitalization should be treated with a single bronchodilator (mMRC score 0–1, CAT score < 10) or with LABA + LAMA (mMRC score ≥ 2, CAT score > 10). When patients experience recurrent exacerbations despite treatment with a dual bronchodilator (LABA + LAMA), the GOLD guidelines recommends switching to triple inhaled therapy with LABA + LAMA + ICSs [11].
If there is an indication for the use of ICSs, the new 2024 GOLD document recommends triple inhaled therapy with LABA + LAMA + ICSs over LABA + ICS therapy [11] according to the results from recent ETHOS and IMPACT trials, which suggest a reduction in mortality among patients with COPD with the use of triple inhaled therapy compared with dual therapy (LABA + ICS) [201,202].
Although definite indications exist for COPD treatment and management, no recent guidelines exist for the treatment of patients with concomitant COPD and HF, and therefore, many patients do not receive appropriate treatment. The cohort study conducted by Kostikas et al. [149] revealed that among a total of 12,587 patients who had both COPD and newly diagnosed HF, adequate HF therapy was prescribed for 18% of the patients. Inadequate therapy for HF was prescribed for 42% of the patients, and a significant portion (40%) remained untreated for HF [149]. Consistent with these results, Canepa et al. highlighted, in their review on the diagnostic and therapeutic gaps in patients with COPD and HF, that half of these patients are not treated with any β-blocker when they are admitted to the hospital and only one third of them are discharged with a β-blocker prescription [91]. The underuse of β-blockers has been associated with increased mortality [203], and there is no rationale for withholding them from patients with COPD/HF [204] because COPD is not a contraindication to β-blockers [154,205] and asthma is only a relative contraindication; in fact, β-blockers seem to improve survival in patients with COPD who do not have HF [203]. Based upon a Cochrane review, the use of β1-selective blockers (i.e., nebivolol and bisoprolol) is preferred and recommended in patients with chronic HF and COPD because they are associated with a lower risk for bronchoconstriction [206]. The reason underlying the low prescription rates of β-blockers for patients with COPD/HF seems to be due more to clinician inertia rather than to a lack of patient compliance [91,207]. SGLT2-is may represent a valid option for the treatment of patients with HF in all the EF ranges (HFrEF, HFmrEF, HFpEF) and concomitant COPD as an additional treatment or, occasionally, as an alternative therapy for patients who are intolerant to β-blockers, as demonstrated in a study by Dewan and colleagues, which showed that dapagliflozin, an SGLT2-i, had a favorable effect in a population of patients with HFrEF and COPD [208,209]. In all cases, optimal blood pressure control must be pursued using combinations of renin–angiotensin–aldosterone inhibitors (RAASis) and appropriate diuretics, especially in people with a tendency for salt and water retention. RAASis are also recommended in patients with HFrEF. ARNIs can also be indicated and ARBs should be preferred before their use, also to avoid confusion from ACE-inhibitor-induced cough. The glucagon-like 1 peptide receptor agonist (GLP-1 RA) might have an increasing role in obese COPD patients, especially if there is concomitant HF. The serial testing of NT-proBNP levels should be included in the post-discharge indications for follow-ups due to its potential usefulness in titrating the therapy for HF [210,211]; the optimal monitoring interval needs to be tailored to the underlying risk in the patient [212]. The attention from the cardiology community needs to greatly improve regarding the consequences of COPD as a dangerous comorbidity of HF and its relationship to other cardiopulmonary conditions. All these recommendations are especially important in older patients.

8. Outpatient Care

In cases of AECOPD not requiring hospitalization, appropriate outpatient care is also fundamental for the optimal management of the disease to reduce the risk of AECOPD and future hospital admissions. Several studies have demonstrated that each exacerbation increases the risk of future exacerbations in a graduated fashion [213,214,215,216] and that the grade of severity of each exacerbation increases the risk of death [215]. The effective prevention of further AECOPD occurrences relies on the timely management of exacerbations through appropriate therapy. The EROS real-world retrospective study demonstrated that initiating triple therapy with budesonide/glycopyrronium/formoterol fumarate within 30 days of a moderate or severe exacerbation in COPD patients is associated with a reduced risk of future exacerbations [217]. The study revealed a 24% decrease in exacerbation risk compared to delaying treatment by one to six months and a 34% decrease compared to delaying treatment for six months to one year [217].
Exacerbations increase the cardiovascular risk in COPD patients, as demonstrated by a study on 25,857 patients with COPD [218]. The study showed that the exacerbation increases the risk of myocardial infarction 2.7-fold and the risk of stroke 1.26-fold 1–5 days and 1–49 days after exacerbation, respectively in patients without a previous history of cardiovascular abnormalities [218]. According to a retrospective cohort study involving 355,978 patients with COPD, the occurrence of a single acute moderate or severe COPD exacerbation was found to increase the risk of an acute severe cardiovascular event by 32% within the first 30 days compared to individuals without a prior COPD exacerbation [219]. The study also revealed that the risk of cardiovascular events was most pronounced during the initial 90 days following the COPD exacerbation, and it remained elevated for a duration of one year [219]. Furthermore, each subsequent COPD exacerbation was associated with an even higher risk of acute cardiovascular events [219].
A phase 3 randomized, double-blind, 52-week trial comparing fluticasone furoate, umeclidinium, and vilanterol in different combinations in patients with COPD demonstrated an increased risk of cardiovascular adverse events, hospitalization, or death during moderate or severe AECOPD, which decreased over time [220]. Other risk factors for HF in patients affected by COPD are summarized in Figure 1.
Hospital admissions markedly increase the risk for death, as demonstrated by a study on a large population-based inception cohort of 73,106 patients with COPD with long-term follow-ups; 50% of patients with COPD died within 3.6 years after the first hospitalization and 75% within 7.7 years [213].
For these reasons, it is paramount that outpatient care provides an evaluation of COPD and cardiovascular risk. Each patient who accesses an outpatient facility with symptoms potentially caused by COPD and/or HF should receive an accurate risk stratification and be treated appropriately in relation to the identified condition. Clinicians should remember that the ultimate aim of outpatient care is to avoid exacerbations and hospital admissions. A true commitment of outpatient care to adequately treat patients with COPD, both with and without concomitant HF, with a synergistic approach by pulmonologists and cardiologists, with the monitoring of pulmonary and cardiac functions (including, if possible, serial testing of NT-proBNP levels to identify patients at high risk who need a modification of the therapy) at each visit and an appropriate use of β1-blockers when necessary would greatly reduce the risk for AECOPD and, as a consequence, the need for hospital admissions and the risk of death.

9. Conclusions

In this article, the authors have underlined several aspects that can help differentiate between COPD and HF, with a special emphasis on the need to differentiate between dyspnea due to COPD and due to HF in its different forms. At the same time the authors have underlined the importance of both diagnoses because COPD and HF often coexist, especially in older patients. To this end, the authors have provided a list of recommended laboratory and diagnostic tests, highlighting the value of NT-proBNP levels, the use of spirometry, and the importance of performing chest radiography in a seated position to rule out confounding factors due to pulmonary congestion and/or edema. Lung sonography also has an increasing role in internal medicine.
The authors have proposed an algorithm to improve the management of hospitalized patients, but greater efforts should be directed towards preventing hospitalizations due to AECOPD because they markedly increase the risk of future hospitalized exacerbations, cardiovascular complications, and death. Outpatient care plays a fundamental role as proper outpatient management could prevent hospital admissions of patients with COPD who have never been hospitalized before, greatly improving their prognosis. In this setting, the appropriate evaluation of global cardiovascular risk and appropriate treatment to reduce it are mandatory in patients with COPD who are often elderly and have several cardiovascular risk factors. The authors have also highlighted the need to treat COPD and HF appropriately even when they coexist, underlining the fact that selective β1-blockers, currently underused in patients with COPD and concomitant HF, are beneficial in these patients. While the connection between COPD and HF is the focus, COPD patients may have several other cardiopulmonary comorbidities and risks to manage; several recommendations (e.g., spirometry, cardiac risk assessment, etc.) could be applicable to these other conditions as well. The diagnostic and therapeutic path illustrated in this article may help avoid misdiagnosis and achieve an overall reduction in hospital readmissions, with better prognosis for patients and reduced costs related to COPD, especially when it coexists with HF.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm13061621/s1, Table S1: Management recommendations for clinicians at different stages of COPD patient journey; Table S2: COPD management recommendations for patient/caregiver.

Author Contributions

Conceptualization, all the authors; methodology, all the authors; formal analysis, all the authors; investigation, all the authors; resources, all the authors; data curation, all the authors; writing—original draft preparation, all the authors; writing—review and editing, all the authors; visualization, all the authors; supervision, all the authors; project administration, all the authors. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by an unconditional grant from AstraZeneca.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Editorial and medical writing assistance were provided, through EDRA S.p.A., by Alessia Scotton (Editorial Project Manager, Edra S.p.A, Milan, Italy) and Valeria Benedusi, respectively.

Conflicts of Interest

Andrea Bianco reports support for the present manuscript from AstraZeneca. Marco Canepa received two investigator-initiated grants from Pfizer; Giosuè Angelo Catapano received payments for lectures and presentations, speaker bureaus, manuscript writing or educational events, and support for attending meetings and traveling from Chiesi, AstraZeneca, GSK, Guidotti, and Menarini; Maurizio Marvisi has no conflict to declare; Fabrizio Oliva has no conflict to declare; Andrea Passantino received payments for presentations from AstraZeneca, Novordisk, Novartis, and Boehringer Ing.; Riccardo Sarzani received a scientific board participation fee from AstraZeneca; Paolo Tarsia declares payment for lectures, manuscript writing, or educational events from AstraZeneca, Daaiki Sankyo, GSK, Incyte, and Medicair; Giovanni Versace has nothing to declare. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Alshabanat, A.; Otterstatter, M.C.; Sin, D.D.; Road, J.; Rempel, C.; Burns, J.; van Eeden, S.F.; FitzGerald, J.M. Impact of a COPD comprehensive case management program on hospital length of stay and readmission rates. Int. J. Chronic Obstr. Pulm. Dis. 2017, 12, 961–971. [Google Scholar] [CrossRef]
  2. Mulpuru, S.; McKay, J.; Ronksley, P.E.; Thavorn, K.; Kobewka, D.M.; Forster, A.J. Factors contributing to high-cost hospital care for patients with COPD. Int. J. Chronic Obstr. Pulm. Dis. 2017, 12, 989–995. [Google Scholar] [CrossRef]
  3. Miravitlles, M.; Murio, C.; Guerrero, T.; Gisbert, R.; DAFNE Study Group. Decisiones sobre Antibioticoterapia y Farmacoeconomía en la EPOC, Pharmacoeconomic evaluation of acute exacerbations of chronic bronchitis and COPD. Chest 2002, 121, 1449–1455. [Google Scholar] [CrossRef] [PubMed]
  4. Press, V.G.; Konetzka, R.T.; White, S.R. Insights about the economic impact of chronic obstructive pulmonary disease readmissions post implementation of the hospital readmission reduction program. Curr. Opin. Pulm. Med. 2018, 24, 138–146. [Google Scholar] [CrossRef] [PubMed]
  5. McIlvennan, C.K.; Eapen, Z.J.; Allen, L.A. Hospital readmissions reduction program. Circulation 2015, 131, 1796–1803. [Google Scholar] [CrossRef]
  6. Criner, G.J.; Bourbeau, J.; Diekemper, R.L.; Ouellette, D.R.; Goodridge, D.; Hernandez, P.; Curren, K.; Balter, M.S.; Bhutani, M.; Camp, P.G.; et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest 2015, 147, 894–942. [Google Scholar] [CrossRef]
  7. Iheanacho, I.; Zhang, S.; King, D.; Rizzo, M.; Ismaila, A.S. Economic Burden of Chronic Obstructive Pulmonary Disease (COPD): A Systematic Literature Review. Int. J. Chronic Obstr. Pulm. Dis. 2020, 15, 439–460. [Google Scholar] [CrossRef]
  8. Green, S.A.; Bell, D.; Mays, N. Identification of factors that support successful implementation of care bundles in the acute medical setting: A qualitative study. BMC Health Serv. Res. 2017, 17, 120. [Google Scholar] [CrossRef]
  9. Dhalla, I.A.; O’Brien, T.; Ko, F.; Laupacis, A. Toward safer transitions: How can we reduce post-discharge adverse events? Healthc. Q. 2012, 15, 63–67. [Google Scholar] [CrossRef]
  10. Hussey, P.S.; Schneider, E.C.; Rudin, R.S.; Fox, D.S.; Lai, J.; Pollack, C.E. Continuity and the costs of care for chronic disease. JAMA Intern. Med. 2014, 174, 742–748. [Google Scholar] [CrossRef]
  11. 2024 GOLD Report. Global Initiative for Chronic Obstructive Lung Disease—GOLD (n.d.). Available online: https://goldcopd.org/2024-gold-report/ (accessed on 22 January 2024).
