• No results found

University of Groningen Effectiveness and safety of medicines used in COPD patients Wang, Yuanyuan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Effectiveness and safety of medicines used in COPD patients Wang, Yuanyuan"

Copied!
212
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Effectiveness and safety of medicines used in COPD patients Wang, Yuanyuan

DOI:

10.33612/diss.123921981

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Wang, Y. (2020). Effectiveness and safety of medicines used in COPD patients: pharmacoepidemiological studies. University of Groningen. https://doi.org/10.33612/diss.123921981

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Effectiveness and safety of

medicines used in COPD patients

Pharmacoepidemiological studies

(3)

Effectiveness and safety of medicines used in COPD patients Pharmacoepidemiological studies

ISBN: 978-94-034-2555-9 (printed version) ISBN: 978-94-034-2554-2 (electric version)

Author: Yuanyuan Wang

Cover-design: IRINA SHI (photo) & Off Page (content) Printing: Off Page (www.offpage.nl)

The studies presented in this thesis were funded by University of Groningen and the China Scholarship Council (CSC) Scholarship. This thesis was conducted within the Groningen University Institute for Drug Exploration (GUIDE). Printing of this thesis was financially supported by the University of Groningen and the Graduate School of Science and Engineering (GSSE).

Copyright ©Yuanyuan Wang, 2020, Groningen, The Netherlands.

All rights reserved. No part of this book may be reproduced or transmitted in any form by any means, electronically or mechanically by photocopying, recording, or otherwise, without the written permission of the author. The copy right of previously published chapters of this thesis remains with the publisher or journal.

(4)

Effectiveness and safety of

medicines used in COPD patients

Pharmacoepidemiological studies

PhD thesis

to obtain the degree of PhD at the University of Groningen

on the authority of the

Rector Magnificus Prof. C. Wijmenga and in accordance with

the decision by the College of Deans. This thesis will be defended in public on

Friday 8 May 2020 at 14.30 hours

by

Yuanyuan Wang

born on 15 February 1988 in Henan, China

(5)

Prof. H.M. oe en

Assessment Committee

Prof. T. .M. erheij

Prof. . van der Palen Prof. . tienstra

(6)

TABLE OF CONTENTS

Chapter 1 General Introduction 7

Part I Effects of antibiotic use for COPD exacerbations and

potential DDIs during COPD exacerbation management

Chapter 2 Effects of Prophylactic Antibiotics on Patients with Stable COPD: 21

A Systematic Review and Meta-Analysis of Randomized Controlled Trials

Chapter 3 The influence of age on real-life effects of doxycycline for 43

acute exacerbations among COPD outpatients: a population-based cohort study

Chapter 4 Real-world short- and long-term effects of antibiotic therapy on 61

acute exacerbations of COPD in outpatients: a cohort study under the PharmLines Initiative

Chapter 5 Improving antibacterial prescribing safety in 81

the management of COPD exacerbations:

systematic review of observational and clinical studies on

potential drug interactions associated with frequently prescribed antibacterials among COPD Patients

Part II Neuropsychiatric safety of varenicline use for smoking cessation

and the application of prescription sequence symmetry analysis in drug safety evaluation

Chapter 6 Neuropsychiatric safety of varenicline in the general and 119

COPD population with and without psychiatric disorders: a retrospective inception cohort study in a real-world setting

Chapter 7 Risk of neuropsychiatric adverse events associated with 145

varenicline treatment for smoking cessation: a prescription sequence symmetry analysis

Chapter 8 Effect estimate comparison between the prescription 165

sequence symmetry analysis and parallel group study designs: a systematic review

Chapter 9 General discussion 183

Chapter 10 Summary 197

Samenvatting 201

Acknowledgement 204

List of publications 208

About the author 211

Supervisors

Prof. . Ha Prof. H.M. oe en

Assessment Committee

Prof. T. .M. erheij

Prof. . van der Palen Prof. . tienstra

(7)
(8)
(9)
(10)

General Introduction

1

GENERAL INTRODUCTION

Chronic obstructive pulmonary disease

According to the Global Initiative for Chronic Obstructive Lung Disease (GOLD) report 2020, Chronic Obstructive Pulmonary Disease (COPD) is a common, preventable and treatable disease that is characterized by persistent respiratory symptoms and airflow limitation. Such symptoms are caused by airway and/or alveolar abnormalities usually triggered by significant exposure to noxious particles or gases.1 Cigarette smoking is

the main independent causal risk factor for COPD with indoor and outdoor air pollution and occupational exposure to dust and noxious particles also being the risk factors for COPD.2 Moreover, host factors that may contribute to the development of COPD include