  12. Papi, A.; Bellettato, C.M.; Braccioni, F.; Romagnoli, M.; Casolari, P.; Caramori, G.; Fabbri, L.M.; Johnston, S.L. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am. J. Respir. Crit. Care Med. 2006, 173, 1114–1121. [Google Scholar] [CrossRef]
  13. Seemungal, T.; Harper-Owen, R.; Bhowmik, A.; Moric, I.; Sanderson, G.; Message, S.; Maccallum, P.; Meade, T.W.; Jeffries, D.J.; Johnston, S.L.; et al. Respiratory viruses; symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2001, 164, 1618–1623. [Google Scholar] [CrossRef]
  14. Sethi, S. Infectious etiology of acute exacerbations of chronic bronchitis. Chest 2000, 117, 380S–385S. [Google Scholar] [CrossRef]
  15. Wedzicha, J.A. Exacerbations: Etiology and pathophysiologic mechanisms. Chest 2002, 121, 136S–141S. [Google Scholar] [CrossRef]
  16. Sethi, S.; Murphy, T.F. Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N. Engl. J. Med. 2008, 359, 2355–2365. [Google Scholar] [CrossRef]
  17. Woodhead, M.; Blasi, F.; Ewig, S.; Huchon, G.; Ieven, M.; Leven, M.; Ortqvist, A.; Schaberg, T.; Torres, A.; van der Heijden, G.; et al. European Society of Clinical Microbiology and Infectious Diseases, Guidelines for the management of adult lower respiratory tract infections. Eur. Respir. J. 2005, 26, 1138–1180. [Google Scholar] [CrossRef]
  18. Hurst, J.R.; Wedzicha, J.A. The biology of a chronic obstructive pulmonary disease exacerbation. Clin. Chest Med. 2007, 28, 525–536. [Google Scholar] [CrossRef]
  19. Wedzicha, J.A.; Donaldson, G.C. Exacerbations of chronic obstructive pulmonary disease. Respir. Care 2003, 48, 1204–1213; discussion 1213–1215. [Google Scholar] [PubMed]
  20. Celli, B.R.; Barnes, P.J. Exacerbations of chronic obstructive pulmonary disease. Eur. Respir. J. 2007, 29, 1224–1238. [Google Scholar] [CrossRef] [PubMed]
  21. Anthonisen, N.R.; Manfreda, J.; Warren, C.P.; Hershfield, E.S.; Harding, G.K.; Nelson, N.A. Antibiotic therapy in exacerbations of chronic obstructive pulmonary disease. Ann. Intern. Med. 1987, 106, 196–204. [Google Scholar] [CrossRef] [PubMed]
  22. Stockley, R.A.; O’Brien, C.; Pye, A.; Hill, S.L. Relationship of sputum color to nature and outpatient management of acute exacerbations of COPD. Chest 2000, 117, 1638–1645. [Google Scholar] [CrossRef]
  23. Bozinovski, S.; Hutchinson, A.; Thompson, M.; Macgregor, L.; Black, J.; Giannakis, E.; Karlsson, A.-S.; Silvestrini, R.; Smallwood, D.; Vlahos, R.; et al. Serum amyloid a is a biomarker of acute exacerbations of chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2008, 177, 269–278. [Google Scholar] [CrossRef]
  24. Gallego, M.; Pomares, X.; Capilla, S.; Marcos, M.A.; Suárez, D.; Monsó, E.; Montón, C. C-reactive protein in outpatients with acute exacerbation of COPD: Its relationship with microbial etiology and severity. Int. J. Chronic Obstr. Pulm. Dis. 2016, 11, 2633–2640. [Google Scholar] [CrossRef]
  25. Hurst, J.R.; Donaldson, G.C.; Perera, W.R.; Wilkinson, T.M.A.; Bilello, J.A.; Hagan, G.W.; Vessey, R.S.; Wedzicha, J.A. Use of plasma biomarkers at exacerbation of chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2006, 174, 867–874. [Google Scholar] [CrossRef] [PubMed]
  26. Li, J.; Sun, S.; Tang, R.; Qiu, H.; Huang, Q.; Mason, T.G.; Tian, L. Major air pollutants and risk of COPD exacerbations: A systematic review and meta-analysis. Int. J. Chronic Obstr. Pulm. Dis. 2016, 11, 3079–3091. [Google Scholar] [CrossRef]
  27. Miravitlles, M.; Moragas, A.; Hernández, S.; Bayona, C.; Llor, C. Is it possible to identify exacerbations of mild to moderate COPD that do not require antibiotic treatment? Chest 2013, 144, 1571–1577. [Google Scholar] [CrossRef] [PubMed]
  28. Perera, W.R.; Hurst, J.R.; Wilkinson, T.M.A.; Sapsford, R.J.; Müllerova, H.; Donaldson, G.C.; Wedzicha, J.A. Inflammatory changes, recovery and recurrence at COPD exacerbation. Eur. Respir. J. 2007, 29, 527–534. [Google Scholar] [CrossRef] [PubMed]
  29. Sethi, S.; Evans, N.; Grant, B.J.B.; Murphy, T.F. New strains of bacteria and exacerbations of chronic obstructive pulmonary disease. N. Engl. J. Med. 2002, 347, 465–471. [Google Scholar] [CrossRef] [PubMed]
  30. Bakke, P.S.; Baste, V.; Hanoa, R.; Gulsvik, A. Prevalence of obstructive lung disease in a general population: Relation to occupational title and exposure to some airborne agents. Thorax 1991, 46, 863–870. [Google Scholar] [CrossRef]
  31. Tan, W.C.; Sin, D.D.; Bourbeau, J.; Hernandez, P.; Chapman, K.R.; Cowie, R.; FitzGerald, J.M.; Marciniuk, D.D.; Maltais, F.; Buist, A.S.; et al. Characteristics of COPD in never-smokers and ever-smokers in the general population: Results from the CanCOLD study. Thorax 2015, 70, 822–829. [Google Scholar] [CrossRef] [PubMed]
  32. Fuller-Thomson, E.; Chisholm, R.S.; Brennenstuhl, S. COPD in a Population-Based Sample of Never-Smokers: Interactions among Sex, Gender, and Race. Int. J. Chronic Dis. 2016, 2016, 5862026. [Google Scholar] [CrossRef]
  33. Smith, B.M.; Kirby, M.; Hoffman, E.A.; Kronmal, R.A.; Aaron, S.D.; Allen, N.B.; Bertoni, A.; Coxson, H.O.; Cooper, C.; Couper, D.J.; et al. Association of Dysanapsis With Chronic Obstructive Pulmonary Disease Among Older Adults. JAMA 2020, 323, 2268–2280. [Google Scholar] [CrossRef]
  34. Moran-Mendoza, O.; Pérez-Padilla, J.R.; Salazar-Flores, M.; Vazquez-Alfaro, F. Wood smoke-associated lung disease: A clinical; functional, radiological and pathological description. Int. J. Tuberc. Lung Dis. 2008, 12, 1092–1098. [Google Scholar]
  35. Lee, S.H.; Hwang, E.D.; Lim, J.E.; Moon, S.; Kang, Y.A.; Jung, J.Y.; Park, M.S.; Kim, S.K.; Chang, J.; Kim, Y.S.; et al. The Risk Factors and Characteristics of COPD Among Nonsmokers in Korea: An Analysis of KNHANES IV and V. Lung 2016, 194, 353–361. [Google Scholar] [CrossRef]
  36. Zhou, Y.; Wang, C.; Yao, W.; Chen, P.; Kang, J.; Huang, S.; Chen, B.; Wang, C.; Ni, D.; Wang, X.; et al. COPD in Chinese nonsmokers. Eur. Respir. J. 2009, 33, 509–518. [Google Scholar] [CrossRef]
  37. Thomsen, M.; Nordestgaard, B.G.; Vestbo, J.; Lange, P. Characteristics and outcomes of chronic obstructive pulmonary disease in never smokers in Denmark: A prospective population study. Lancet Respir. Med. 2013, 1, 543–550. [Google Scholar] [CrossRef] [PubMed]
  38. Golpe, R.; López, P.S.; Jiménez, E.C.; Añón, O.C.; de Llano, L.A.P. Distribution of clinical phenotypes in patients with chronic obstructive pulmonary disease caused by biomass and tobacco smoke. Arch. Bronconeumol. 2014, 50, 318–324. [Google Scholar] [CrossRef] [PubMed]
  39. Salvi, S.S.; Brashier, B.B.; Londhe, J.; Pyasi, K.; Vincent, V.; Kajale, S.S.; Tambe, S.; Mandani, K.; Nair, A.; Mak, S.M.; et al. Phenotypic comparison between smoking and non-smoking chronic obstructive pulmonary disease. Respir. Res. 2020, 21, 50. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, J.; Lin, X.; Bai, C. Comparison of clinical features between non-smokers with COPD and smokers with COPD: A retrospective observational study. Int. J. Chronic Obstr. Pulm. Dis. 2014, 9, 57–63. [Google Scholar] [CrossRef] [PubMed]
  41. Yang, I.A.; Jenkins, C.R.; Salvi, S.S. Chronic obstructive pulmonary disease in never-smokers: Risk factors, pathogenesis, and implications for prevention and treatment. Lancet Respir. Med. 2022, 10, 497–511. [Google Scholar] [CrossRef] [PubMed]
  42. Zhao, D.; Zhou, Y.; Jiang, C.; Zhao, Z.; He, F.; Ran, P. Small airway disease: A different phenotype of early stage COPD associated with biomass smoke exposure. Respirology 2018, 23, 198–205. [Google Scholar] [CrossRef]
  43. Krimmer, D.; Ichimaru, Y.; Burgess, J.; Black, J.; Oliver, B. Exposure to biomass smoke extract enhances fibronectin release from fibroblasts. PLoS ONE 2013, 8, e83938. [Google Scholar] [CrossRef]
  44. Bhome, A.B.; Brashier, B. Profiles of chronic obstructive lung disease: Characteristics of stable chronic obstructive lung disease in different parts of Asia. Curr. Opin. Pulm. Med. 2014, 20, 165–172. [Google Scholar] [CrossRef]
  45. Almagro, P.; Barreiro, B.; de Echaguen, A.O.; Quintana, S.; Carballeira, M.R.; Heredia, J.L.; Garau, J. Risk factors for hospital readmission in patients with chronic obstructive pulmonary disease. Respiration 2006, 73, 311–317. [Google Scholar] [CrossRef] [PubMed]
  46. Garcia-Aymerich, J.; Farrero, E.; Félez, M.A.; Izquierdo, J.; Marrades, R.M.; Antó, J.M. Estudi del Factors de Risc d’Agudització de la MPOC investigators, Risk factors of readmission to hospital for a COPD exacerbation: A prospective study. Thorax 2003, 58, 100–105. [Google Scholar] [CrossRef] [PubMed]
  47. Gudmundsson, G.; Gislason, T.; Janson, C.; Lindberg, E.; Hallin, R.; Ulrik, C.S.; Brøndum, E.; Nieminen, M.M.; Aine, T.; Bakke, P. Risk factors for rehospitalisation in COPD: Role of health status, anxiety and depression. Eur. Respir. J. 2005, 26, 414–419. [Google Scholar] [CrossRef]
  48. Mercer, P.; Shute, J.; Bhowmik, A.; Donaldson, G.; Wedzicha, J.; Warner, J. MMP-9, TIMP-1 and inflammatory cells in sputum from COPD patients during exacerbation. Respir. Res. 2005, 6, 151. [Google Scholar] [CrossRef] [PubMed]
  49. Donaldson, G.C.; Seemungal, T.A.R.; Bhowmik, A.; Wedzicha, J.A. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax 2002, 57, 847–852. [Google Scholar] [CrossRef] [PubMed]
  50. McGhan, R.; Radcliff, T.; Fish, R.; Sutherland, E.R.; Welsh, C.; Make, B. Predictors of rehospitalization and death after a severe exacerbation of COPD. Chest 2007, 132, 1748–1755. [Google Scholar] [CrossRef] [PubMed]
  51. Lau, A.; YAM, L.; Poon, E. Hospital re-admission in patients with acute exacerbation of chronic obstructive pulmonary disease. Respir. Med. 2001, 95, 876–884. [Google Scholar] [CrossRef] [PubMed]
  52. Nantsupawat, T.; Limsuwat, C.; Nugent, K. Factors affecting chronic obstructive pulmonary disease early rehospitalization. Chronic Respir. Dis. 2012, 9, 93–98. [Google Scholar] [CrossRef] [PubMed]
  53. Papaioannou, A.I.; Bartziokas, K.; Tsikrika, S.; Karakontaki, F.; Kastanakis, E.; Banya, W.; Haniotou, A.; Papiris, S.; Loukides, S.; Polychronopoulos, V.; et al. The impact of depressive symptoms on recovery and outcome of hospitalised COPD exacerbations. Eur. Respir. J. 2013, 41, 815–823. [Google Scholar] [CrossRef] [PubMed]
  54. Dransfield, M.T.; Criner, G.J.; Halpin, D.M.G.; Han, M.K.; Hartley, B.; Kalhan, R.; Lange, P.; Lipson, D.A.; Martinez, F.J.; Midwinter, D.; et al. Time-Dependent Risk of Cardiovascular Events following an Exacerbation in Patients with Chronic Obstructive Pulmonary Disease: Post Hoc Analysis from the IMPACT Trial. J. Am. Heart Assoc. 2022, 11, e024350. [Google Scholar] [CrossRef]
  55. Liu, X.; Chen, Z.; Li, S.; Xu, S. Association of Chronic Obstructive Pulmonary Disease With Arrhythmia Risks: A Systematic Review and Meta-Analysis. Front. Cardiovasc. Med. 2021, 8, 732349. [Google Scholar] [CrossRef]
  56. Terzano, C.; Romani, S.; Conti, V.; Paone, G.; Oriolo, F.; Vitarelli, A. Atrial fibrillation in the acute, hypercapnic exacerbations of COPD. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 2908–2917. [Google Scholar] [PubMed]
  57. Houben-Wilke, S.; Jörres, R.A.; Bals, R.; Franssen, F.M.E.; Gläser, S.; Holle, R.; Karch, A.; Koch, A.; Magnussen, H.; Obst, A.; et al. Peripheral Artery Disease and Its Clinical Relevance in Patients with Chronic Obstructive Pulmonary Disease in the COPD and Systemic Consequences-Comorbidities Network Study. Am. J. Respir. Crit. Care Med. 2017, 195, 189–197. [Google Scholar] [CrossRef]
  58. Le Jemtel, T.H.; Padeletti, M.; Jelic, S. Diagnostic and therapeutic challenges in patients with coexistent chronic obstructive pulmonary disease and chronic heart failure. J. Am. Coll. Cardiol. 2007, 49, 171–180. [Google Scholar] [CrossRef]
  59. Rutten, F.H.; Cramer, M.-J.M.; Grobbee, D.E.; Sachs, A.P.E.; Kirkels, J.H.; Lammers, J.-W.J.; Hoes, A.W. Unrecognized heart failure in elderly patients with stable chronic obstructive pulmonary disease. Eur. Heart J. 2005, 26, 1887–1894. [Google Scholar] [CrossRef]
  60. Canepa, M.; Temporelli, P.L.; Rossi, A.; Rossi, A.; Gonzini, L.; Nicolosi, G.L.; Staszewsky, L.; Marchioli, R.; Maggioni, A.P.; Tavazzi, L. Prevalence and Prognostic Impact of Chronic Obstructive Pulmonary Disease in Patients with Chronic Heart Failure: Data from the GISSI-HF Trial. Cardiology 2017, 136, 128–137. [Google Scholar] [CrossRef]
  61. Hawkins, N.M.; Petrie, M.C.; Jhund, P.S.; Chalmers, G.W.; Dunn, F.G.; McMurray, J.J.V. Heart failure and chronic obstructive pulmonary disease: Diagnostic pitfalls and epidemiology. Eur. J. Heart Fail. 2009, 11, 130–139. [Google Scholar] [CrossRef]
  62. Güder, G.; Brenner, S.; Störk, S.; Hoes, A.; Rutten, F.H. Chronic obstructive pulmonary disease in heart failure: Accurate diagnosis and treatment. Eur. J. Heart Fail. 2014, 16, 1273–1282. [Google Scholar] [CrossRef] [PubMed]
  63. Gustafsson, F.; Torp-Pedersen, C.; Brendorp, B.; Seibaek, M.; Burchardt, H.; Køber, L. Long-term survival in patients hospitalized with congestive heart failure: Relation to preserved and reduced left ventricular systolic function. Eur. Heart J. 2003, 24, 863–870. [Google Scholar] [CrossRef] [PubMed]
  64. Wilhelmsen, L.; Rosengren, A.; Eriksson, H.; Lappas, G. Heart failure in the general population of men—Morbidity, risk factors and prognosis. J. Intern. Med. 2001, 249, 253–261. [Google Scholar] [CrossRef]
  65. He, J.; Ogden, L.G.; Bazzano, L.A.; Vupputuri, S.; Loria, C.; Whelton, P.K. Risk factors for congestive heart failure in US men and women: NHANES I epidemiologic follow-up study. Arch. Intern. Med. 2001, 161, 996–1002. [Google Scholar] [CrossRef] [PubMed]
  66. Sin, D.D.; Man, S.F.P. Why are patients with chronic obstructive pulmonary disease at increased risk of cardiovascular diseases? The potential role of systemic inflammation in chronic obstructive pulmonary disease. Circulation 2003, 107, 1514–1519. [Google Scholar] [CrossRef]
  67. Ross, R. Atherosclerosis—An inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef]
  68. Man, S.F.P.; Connett, J.E.; Anthonisen, N.R.; Wise, R.A.; Tashkin, D.P.; Sin, D.D. C-reactive protein and mortality in mild to moderate chronic obstructive pulmonary disease. Thorax 2006, 61, 849–853. [Google Scholar] [CrossRef]
  69. Ouwerkerk, W.; Voors, A.A.; Anker, S.D.; Cleland, J.G.; Dickstein, K.; Filippatos, G.; van der Harst, P.; Hillege, H.L.; Lang, C.C.; Maaten, J.M.T.; et al. Determinants and clinical outcome of uptitration of ACE-inhibitors and beta-blockers in patients with heart failure: A prospective European study. Eur. Heart J. 2017, 38, 1883–1890. [Google Scholar] [CrossRef]
  70. Staszewsky, L.; Wong, M.; Masson, S.; Barlera, S.; Carretta, E.; Maggioni, A.P.; Anand, I.S.; Cohn, J.N.; Tognoni, G.; Latini, R. Valsartan Heart Failure Trial Investigators, Clinical, neurohormonal, and inflammatory markers and overall prognostic role of chronic obstructive pulmonary disease in patients with heart failure: Data from the Val-HeFT heart failure trial. J. Card. Fail. 2007, 13, 797–804. [Google Scholar] [CrossRef]
  71. Rabe, K.F.; Hurst, J.R.; Suissa, S. Cardiovascular disease and COPD: Dangerous liaisons? Eur. Respir. Rev. 2018, 27, 180057. [Google Scholar] [CrossRef]
  72. Hawkins, N.M.; Virani, S.; Ceconi, C. Heart failure and chronic obstructive pulmonary disease: The challenges facing physicians and health services. Eur. Heart J. 2013, 34, 2795–2803. [Google Scholar] [CrossRef] [PubMed]
  73. Müllerova, H.; Agusti, A.; Erqou, S.; Mapel, D.W. Cardiovascular comorbidity in COPD: Systematic literature review. Chest 2013, 144, 1163–1178. [Google Scholar] [CrossRef]
  74. Axson, E.L.; Ragutheeswaran, K.; Sundaram, V.; Bloom, C.I.; Bottle, A.; Cowie, M.R.; Quint, J.K. Hospitalisation and mortality in patients with comorbid COPD and heart failure: A systematic review and meta-analysis. Respir. Res. 2020, 21, 54. [Google Scholar] [CrossRef] [PubMed]
  75. Canepa, M.; Straburzynska-Migaj, E.; Drozdz, J.; Fernandez-Vivancos, C.; Pinilla, J.M.G.; Nyolczas, N.; Temporelli, P.L.; Mebazaa, A.; Lainscak, M.; Laroche, C.; et al. ESC-HFA Heart Failure Long-Term Registry Investigators, Characteristics, treatments and 1-year prognosis of hospitalized and ambulatory heart failure patients with chronic obstructive pulmonary disease in the European Society of Cardiology Heart Failure Long-Term Registry. Eur. J. Heart Fail. 2018, 20, 100–110. [Google Scholar] [CrossRef]