age, genetics, airway hyper responsiveness and abnormal lung development.3-5

The prevalence of COPD varies across countries as well as regions within countries. According to the findings of a global meta-analysis, the number of COPD cases increased to 384 million in 2010, with a global prevalence of 12% (ranging between 8% and 15%).6 COPD is commonly diagnosed in individuals aged 40 years or older

based on the presence of associated symptoms and risk factors. However, a definitive COPD diagnosis requires the performance of spirometry. The presence of a post-bronchodilator FEV1/FVC < 0.7 confirms the presence of airflow limitation and results in a COPD diagnosis. COPD patients are currently categorized into four GOLD stages of severity of airflow limitations based on the predicted value of FEV1: mild (stage I, FEV1 ≥ 80% predicted), moderate (stage II, 50% ≤ FEV1 < 80% predicted), severe (stage III, 30% ≤ FEV1 < 80% predicted) and very severe (stage IV, FEV1 < 30% predicted).1 A large

prevalence study estimated that the rate of COPD for GOLD stage II and higher is around 10% in the general population, and a little higher in men than women (11.8% and 8.5%, respectively).7

COPD is one of the leading causes of morbidity and mortality worldwide.8 Its burden is

predicted to increase in the coming decades as a result of continuous exposure to risk factors in developing countries and aging of the population worldwide, particularly in high-income countries.9 Smoking cessation interventions, increased physical activity,

and early diagnosis and treatment of related comorbidities are considered key measures for reducing the health-economic burden of COPD. 10

Up to now, the key goals of COPD treatment have been to improve patients’ prognosis and prevent the disease from worsening. The main treatment used in the daily management of mild and moderate COPD is pharmacotherapy. Bronchodilators, including short- and long-acting β2-agonists (SABA and LABA) and short- and long-acting anticholinergics (SAMA and LAMA) are essential for managing and preventing symptoms, and combined treatment (SABA/SAMA, LABA/LAMA or LABA/ICS) may be used as appropriate.

(11)

Non-pharmacological treatment comprises pulmonary rehabilitation (e.g., exercise training, education, and behavioral change).11 Oxygen therapy is necessary for patients with very

severe COPD and lung surgery may also be necessary.

Exacerbations of COPD and antibiotic use

An exacerbation of COPD is defined as an acute worsening of respiratory symptoms that necessitates additional therapy.1 COPD patients can periodically experience acute

exacerbations that may accelerate the decline in lung function, reduce the quality of life, and increase mortality and health-care costs.12,13 14 Infections, especially bacterial

infections, and inflammation are thought to be an important trigger for exacerbations of COPD. Previous studies have found that bacteria are responsible for around 40% to 50% of exacerbations.15,16 S. pneumoniae, H. influenza, P. aeruginosa, M. catarrhalis,

A. baumannii, and S. aureus were the most frequently reported bacteria that cause exacerbations of COPD.16-18 According to the GOLD guideline, the main goals in

treatment of COPD exacerbations are minimizing the negative impact of the current exacerbation and preventing subsequent exacerbations.1 Because almost 40% of

exacerbations are bacteria-caused respiratory tract infections,16 the use of antibiotics

has become a common component in the management of acute exacerbations among COPD patients, both in terms of treatment and prevention.1,19

Notably, recommendations in prophylactic use of antibiotics in the management of COPD exacerbations are conditional and unspecific. Only long-term macrolides are currently mentioned as first-line therapy.1,19 Moreover, in terms of current evidence, an optimal

regimen of prophylactic antibiotics for exacerbations has not been well established, and related recommendations regarding an appropriate schedule (continuous vs. intermittent) and the duration of a specific antibiotic intervention (below or equal to 6 months vs. above 6 months) are still lacking. Besides, the effects of even the most extensively researched antibiotics macrolides—let alone other potentially suitable antibiotics—on the time to the first exacerbation, changes in lung function, the bacterial load, and airway inflammation have not been adequately evaluated.20 Knowledge of

these outcomes is also vital for elucidating possible mechanisms behind the reduction of exacerbations through the prophylactic use of antibiotics, and for weighing benefits and risks.21

The beneficial effects of oral corticosteroids as an effective treatment for acute exacerbations of COPD (AECOPD) in improving COPD symptoms and lung function are well established.22 However, although antibiotics have been recommended for

the treatment of AECOPOD when signs of bacterial infection are present,1 there

is still uncertainty regarding the beneficial effects of antibiotic treatment used in the combination with oral glucocorticoids for AECOPD, particularly in the case of outpatients in real-world settings. In 2012, a Cochrane review that pooled the results

(12)

General Introduction

1

of five RCTs conducted among outpatients did not reveal a significantly reduced risk of treatment failure associated with antibiotics currently prescribed for outpatients.23