  76. Fisher, K.A.; Stefan, M.S.; Darling, C.; Lessard, D.; Goldberg, R.J. Impact of COPD on the mortality and treatment of patients hospitalized with acute decompensated heart failure: The Worcester Heart Failure Study. Chest 2015, 147, 637–645. [Google Scholar] [CrossRef] [PubMed]
  77. Roversi, S.; Fabbri, L.; Sin, D.; Hawkins, N.; Agustì, A. Chronic Obstructive Pulmonary Disease and Cardiac Diseases. An Urgen. Need for Integrated Care. Am. J. Respir. Crit. Care Med. 2016, 194, 1319–1336. [Google Scholar] [CrossRef]
  78. Braunstein, J.B.; Anderson, G.F.; Gerstenblith, G.; Weller, W.; Niefeld, M.; Herbert, R.; Wu, A.W. Noncardiac comorbidity increases preventable hospitalizations and mortality among Medicare beneficiaries with chronic heart failure. J. Am. Coll. Cardiol. 2003, 42, 1226–1233. [Google Scholar] [CrossRef]
  79. Rusinaru, D.; Saaidi, I.; Godard, S.; Mahjoub, H.; Battle, C.; Tribouilloy, C. Impact of chronic obstructive pulmonary disease on long-term outcome of patients hospitalized for heart failure. Am. J. Cardiol. 2008, 101, 353–358. [Google Scholar] [CrossRef]
  80. Mentz, R.J.; Fiuzat, M.; Wojdyla, D.M.; Chiswell, K.; Gheorghiade, M.; Fonarow, G.C.; O’Connor, C.M. Clinical characteristics and outcomes of hospitalized heart failure patients with systolic dysfunction and chronic obstructive pulmonary disease: Findings from OPTIMIZE-HF. Eur. J. Heart Fail. 2012, 14, 395–403. [Google Scholar] [CrossRef]
  81. Jacob, J.; Tost, J.; Miró, Ò.; Herrero, P.; Martín-Sánchez, F.J.; Llorens, P. Impact of chronic obstructive pulmonary disease on clinical course after an episode of acute heart failure. EAHFE–COPD study. Int. J. Cardiol. 2017, 227, 450–456. [Google Scholar] [CrossRef]
  82. Tavazzi, L.; Swedberg, K.; Komajda, M.; Böhm, M.; Borer, J.S.; Lainscak, M.; Robertson, M.; Ford, I.; Investigators, S.H.F. Clinical profiles and outcomes in patients with chronic heart failure and chronic obstructive pulmonary disease: An efficacy and safety analysis of SHIFT study. Int. J. Cardiol. 2013, 170, 182–188. [Google Scholar] [CrossRef]
  83. De Blois, J.; Simard, S.; Atar, D.; Agewall, S. Norwegian Heart Failure Registry, COPD predicts mortality in HF: The Norwegian Heart Failure Registry. J. Card. Fail. 2010, 16, 225–229. [Google Scholar] [CrossRef]
  84. Walke, L.M.; Byers, A.L.; Tinetti, M.E.; Dubin, J.A.; McCorkle, R.; Fried, T.R. Range and severity of symptoms over time among older adults with chronic obstructive pulmonary disease and heart failure. Arch. Intern. Med. 2007, 167, 2503–2508. [Google Scholar] [CrossRef]
  85. de Souza Caroci, A.; Lareau, S.C. Descriptors of dyspnea by patients with chronic obstructive pulmonary disease versus congestive heart failure. Heart Lung 2004, 33, 102–110. [Google Scholar] [CrossRef]
  86. Zvezdin, B.; Milutinov, S.; Kojicic, M.; Hadnadjev, M.; Hromis, S.; Markovic, M.; Gajic, O. A postmortem analysis of major causes of early death in patients hospitalized with COPD exacerbation. Chest 2009, 136, 376–380. [Google Scholar] [CrossRef]
  87. Mannino, D.M.; Doherty, D.E.; Buist, A.S. Global Initiative on Obstructive Lung Disease (GOLD) classification of lung disease and mortality: Findings from the Atherosclerosis Risk in Communities (ARIC) study. Respir. Med. 2006, 100, 115–122. [Google Scholar] [CrossRef]
  88. Masa, J.F.; Utrabo, I.; de Terreros, J.G.; Aburto, M.; Esteban, C.; Prats, E.; Núñez, B.; Ortega-González, Á.; Jara-Palomares, L.; Martin-Vicente, M.J.; et al. Noninvasive ventilation for severely acidotic patients in respiratory intermediate care units: Precision medicine in intermediate care units. BMC Pulm. Med. 2016, 16, 97. [Google Scholar] [CrossRef] [PubMed]
  89. Dransfield, M.T.; Voelker, H.; Bhatt, S.P.; Brenner, K.; Casaburi, R.; Come, C.E.; Cooper, J.A.D.; Criner, G.J.; Curtis, J.L.; Han, M.K.; et al. Metoprolol for the Prevention of Acute Exacerbations of COPD. N. Engl. J. Med. 2019, 381, 2304–2314. [Google Scholar] [CrossRef] [PubMed]
  90. Vogelmeier, C.F.; Criner, G.J.; Martinez, F.J.; Anzueto, A.; Barnes, P.J.; Bourbeau, J.; Celli, B.R.; Chen, R.; Decramer, M.; Fabbri, L.M.; et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD Executive Summary. Am. J. Respir. Crit. Care Med. 2017, 195, 557–582. [Google Scholar] [CrossRef] [PubMed]
  91. Canepa, M.; Franssen, F.M.E.; Olschewski, H.; Lainscak, M.; Böhm, M.; Tavazzi, L.; Rosenkranz, S. Diagnostic and Therapeutic Gaps in Patients With Heart Failure and Chronic Obstructive Pulmonary Disease. JACC Heart Fail. 2019, 7, 823–833. [Google Scholar] [CrossRef] [PubMed]
  92. Kong, C.W.; Wilkinson, T.M.A. Predicting and preventing hospital readmission for exacerbations of COPD. ERJ Open Res. 2020, 6, 00325-2019. [Google Scholar] [CrossRef]
  93. Press, V.G.; Myers, L.C.; Feemster, L.C. Preventing COPD Readmissions Under the Hospital Readmissions Reduction Program. Chest 2021, 159, 996–1006. [Google Scholar] [CrossRef]
  94. Sharpe, I.; Bowman, M.; Kim, A.; Srivastava, S.; Jalink, M.; Wijeratne, D.T. Strategies to Prevent Readmissions to Hospital for COPD: A Systematic Review. COPD J. Chronic Obstr. Pulm. Dis. 2021, 18, 456–468. [Google Scholar] [CrossRef]
  95. Wang, L.; Li, G.; Ezeana, C.F.; Ogunti, R.; Puppala, M.; He, T.; Yu, X.; Wong, S.S.Y.; Yin, Z.; Roberts, A.W.; et al. An AI-driven clinical care pathway to reduce 30-day readmission for chronic obstructive pulmonary disease (COPD) patients. Sci. Rep. 2022, 12, 20633. [Google Scholar] [CrossRef]
  96. Bamforth, R.J.; Chhibba, R.; Ferguson, T.W.; Sabourin, J.; Pieroni, D.; Askin, N.; Tangri, N.; Komenda, P.; Rigatto, C. Strategies to prevent hospital readmission and death in patients with chronic heart failure, chronic obstructive pulmonary disease, and chronic kidney disease: A systematic review and meta-analysis. PLoS ONE 2021, 16, e0249542. [Google Scholar] [CrossRef]
  97. Morgenthaler, T.I.; Lim, K.; Larson, M.; Helfinstine, K.; Homan, J.; Schwarz, R.; Dankbar, G. A Practice Redesign Collaborative for Reducing Hospital Readmission for Chronic Obstructive Pulmonary Disease in an Affiliated Network of Health Care Organizations. Jt. Comm. J. Qual. Patient Saf. 2021, 47, 412–421. [Google Scholar] [CrossRef]
  98. Kalhan, R.; Mutharasan, R.K. Reducing Readmissions in Patients With Both Heart Failure and COPD. Chest 2018, 154, 1230–1238. [Google Scholar] [CrossRef]
  99. Celli, B.R.; Fabbri, L.M.; Aaron, S.D.; Agusti, A.; Brook, R.D.; Criner, G.J.; Franssen, F.M.E.; Humbert, M.; Hurst, J.R.; de Oca, M.M.; et al. Differential Diagnosis of Suspected COPD Exacerbations in the Acute Care Setting: Best Practice. Am. J. Respir. Crit. Care Med. 2023, 207, 1134–1144. [Google Scholar] [CrossRef] [PubMed]
  100. Using Care Bundles to Improve Health Care Quality|IHI—Institute for Healthcare Improvement, (n.d.). Available online: https://www.ihi.org:443/resources/Pages/IHIWhitePapers/UsingCareBundles.aspx (accessed on 17 December 2022).
  101. Turner, A.M.; Lim, W.S.; Rodrigo, C.; Welham, S.A.; Calvert, J.M. A care-bundles approach to improving standard of care in AECOPD admissions: Results of a national project. Thorax 2015, 70, 992–994. [Google Scholar] [CrossRef] [PubMed]
  102. Stone, P.W.; Adamson, A.; Hurst, J.R.; Roberts, C.M.; Quint, J.K. Does pay-for-performance improve patient outcomes in acute exacerbation of COPD admissions? Thorax 2022, 77, 239–246. [Google Scholar] [CrossRef] [PubMed]
  103. Wedzicha, J.A.; Calverley, P.M.A.; Albert, R.K.; Anzueto, A.; Criner, G.J.; Hurst, J.R.; Miravitlles, M.; Papi, A.; Rabe, K.F.; Rigau, D.; et al. Prevention of COPD exacerbations: A European Respiratory Society/American Thoracic Society guideline. Eur. Respir. J. 2017, 50, 1602265. [Google Scholar] [CrossRef] [PubMed]
  104. Laennec, R.; Forbes, J.; Fisher, J.; Andral, G. A Treatise on the Diseases of the Chest, and on Mediate Auscultation—Digital Collections—National Library of Medicine, Medicine in the Americas, 1610–1920 (n.d.). Available online: https://collections.nlm.nih.gov/catalog/nlm:nlmuid-9308216-bk (accessed on 21 November 2022).
  105. Calverley, P.M.A. Minimal clinically important difference—Exacerbations of COPD. COPD J. Chronic Obstr. Pulm. Dis. 2005, 2, 143–148. [Google Scholar] [CrossRef] [PubMed]
  106. Effing, T.W.; Kerstjens, H.A.M.; Monninkhof, E.M.; van der Valk, P.D.L.P.M.; Wouters, E.F.M.; Postma, D.S.; Zielhuis, G.A.; van der Palen, J. Definitions of exacerbations: Does it really matter in clinical trials on COPD? Chest 2009, 136, 918–923. [Google Scholar] [CrossRef] [PubMed]
  107. Stolz, D.; Mkorombindo, T.; Schumann, D.M.; Agusti, A.; Ash, S.Y.; Bafadhel, M.; Bai, C.; Chalmers, J.D.; Criner, G.J.; Dharmage, S.C.; et al. Towards the elimination of chronic obstructive pulmonary disease: A Lancet Commission. Lancet 2022, 400, 921–972. [Google Scholar] [CrossRef]
  108. Rennard, S.I. Overview of causes of COPD. New understanding of pathogenesis and mechanisms can guide future therapy. Postgrad. Med. 2002, 111, 28–30, 33–34, 37–38. [Google Scholar] [CrossRef] [PubMed]
  109. Renier, W.; Winckelmann, K.H.; Verbakel, J.Y.; Aertgeerts, B.; Buntinx, F. Signs and symptoms in adult patients with acute dyspnea: A systematic review and meta-analysis. Eur. J. Emerg. Med. 2018, 25, 3–11. [Google Scholar] [CrossRef]
  110. Collet, J.-P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Rev. Esp. Cardiol. Engl. Ed. 2021, 74, 544. [Google Scholar] [CrossRef]
  111. Hawkins, N.M.; Peterson, S.; Ezzat, A.M.; Vijh, R.; Virani, S.A.; Gibb, A.; Mancini, G.B.J.; Wong, S.T. Control of Cardiovascular Risk Factors in Patients with Chronic Obstructive Pulmonary Disease. Ann. Am. Thorac. Soc. 2022, 19, 1102–1111. [Google Scholar] [CrossRef]
  112. Crisafulli, E.; Manco, A.; Ferrer, M.; Huerta, A.; Micheletto, C.; Girelli, D.; Clini, E.; Torres, A. Pneumonic versus Nonpneumonic Exacerbations of Chronic Obstructive Pulmonary Disease. Semin. Respir. Crit. Care Med. 2020, 41, 817–829. [Google Scholar] [CrossRef]
  113. Torres, A.; Blasi, F.; Dartois, N.; Akova, M. Which individuals are at increased risk of pneumococcal disease and why? Impact of COPD, asthma, smoking, diabetes, and/or chronic heart disease on community-acquired pneumonia and invasive pneumococcal disease. Thorax 2015, 70, 984–989. [Google Scholar] [CrossRef]
  114. Davie, A.P.; Francis, C.M.; Love, M.P.; Caruana, L.; Starkey, I.R.; Shaw, T.R.; Sutherland, G.R.; McMurray, J.J. Value of the electrocardiogram in identifying heart failure due to left ventricular systolic dysfunction. BMJ 1996, 312, 222. [Google Scholar] [CrossRef]
  115. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 2021, 42, 373–498. [Google Scholar] [CrossRef] [PubMed]
  116. Ferry, O.R.; Huang, Y.C.; Masel, P.J.; Hamilton, M.; Fong, K.M.; Bowman, R.V.; McKenzie, S.C.; Yang, I.A. Diagnostic approach to chronic dyspnoea in adults. J. Thorac. Dis. 2019, 11, S2117–S2128. [Google Scholar] [CrossRef] [PubMed]
  117. Allen, C.J.; Guha, K.; Sharma, R. How to Improve Time to Diagnosis in Acute Heart Failure—Clinical Signs and Chest X-ray. Card. Fail. Rev. 2015, 1, 69–74. [Google Scholar] [CrossRef]
  118. Debray, M.P.; Carette, M.F.; Loubet, P.; Pasquet, B.; Fidouh, N.H.; Benjoar, M.; Varon, E.; Brun, A.L.; Claessens, Y.E.; Duval, X.; et al. CT features of community-acquired pneumonia at the emergency department. Respir. Med. Res. 2022, 81, 100892. [Google Scholar] [CrossRef]
  119. Bafadhel, M.; Clark, T.W.; Reid, C.; Medina, M.-J.; Batham, S.; Barer, M.R.; Nicholson, K.G.; Brightling, C.E. Procalcitonin and C-reactive protein in hospitalized adult patients with community-acquired pneumonia or exacerbation of asthma or COPD. Chest 2011, 139, 1410–1418. [Google Scholar] [CrossRef]
  120. Elmenawi, K.A.; Anil, V.; Gosal, H.; Kaur, H.; Ngassa, H.C.; Mohammed, L. The Importance of Measuring Troponin in Chronic Obstructive Pulmonary Disease Exacerbations: A Systematic Review. Cureus 2021, 13, e17451. [Google Scholar] [CrossRef] [PubMed]
  121. Thygesen, K.; Alpert, J.S.; Jaffe, A.S.; Chaitman, B.R.; Bax, J.J.; Morrow, D.A.; White, H.D. Executive Group on behalf of the Joint European Society of Cardiology (ESC)/American College of Cardiology (ACC)/American Heart Association (AHA)/World Heart Federation (WHF) Task Force for the Universal Definition of Myocardial Infarction, Fourth Universal Definition of Myocardial Infarction. Circulation 2018, 138, e618–e651. [Google Scholar] [CrossRef]
  122. Silver, M.A.; Maisel, A.; Yancy, C.W.; McCullough, P.A.; Burnett, J.C.; Francis, G.S.; Mehra, M.R.; Peacock, W.F.; Fonarow, G.; Gibler, W.B.; et al. BNP Consensus Panel 2004: A clinical approach for the diagnostic, prognostic, screening, treatment monitoring, and therapeutic roles of natriuretic peptides in cardiovascular diseases. Congest. Heart Fail. 2004, 10, 1–30. [Google Scholar] [CrossRef]
  123. Maisel, A.S.; Krishnaswamy, P.; Nowak, R.M.; McCord, J.; Hollander, J.E.; Duc, P.; Omland, T.; Storrow, A.B.; Abraham, W.T.; Wu, A.H.B.; et al. Breathing Not Properly Multinational Study Investigators, Rapid measurement of B-type natriuretic peptide in the emergency diagnosis of heart failure. N. Engl. J. Med. 2002, 347, 161–167. [Google Scholar] [CrossRef]
  124. Januzzi, J.L.; van Kimmenade, R.; Lainchbury, J.; Bayes-Genis, A.; Ordonez-Llanos, J.; Santalo-Bel, M.; Pinto, Y.M.; Richards, M. NT-proBNP testing for diagnosis and short-term prognosis in acute destabilized heart failure: An. international pooled analysis of 1256 patients: The International Collaborative of NT-proBNP Study. Eur. Heart J. 2006, 27, 330–337. [Google Scholar] [CrossRef]
  125. Sarzani, R.; Allevi, M.; Di Pentima, C.; Schiavi, P.; Spannella, F.; Giulietti, F. Role of Cardiac Natriuretic Peptides in Heart Structure and Function. Int. J. Mol. Sci. 2022, 23, 14415. [Google Scholar] [CrossRef]
  126. McCullough, P.A.; Hollander, J.E.; Nowak, R.M.; Storrow, A.B.; Duc, P.; Omland, T.; McCord, J.; Herrmann, H.C.; Steg, P.G.; Westheim, A.; et al. BNP Multinational Study Investigators, Uncovering heart failure in patients with a history of pulmonary disease: Rationale for the early use of B-type natriuretic peptide in the emergency department. Acad. Emerg. Med. 2003, 10, 198–204. [Google Scholar] [CrossRef]
  127. Morrison, L.K.; Harrison, A.; Krishnaswamy, P.; Kazanegra, R.; Clopton, P.; Maisel, A. Utility of a rapid B-natriuretic peptide assay in differentiating congestive heart failure from lung disease in patients presenting with dyspnea. J. Am. Coll. Cardiol. 2002, 39, 202–209. [Google Scholar] [CrossRef]
  128. Hawkins, N.M.; Khosla, A.; Virani, S.A.; McMurray, J.J.V.; FitzGerald, J.M. B-type natriuretic peptides in chronic obstructive pulmonary disease: A systematic review. BMC Pulm. Med. 2017, 17, 11. [Google Scholar] [CrossRef]
  129. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: Executive Summary: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, e876–e894. [Google Scholar] [CrossRef] [PubMed]
  130. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: Developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) With the special contribution of the Heart Failure Association (HFA) of the ESC. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  131. Stolz, D.; Breidthardt, T.; Christ-Crain, M.; Bingisser, R.; Miedinger, D.; Leuppi, J.; Mueller, B.; Tamm, M.; Mueller, C. Use of B-type natriuretic peptide in the risk stratification of acute exacerbations of COPD. Chest 2008, 133, 1088–1094. [Google Scholar] [CrossRef] [PubMed]
  132. Sarzani, R.; Spannella, F.; Giulietti, F.; Fedecostante, M.; Giordano, P.; Gattafoni, P.; Espinosa, E.; Busco, F.; Piccinini, G.; Dessì-Fulgheri, P. NT-proBNP and Its Correlation with In-Hospital Mortality in the Very Elderly without an Admission Diagnosis of Heart Failure. PLoS ONE 2016, 11, e0153759. [Google Scholar] [CrossRef] [PubMed]
  133. Januzzi, J.L.; Chen-Tournoux, A.A.; Moe, G. Amino-terminal pro-B-type natriuretic peptide testing for the diagnosis or exclusion of heart failure in patients with acute symptoms. Am. J. Cardiol. 2008, 101, 29–38. [Google Scholar] [CrossRef]
  134. Di Somma, S.; Magrini, L.; Pittoni, V.; Marino, R.; Mastrantuono, A.; Ferri, E.; Ballarino, P.; Semplicini, A.; Bertazzoni, G.; Carpinteri, G.; et al. In-hospital percentage BNP reduction is highly predictive for adverse events in patients admitted for acute heart failure: The Italian RED Study. Crit. Care 2010, 14, R116. [Google Scholar] [CrossRef] [PubMed]
  135. Di Somma, S.; Marino, R.; Zampini, G.; Magrini, L.; Ferri, E.; Shah, K.; Clopton, P.; Maisel, A.S. Predictive value for death and rehospitalization of 30-day postdischarge B-type natriuretic peptide (BNP) in elderly patients with heart failure. Sub-analysis of Italian RED Study. Clin. Chem. Lab. Med. 2015, 53, 507–513. [Google Scholar] [CrossRef] [PubMed]
  136. Di Somma, S.; Magrini, L.; Tabacco, F.; Marino, R.; Talucci, V.; Marrocco, F.; Cardelli, P.; Ferri, E.; Pittoni, V. Brain natriuretic peptide and N-terminal pro-B-type natriuretic peptide show a different profile in response to acute decompensated heart failure treatment. Congest. Heart Fail. 2008, 14, 245–250. [Google Scholar] [CrossRef] [PubMed]
  137. Oudejans, I.; Mosterd, A.; Bloemen, J.A.; Valk, M.J.; van Velzen, E.; Wielders, J.P.; Zuithoff, N.P.; Rutten, F.H.; Hoes, A.W. Clinical evaluation of geriatric outpatients with suspected heart failure: Value of symptoms, signs, and additional tests. Eur. J. Heart Fail. 2011, 13, 518–527. [Google Scholar] [CrossRef] [PubMed]
  138. Nagueh, S.F.; Appleton, C.P.; Gillebert, T.C.; Marino, P.N.; Oh, J.K.; Smiseth, O.A.; Waggoner, A.D.; Flachskampf, F.A.; Pellikka, P.A.; Evangelisa, A. Recommendations for the evaluation of left ventricular diastolic function by echocardiography. Eur. J. Echocardiogr. 2009, 10, 165–193. [Google Scholar] [CrossRef] [PubMed]
  139. Vaes, B.; Delgado, V.; Bax, J.; Degryse, J.; Westendorp, R.G.J.; Gussekloo, J. Diagnostic accuracy of plasma NT-proBNP levels for excluding cardiac abnormalities in the very elderly. BMC Geriatr. 2010, 10, 85. [Google Scholar] [CrossRef]
  140. Vaes, B.; de Ruijter, W.; Degryse, J.; Westendorp, R.G.J.; Gussekloo, J. Clinical relevance of a raised plasma N-terminal pro-brain natriuretic peptide level in a population-based cohort of nonagenarians. J. Am. Geriatr. Soc. 2009, 57, 823–829. [Google Scholar] [CrossRef]
  141. Richards, M.; Di Somma, S.; Mueller, C.; Nowak, R.; Peacock, W.F.; Ponikowski, P.; Mockel, M.; Hogan, C.; Wu, A.H.B.; Clopton, P.; et al. Atrial fibrillation impairs the diagnostic performance of cardiac natriuretic peptides in dyspneic patients: Results from the BACH Study (Biomarkers in ACute Heart Failure). JACC Heart Fail. 2013, 1, 192–199. [Google Scholar] [CrossRef]
  142. Leuchte, H.H.; Baumgartner, R.A.; Nounou, M.E.; Vogeser, M.; Neurohr, C.; Trautnitz, M.; Behr, J. Brain natriuretic peptide is a prognostic parameter in chronic lung disease. Am. J. Respir. Crit. Care Med. 2006, 173, 744–750. [Google Scholar] [CrossRef]
  143. Bayes-Genis, A.; Docherty, K.F.; Petrie, M.C.; Januzzi, J.L.; Mueller, C.; Anderson, L.; Bozkurt, B.; Butler, J.; Chioncel, O.; Cleland, J.G.F.; et al. Practical algorithms for early diagnosis of heart failure and heart stress using NT-proBNP: A clinical consensus statement from the Heart Failure Association of the ESC. Eur. J. Heart Fail. 2023, 25, 1891–1898. [Google Scholar] [CrossRef]
  144. COPD & Spirometry|British Thoracic Society|Better Lung Health for All, (n.d.). Available online: https://www.brit-thoracic.org.uk/quality-improvement/clinical-resources/copd-spirometry/ (accessed on 17 December 2022).