However, an updated version of this Cochrane review conducted in 2018 presented statistically significant beneficial effects of current antibiotics prescribed for outpatients.24 Two new RCTs were included in this later study in relation to the earlier

review conducted in 2012.25,26 Notably, one of the two RCTs did not support the beneficial

effects of antibiotics treatment on AECOPD, although it did contribute to almost 25% of the sample size of the updated pooled results.25

Hence, most of the available scientific evidence on the effects of antibiotics for AECOPD is basically derived from RCTs. It is widely accepted that RCTs provide solid evidence with high internal validity, however, their generalizability in the real world, especially in outpatient settings is low. COPD is a chronic disease and is mostly managed on an outpatient basis within a population that is more heterogeneous compared with populations from RCTs. Moreover, the use of antibiotics for AECOPD treatment is not always appropriate and in line with related guidelines.27,28 Therefore, the effect of

antibiotic treatment for AECOPD in real-world settings may differ from those obtained in clinical trials and require further investigation.

Comorbidities of COPD and potential drug-drug interactions

COPD is a chronic disease and its prevalence increases with age; around 15% of the general population over 65 years is affected by COPD.29 Hence, age-related comorbidities

frequently co-exist with COPD.30 The most common concomitant chronic conditions

associated with COPD include cardiovascular disease (e.g., heart failure, ischemic heart disease, and arrhythmias), metabolic disease (e.g., diabetes), osteoporosis, depression and anxiety, lung cancer and gastroesophageal reflux (GERD).1,30

COPD itself is a complex disease that entails the need for a variety of medications to improve lung function and treat exacerbations.1 Multiple comorbidities further

complicate the medical management of COPD, resulting in polypharmacy among a large section of COPD patients. Polypharmacy poses a potential risk of drug-drug interactions (DDIs) that may induce adverse events and treatment failures. Moreover, COPD is an age-related disease that generally manifests at an older age. Therefore, these older patients are more susceptible to DDIs due to gradual physiologic negative changes that may influence their pharmacokinetics and the pharmacodynamics of the drugs used.31

As most evidence about drug effects is from clinical trials, more attention should be paid to issues related to polypharmacy and to potential DDIs in the management of COPD in real-world settings.32 This is especially the case for antibiotic therapy as it

includes different drug classes that vary in their mechanisms relating to absorption and metabolism, making their interaction with other medications more likely. Comprehensive information for clinicians to avoid potential DDIs, however, is lacking.

(13)

Smoking cessation drug therapy and neuropsychiatric safety

Tobacco smoking is the main risk factor for COPD and other physical and mental disorders.33-35 This preventable behavior poses huge threats to global public health.36,37

Although in recent years, strict tobacco control policies have prompted a global decline in smoking,36 the actual numbers of smokers and smoking-related disease burden

continues to increase because of the growing population worldwide.38 More than eight

million people continue to die annually as a result of tobacco consumption.39

Therefore, smoking cessation strategies to prevent smoking-related diseases are imperative.40 Varenicline, which was the first non-nicotine, pharmacotherapeutic,

smoking cessation product, has been found to be more efficacious than other therapies, such as single-dose bupropion and nicotine replacement therapy (NRT).41

However, following varenicline’s approval by the FDA in 2006, safety concerns were raised relating to its neuropsychiatric adverse events, which include suicidal thoughts, aggressive behavior, depression, anxiety, and sleep disorders.42 Numerous RCTs were

subsequently conducted with varenicline to generate evidence on its safety.43 In light of

their findings, the FDA warning was removed in 2016. However, concerns remain, given the strict inclusion and exclusion criteria applied in RCTs that result in the participation of relatively healthy individuals and the lack of consistent real-world evidence. Notably, special risk populations demonstrating increased smoking prevalence, such as COPD patients, have generally been excluded from RCTs.44

As previously noted, most COPD patients are elderly and have multi-morbidities, making them more susceptible to adverse drug events (ADEs). Similarly, there is evidence that individuals with psychiatric disorders experience relapses of psychiatric symptoms more frequently than those without these disorders.45,46 The safety of varenicline use for these

specific populations has not been established. Although a few studies were conducted among patients with COPD or psychiatric disorders,47,48 the results were inconsistent.

Consequently, more observational studies are still needed to generate the real-world evidence relating to the safety of varenicline use.

PSSA and observational study designs in drug safety evaluation

Most evidence regarding the effects of drugs is derived from strictly regulated clinical trials. However, the results from RCTs may not reflect the real-world situations, given that the participants are relatively healthy and because of the limited scope for detecting rare events with clinical trials. Therefore, real-world evidence derived