  145. Evans, K.A.; Pollack, M.; Portillo, E.; Strange, C.; Touchette, D.R.; Staresinic, A.; Patel, S.; Tkacz, J.; Feigler, N. Prompt initiation of triple therapy following hospitalization for a chronic obstructive pulmonary disease exacerbation in the United States: An analysis of the PRIMUS study. J. Manag. Care Spec. Pharm. 2022, 28, 1366–1377. [Google Scholar] [CrossRef]
  146. Halpin, D.M.G.; Dickens, A.P.; Skinner, D.; Murray, R.; Singh, M.; Hickman, K.; Carter, V.; Couper, A.; Evans, A.; Pullen, R.; et al. Identification of key opportunities for optimising the management of high-risk COPD patients in the UK using the CONQUEST quality standards: An observational longitudinal study. Lancet Reg. Health Eur. 2023, 29, 100619. [Google Scholar] [CrossRef]
  147. Light, R.W.; George, R.B. Serial pulmonary function in patients with acute heart failure. Arch. Intern. Med. 1983, 143, 429–433. [Google Scholar] [CrossRef]
  148. Brenner, S.; Güder, G.; Berliner, D.; Deubner, N.; Fröhlich, K.; Ertl, G.; Jany, B.; Angermann, C.E.; Störk, S. Airway obstruction in systolic heart failure—COPD or congestion? Int. J. Cardiol. 2013, 168, 1910–1916. [Google Scholar] [CrossRef]
  149. Kostikas, K.; Rhee, C.K.; Hurst, J.R.; Agostoni, P.; Cao, H.; Fogel, R.; Jones, R.; Kocks, J.W.H.; Mezzi, K.; Ming, S.W.Y.; et al. Adequacy of Therapy for People with Both COPD and Heart Failure in the UK: Historical Cohort Study. Pragmat. Obs. Res. 2020, 11, 55–66. [Google Scholar] [CrossRef]
  150. Ansari, M.; Alexander, M.; Tutar, A.; Bello, D.; Massie, B.M. Cardiology participation improves outcomes in patients with new-onset heart failure in the outpatient setting. J. Am. Coll. Cardiol. 2003, 41, 62–68. [Google Scholar] [CrossRef] [PubMed]
  151. Auerbach, A.D.; Hamel, M.B.; Davis, R.B.; Connors, A.F.; Regueiro, C.; Desbiens, N.; Goldman, L.; Califf, R.M.; Dawson, N.V.; Wenger, N.; et al. Resource use and survival of patients hospitalized with congestive heart failure: Differences in care by specialty of the attending physician. SUPPORT Investigators. Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments. Ann. Intern. Med. 2000, 132, 191–200. [Google Scholar] [CrossRef] [PubMed]
  152. Philbin, E.F.; Jenkins, P.L. Differences between patients with heart failure treated by cardiologists, internists, family physicians, and other physicians: Analysis of a large, statewide database. Am. Heart J. 2000, 139, 491–496. [Google Scholar] [CrossRef] [PubMed]
  153. Jong, P.; Gong, Y.; Liu, P.P.; Austin, P.C.; Lee, D.S.; Tu, J.V. Care and outcomes of patients newly hospitalized for heart failure in the community treated by cardiologists compared with other specialists. Circulation 2003, 108, 184–191. [Google Scholar] [CrossRef] [PubMed]
  154. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.F.; Coats, A.J.S.; Falk, V.; González-Juanatey, J.R.; Harjola, V.-P.; Jankowska, E.A.; 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. [Google Scholar] [CrossRef] [PubMed]
  155. Engström, G. The restrictive-obstructive continuum and the failing heart. Thorax 2016, 71, 487–488. [Google Scholar] [CrossRef]
  156. Hawkins, N.M. Chronic obstructive pulmonary disease and heart failure in Europe-further evidence of the need for integrated care. Eur. J. Heart Fail. 2018, 20, 111–113. [Google Scholar] [CrossRef]
  157. Magnussen, H.; Canepa, M.; Zambito, P.E.; Brusasco, V.; Meinertz, T.; Rosenkranz, S. What can we learn from pulmonary function testing in heart failure? Eur. J. Heart Fail. 2017, 19, 1222–1229. [Google Scholar] [CrossRef]
  158. Pellegrino, R.; Viegi, G.; Brusasco, V.; Crapo, R.O.; Burgos, F.; Casaburi, R.; Coates, A.; van der Grinten, C.P.M.; Gustafsson, P.; Hankinson, J.; et al. Interpretative strategies for lung function tests. Eur. Respir. J. 2005, 26, 948–968. [Google Scholar] [CrossRef] [PubMed]
  159. Celli, B.R.; MacNee, W.; Force, E.R.T. Standards for the diagnosis and treatment of patients with COPD: A summary of the ATS/ERS position paper. Eur. Respir. J. 2004, 23, 932–946. [Google Scholar] [CrossRef] [PubMed]
  160. Mannino, D.M.; Buist, A.S.; Vollmer, W.M. Chronic obstructive pulmonary disease in the older adult: What defines abnormal lung function? Thorax 2007, 62, 237–241. [Google Scholar] [CrossRef] [PubMed]
  161. Barrecheguren, M.; Pinto, L.; Mostafavi-Pour-Manshadi, S.-M.-Y.; Tan, W.C.; Li, P.Z.; Aaron, S.D.; Benedetti, A.; Chapman, K.R.; Walker, B.; Fitzgerald, J.M.; et al. Identification and definition of asthma-COPD overlap: The CanCOLD study. Respirology 2020, 25, 836–849. [Google Scholar] [CrossRef] [PubMed]
  162. Reddel, H.K.; Bateman, E.D.; Becker, A.; Boulet, L.-P.; Cruz, A.A.; Drazen, J.M.; Haahtela, T.; Hurd, S.S.; Inoue, H.; de Jongste, J.C.; et al. A summary of the new GINA strategy: A roadmap to asthma control. Eur. Respir. J. 2015, 46, 622–639. [Google Scholar] [CrossRef] [PubMed]
  163. Gibson, P.G.; Simpson, J.L. The overlap syndrome of asthma and COPD: What are its features and how important is it? Thorax 2009, 64, 728–735. [Google Scholar] [CrossRef] [PubMed]
  164. Leung, J.M.; Sin, D.D. Asthma-COPD overlap syndrome: Pathogenesis; clinical features; therapeutic targets. BMJ 2017, 358, j3772. [Google Scholar] [CrossRef]
  165. Carolan, B.J.; Sutherland, E.R. Clinical phenotypes of chronic obstructive pulmonary disease and asthma: Recent advances. J. Allergy Clin. Immunol. 2013, 131, 627–634; quiz 635. [Google Scholar] [CrossRef]
  166. Sharma, G.; Kuo, Y.-F.; Freeman, J.L.; Zhang, D.D.; Goodwin, J.S. Outpatient follow-up visit and 30-day emergency department visit and readmission in patients hospitalized for chronic obstructive pulmonary disease. Arch. Intern. Med. 2010, 170, 1664–1670. [Google Scholar] [CrossRef]
  167. Sin, D.D.; Bell, N.R.; Svenson, L.W.; Man, S.F.P. The impact of follow-up physician visits on emergency readmissions for patients with asthma and chronic obstructive pulmonary disease: A population-based study. Am. J. Med. 2002, 112, 120–125. [Google Scholar] [CrossRef]
  168. Atwood, C.E.; Bhutani, M.; Ospina, M.B.; Rowe, B.H.; Leigh, R.; Deuchar, L.; Faris, P.; Michas, M.; Mrklas, K.J.; Graham, J.; et al. Optimizing COPD Acute Care Patient Outcomes Using a Standardized Transition Bundle and Care Coordinator: A Randomized Clinical Trial. Chest 2022, 162, 321–330. [Google Scholar] [CrossRef]
  169. Bourbeau, J.; Echevarria, C. Models of care across the continuum of exacerbations for patients with chronic obstructive pulmonary disease. Chronic Respir. Dis. 2020, 17, 1479973119895457. [Google Scholar] [CrossRef]
  170. Lenferink, A.; Brusse-Keizer, M.; van der Valk, P.D.; Frith, P.A.; Zwerink, M.; Monninkhof, E.M.; van der Palen, J.; Effing, T.W. Self-management interventions including action plans for exacerbations versus usual care in patients with chronic obstructive pulmonary disease. Cochrane Database Syst. Rev. 2017, 8, CD011682. [Google Scholar] [CrossRef] [PubMed]
  171. Benzo, R.; Vickers, K.; Novotny, P.J.; Tucker, S.; Hoult, J.; Neuenfeldt, P.; Connett, J.; Lorig, K.; McEvoy, C. Health Coaching and Chronic Obstructive Pulmonary Disease Rehospitalization. A Randomized Study. Am. J. Respir. Crit. Care Med. 2016, 194, 672–680. [Google Scholar] [CrossRef] [PubMed]
  172. Jencks, S.F.; Williams, M.V.; Coleman, E.A. Rehospitalizations among patients in the Medicare fee-for-service program. N. Engl. J. Med. 2009, 360, 1418–1428. [Google Scholar] [CrossRef] [PubMed]
  173. Gavish, R.; Levy, A.; Dekel, O.K.; Karp, E.; Maimon, N. The Association Between Hospital Readmission and Pulmonologist Follow-up Visits in Patients With COPD. Chest 2015, 148, 375–381. [Google Scholar] [CrossRef] [PubMed]
  174. Shah, T.; Churpek, M.M.; Perraillon, M.C.; Konetzka, R.T. Understanding why patients with COPD get readmitted: A large national study to delineate the Medicare population for the readmissions penalty expansion. Chest 2015, 147, 1219–1226. [Google Scholar] [CrossRef] [PubMed]
  175. Rice, K.L.; Dewan, N.; Bloomfield, H.E.; Grill, J.; Schult, T.M.; Nelson, D.B.; Kumari, S.; Thomas, M.; Geist, L.J.; Beaner, C.; et al. Disease management program for chronic obstructive pulmonary disease: A randomized controlled trial. Am. J. Respir. Crit. Care Med. 2010, 182, 890–896. [Google Scholar] [CrossRef] [PubMed]
  176. Rea, H.; McAuley, S.; Stewart, A.; Lamont, C.; Roseman, P.; Didsbury, P. A chronic disease management programme can reduce days in hospital for patients with chronic obstructive pulmonary disease. Intern. Med. J. 2004, 34, 608–614. [Google Scholar] [CrossRef] [PubMed]
  177. Bourbeau, J.; Julien, M.; Maltais, F.; Rouleau, M.; Beaupré, A.; Bégin, R.; Renzi, P.; Nault, D.; Borycki, E.; Schwartzman, K.; et al. Chronic Obstructive Pulmonary Disease axis of the Respiratory Network Fonds de la Recherche en Santé du Québec, Reduction of hospital utilization in patients with chronic obstructive pulmonary disease: A disease-specific self-management intervention. Arch. Intern. Med. 2003, 163, 585–591. [Google Scholar] [CrossRef] [PubMed]
  178. Zwerink, M.; Brusse-Keizer, M.; van der Valk, P.D.L.P.M.; Zielhuis, G.A.; Monninkhof, E.M.; van der Palen, J.; Frith, P.A.; Effing, T. Self management for patients with chronic obstructive pulmonary disease. Cochrane Database Syst. Rev. 2014, 3, CD002990. [Google Scholar] [CrossRef]
  179. Kessler, R.; Casan-Clara, P.; Koehler, D.; Tognella, S.; Viejo, J.L.; Negro, R.W.D.; Díaz-Lobato, S.; Reissig, K.; González-Moro, J.M.R.; Devouassoux, G.; et al. COMET: A multicomponent home-based disease-management programme versus routine care in severe COPD. Eur. Respir. J. 2018, 51, 1701612. [Google Scholar] [CrossRef] [PubMed]
  180. Bhatt, S.P.; Patel, S.B.; Anderson, E.M.; Baugh, D.; Givens, T.; Schumann, C.; Sanders, J.G.; Windham, S.T.; Cutter, G.R.; Dransfield, M.T. Video Telehealth Pulmonary Rehabilitation Intervention in Chronic Obstructive Pulmonary Disease Reduces 30-Day Readmissions. Am. J. Respir. Crit. Care Med. 2019, 200, 511–513. [Google Scholar] [CrossRef] [PubMed]
  181. Marcos, P.J.; Represas, C.R.; Ramos, C.; Álvarez, B.C.; Villar, A.F.; Liste, A.F.; Nocelo, S.F.; del Río, J.Q.; Sanz, C.Z.; Golpe, R.; et al. Impact of a Home Telehealth Program After a Hospitalized COPD Exacerbation: A Propensity Score Analysis. Arch. Bronconeumol. 2022, 58, 474–481. [Google Scholar] [CrossRef]
  182. Park, Y.; Kim, J.; Kim, S.; Moon, D.; Jo, H. Effects of Transitional Care after Hospital Discharge in Patients with Chronic Obstructive Pulmonary Disease: An Updated Systematic Review and Meta-Analysis. Int. J. Environ. Res. Public Health 2023, 20, 6053. [Google Scholar] [CrossRef]
  183. Hartman, M.; Mináriková, J.; Batalik, L.; Pepera, G.; Su, J.J.; Formiga, M.F.; Cahalin, L.; Dosbaba, F. Effects of Home-Based Training with Internet Telehealth Guidance in COPD Patients Entering Pulmonary Rehabilitation: A Systematic Review. Int. J. Chronic Obstr. Pulm. Dis. 2023, 18, 2305–2319. [Google Scholar] [CrossRef]
  184. Sculley, J.A.; Musick, H.; Krishnan, J.A. Telehealth in chronic obstructive pulmonary disease: Before, during, and after the coronavirus disease 2019 pandemic. Curr. Opin. Pulm. Med. 2022, 28, 93–98. [Google Scholar] [CrossRef]
  185. Barbosa, M.T.; Sousa, C.S.; Morais-Almeida, M.; Simões, M.J.; Mendes, P. Telemedicine in COPD: An Overview by Topics. COPD 2020, 17, 601–617. [Google Scholar] [CrossRef]
  186. Janjua, S.; Carter, D.; Threapleton, C.J.; Prigmore, S.; Disler, R.T. Telehealth interventions: Remote monitoring and consultations for people with chronic obstructive pulmonary disease (COPD). Cochrane Database Syst. Rev. 2021, 7, CD013196. [Google Scholar] [CrossRef]
  187. Casas, A.; Troosters, T.; Garcia-Aymerich, J.; Roca, J.; Hernández, C.; Alonso, A.; del Pozo, F.; de Toledo, P.; Antó, J.M.; Rodríguez-Roisín, R.; et al. Integrated care prevents hospitalisations for exacerbations in COPD patients. Eur. Respir. J. 2006, 28, 123–130. [Google Scholar] [CrossRef]
  188. Annandale, J.; Hurlin, C.; Lewis, K. Reducing COPD admissions with a specialist chronic disease management team. Nurs. Times 2009, 105, 25. [Google Scholar] [PubMed]
  189. Vestbo, J.; Anderson, J.A.; Calverley, P.M.A.; Celli, B.; Ferguson, G.T.; Jenkins, C.; Knobil, K.; Willits, L.R.; Yates, J.C.; Jones, P.W. Adherence to inhaled therapy, mortality and hospital admission in COPD. Thorax 2009, 64, 939–943. [Google Scholar] [CrossRef] [PubMed]
  190. Ashton, C.M.; Kuykendall, D.H.; Johnson, M.L.; Wray, N.P.; Wu, L. The association between the quality of inpatient care and early readmission. Ann. Intern. Med. 1995, 122, 415–421. [Google Scholar] [CrossRef] [PubMed]
  191. Wilkinson, T.M.A.; Donaldson, G.C.; Hurst, J.R.; Seemungal, T.A.R.; Wedzicha, J.A. Early therapy improves outcomes of exacerbations of chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2004, 169, 1298–1303. [Google Scholar] [CrossRef]
  192. Oancea, C.; Fira-Mladinescu, O.; Timar, B.; Tudorache, V. Impact of medical education program on COPD patients: A cohort prospective study. Wien. Klin. Wochenschr. 2015, 127, 388–393. [Google Scholar] [CrossRef] [PubMed]
  193. Wedzicha, J.A.; Miravitlles, M.; Hurst, J.R.; Calverley, P.M.A.; Albert, R.K.; Anzueto, A.; Criner, G.J.; Papi, A.; Rabe, K.F.; Rigau, D.; et al. Management of COPD exacerbations: A European Respiratory Society/American Thoracic Society guideline. Eur. Respir. J. 2017, 49, 1600791. [Google Scholar] [CrossRef] [PubMed]
  194. Sandhu, S.K.; Chu, J.; Yurkovich, M.; Harriman, D.; Taraboanta, C.; Fitzgerald, J.M. Variations in the management of acute exacerbations of chronic obstructive pulmonary disease. Can. Respir. J. 2013, 20, 175–179. [Google Scholar] [CrossRef]
  195. Boulet, L.-P.; Bourbeau, J.; Skomro, R.; Gupta, S. Major care gaps in asthma, sleep and chronic obstructive pulmonary disease: A road map for knowledge translation. Can. Respir. J. 2013, 20, 265–269. [Google Scholar] [CrossRef]
  196. Kripalani, S.; Jackson, A.T.; Schnipper, J.L.; Coleman, E.A. Promoting effective transitions of care at hospital discharge: A review of key issues for hospitalists. J. Hosp. Med. 2007, 2, 314–323. [Google Scholar] [CrossRef]
  197. van Walraven, C.; Oake, N.; Jennings, A.; Forster, A.J. The association between continuity of care and outcomes: A systematic and critical review. J. Eval. Clin. Pract. 2010, 16, 947–956. [Google Scholar] [CrossRef]
  198. Langsetmo, L.; Platt, R.W.; Ernst, P.; Bourbeau, J. Underreporting exacerbation of chronic obstructive pulmonary disease in a longitudinal cohort. Am. J. Respir. Crit. Care Med. 2008, 177, 396–401. [Google Scholar] [CrossRef]
  199. Pullen, R.; Miravitlles, M.; Sharma, A.; Singh, D.; Martinez, F.; Hurst, J.R.; Alves, L.; Dransfield, M.; Chen, R.; Muro, S.; et al. CONQUEST Quality Standards: For the Collaboration on Quality Improvement Initiative for Achieving Excellence in Standards of COPD Care. Int. J. Chronic Obstr. Pulm. Dis. 2021, 16, 2301–2322. [Google Scholar] [CrossRef] [PubMed]
  200. Fidahussein, S.S.; Croghan, I.T.; Cha, S.S.; Klocke, D.L. Posthospital follow-up visits and 30-day readmission rates in chronic obstructive pulmonary disease. Risk Manag. Healthc. Policy 2014, 7, 105–112. [Google Scholar] [CrossRef] [PubMed]
  201. Rabe, K.F.; Martinez, F.J.; Ferguson, G.T.; Wang, C.; Singh, D.; Wedzicha, J.A.; Trivedi, R.; Rose, E.S.; Ballal, S.; McLaren, J.; et al. Triple Inhaled Therapy at Two Glucocorticoid Doses in Moderate-to-Very-Severe COPD. N. Engl. J. Med. 2020, 383, 35–48. [Google Scholar] [CrossRef] [PubMed]
  202. Lipson, D.A.; Barnhart, F.; Brealey, N.; Brooks, J.; Criner, G.J.; Day, N.C.; Dransfield, M.T.; Halpin, D.M.G.; Han, M.K.; Jones, C.E.; et al. Once-Daily Single-Inhaler Triple versus Dual Therapy in Patients with COPD. N. Engl. J. Med. 2018, 378, 1671–1680. [Google Scholar] [CrossRef]
  203. Lipworth, B.; Skinner, D.; Devereux, G.; Thomas, V.; Jie, J.L.Z.; Martin, J.; Carter, V.; Price, D.B. Underuse of β-blockers in heart failure and chronic obstructive pulmonary disease. Heart 2016, 102, 1909–1914. [Google Scholar] [CrossRef]
  204. Du, Q.; Sun, Y.; Ding, N.; Lu, L.; Chen, Y. Beta-blockers reduced the risk of mortality and exacerbation in patients with COPD: A meta-analysis of observational studies. PLoS ONE 2014, 9, e113048. [Google Scholar] [CrossRef]
  205. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L.; et al. American College of Cardiology Foundation, American Heart Association Task Force on Practice Guidelines, 2013 ACCF/AHA guideline for the management of heart failure: A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef]
  206. Salpeter, S.; Ormiston, T.; Salpeter, E. Cardioselective beta-blockers for chronic obstructive pulmonary disease. Cochrane Database Syst. Rev. 2005, 4, CD003566. [Google Scholar] [CrossRef]
  207. Maggioni, A.P.; Anker, S.D.; Dahlström, U.; Filippatos, G.; Ponikowski, P.; Zannad, F.; Amir, O.; Chioncel, O.; Leiro, M.C.; Drozdz, J.; et al. Heart Failure Association of the ESC, Are hospitalized or ambulatory patients with heart failure treated in accordance with European Society of Cardiology guidelines? Evidence from 12,440 patients of the ESC Heart Failure Long-Term Registry. Eur. J. Heart Fail. 2013, 15, 1173–1184. [Google Scholar] [CrossRef]
  208. Dewan, P.; Docherty, K.F.; Bengtsson, O.; de Boer, R.A.; Desai, A.S.; Drozdz, J.; Hawkins, N.M.; Inzucchi, S.E.; Kitakaze, M.; Køber, L.; et al. Effects of dapagliflozin in heart failure with reduced ejection fraction and chronic obstructive pulmonary disease: An analysis of DAPA-HF. Eur. J. Heart Fail. 2021, 23, 632–643. [Google Scholar] [CrossRef] [PubMed]
  209. Canepa, M.; Ameri, P.; Lainscak, M. Chronic obstructive pulmonary disease and comorbidities in heart failure: The next frontier of sodium-glucose co-transporter 2 inhibitors? Eur. J. Heart Fail. 2021, 23, 644–647. [Google Scholar] [CrossRef] [PubMed]
  210. Bayes-Genis, A.; Pascual-Figal, D.; Fabregat, J.; Domingo, M.; Planas, F.; Casas, T.; Ordoñez-Llanos, J.; Valdes, M.; Cinca, J. Serial NT-proBNP monitoring and outcomes in outpatients with decompensation of heart failure. Int. J. Cardiol. 2007, 120, 338–343. [Google Scholar] [CrossRef] [PubMed]
  211. Pascual-Figal, D.A.; Domingo, M.; Casas, T.; Gich, I.; Ordoñez-Llanos, J.; Martínez, P.; Cinca, J.; Valdés, M.; Januzzi, J.L.; Bayes-Genis, A. Usefulness of clinical and NT-proBNP monitoring for prognostic guidance in destabilized heart failure outpatients. Eur. Heart J. 2008, 29, 1011–1018. [Google Scholar] [CrossRef] [PubMed]
  212. Dai, Z.; Asano, T.; Takahashi, O.; Komiyama, N.; Ohde, S. The minimal informative monitoring interval of N-terminal pro-B-type natriuretic peptide in patients with stable heart failure. BMC Cardiovasc. Disord. 2020, 20, 262. [Google Scholar] [CrossRef]
  213. Suissa, S.; Dell’Aniello, S.; Ernst, P. Long-term natural history of chronic obstructive pulmonary disease: Severe exacerbations and mortality. Thorax 2012, 67, 957–963. [Google Scholar] [CrossRef]
  214. Whittaker, H.; Rubino, A.; Müllerová, H.; Morris, T.; Varghese, P.; Xu, Y.; De Nigris, E.; Quint, J.K. Frequency and Severity of Exacerbations of COPD Associated with Future Risk of Exacerbations and Mortality: A UK Routine Health Care Data Study. Int. J. Chronic Obstr. Pulm. Dis. 2022, 17, 427–437. [Google Scholar] [CrossRef]
  215. Rothnie, K.J.; Müllerová, H.; Smeeth, L.; Quint, J.K. Natural History of Chronic Obstructive Pulmonary Disease Exacerbations in a General Practice-based Population with Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2018, 198, 464–471. [Google Scholar] [CrossRef]
  216. Sethi, S.; Make, B.J.; Robinson, S.B.; Kumar, S.; Pollack, M.; Moretz, C.; Dreyfus, J.; Xi, A.; Powell, D.; Feigler, N. Relationship of COPD Exacerbation Severity and Frequency on Risks for Future Events and Economic Burden in the Medicare Fee-For-Service Population. Int. J. Chronic Obstr. Pulm. Dis. 2022, 17, 593–608. [Google Scholar] [CrossRef] [PubMed]
  217. Pollack, M.; Tkacz, J.; Schinkel, J. Exacerbations and real-world outcomes (EROS) among patients with COPD receiving single inhaler triple therapy of budesonide/glycopyrrolate/formoterol fumarate [Poster Discussion]. In Proceedings of the American Thoracic Society International Conference, Washington, DC, USA, 19–24 May 2023. [Google Scholar]
  218. Donaldson, G.C.; Hurst, J.R.; Smith, C.J.; Hubbard, R.B.; Wedzicha, J.A. Increased risk of myocardial infarction and stroke following exacerbation of COPD. Chest 2010, 137, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  219. Daniels, K.; Tave, A.; Neikirk, A. Incidence of acute cardiovascular events following acute exacerbation of chronic obstructive pulmonary disease in a large US claims database [Thematic Poster Session]. In Proceedings of the American Thoracic Society International Conference, Washington, DC, USA, 19–24 May 2023. [Google Scholar]
  220. Dransfield, M.; Halpin, D.; Han, M.L.; Hartley, B.; Jones, C.E.; Kalhan, R.; Kilbride, S.; Kunisaki, K.; Lange, P.; Lipson, D.; et al. Time-Dependent Risk Of Cardiovascular Events Following An Exacerbation In Patients With Copd: Post Hoc Analysis From The Impact Triall. Chest 2020, 158, A1722–A1726. [Google Scholar] [CrossRef]
Figure 1. Risk factors shared by COPD and HF. AECOPD: acute exacerbation of COPD; COPD: chronic obstructive pulmonary disease; HF: heart failure.
Figure 1. Risk factors shared by COPD and HF. AECOPD: acute exacerbation of COPD; COPD: chronic obstructive pulmonary disease; HF: heart failure.
Jcm 13 01621 g001
Figure 2. Flowchart summarizing the path for differential diagnosis of COPD and HF. On the left—pink: acute care setting; blue: hospital setting; green: post discharge. On the right—orange: type of patient. AECOPD: acute exacerbation of COPD; COPD: chronic obstructive pulmonary disease; HF: heart failure; pMDI: pressurized metered dose inhaler.
Figure 2. Flowchart summarizing the path for differential diagnosis of COPD and HF. On the left—pink: acute care setting; blue: hospital setting; green: post discharge. On the right—orange: type of patient. AECOPD: acute exacerbation of COPD; COPD: chronic obstructive pulmonary disease; HF: heart failure; pMDI: pressurized metered dose inhaler.
Jcm 13 01621 g002
Figure 3. Checklist of the main lab-test, signs and imaging that should lead to suspicion of HF as a cause of or a contributor to COPD.
Figure 3. Checklist of the main lab-test, signs and imaging that should lead to suspicion of HF as a cause of or a contributor to COPD.
Jcm 13 01621 g003
Table 1. Clinical evaluation recommended in the acute care setting for a patient with AECOPD and/or HF.
Table 1. Clinical evaluation recommended in the acute care setting for a patient with AECOPD and/or HF.
Laboratory TestsImaging
Blood cell count
Plasma electrolytes
Chest radiography
ESRLung sonography with assessment of diaphragmatic excursion and pleural effusion
CRPChest computed tomography scan
D-dimerDiagnostic tests
Electrocardiography
hs-cardiac troponinPulse oximetry
GlycemiaBlood gas analysis
Eosinophilic and neutrophilic pattern
NT-proBNP (recommended both as a diagnostic/prognostic test for heart overload or HF or to exclude them)
ESR: erythrocyte sedimentation rate; CRP: C-reactive protein; hs-troponin: high-sensitivity troponin; NT-proBNP: N-terminal prohormone of brain natriuretic peptide; HF: heart failure.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bianco, A.; Canepa, M.; Catapano, G.A.; Marvisi, M.; Oliva, F.; Passantino, A.; Sarzani, R.; Tarsia, P.; Versace, A.G. Implementation of the Care Bundle for the Management of Chronic Obstructive Pulmonary Disease with/without Heart Failure. J. Clin. Med. 2024, 13, 1621. https://doi.org/10.3390/jcm13061621

AMA Style

Bianco A, Canepa M, Catapano GA, Marvisi M, Oliva F, Passantino A, Sarzani R, Tarsia P, Versace AG. Implementation of the Care Bundle for the Management of Chronic Obstructive Pulmonary Disease with/without Heart Failure. Journal of Clinical Medicine. 2024; 13(6):1621. https://doi.org/10.3390/jcm13061621

Chicago/Turabian Style

Bianco, Andrea, Marco Canepa, Giosuè Angelo Catapano, Maurizio Marvisi, Fabrizio Oliva, Andrea Passantino, Riccardo Sarzani, Paolo Tarsia, and Antonio Giovanni Versace. 2024. "Implementation of the Care Bundle for the Management of Chronic Obstructive Pulmonary Disease with/without Heart Failure" Journal of Clinical Medicine 13, no. 6: 1621. https://doi.org/10.3390/jcm13061621

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop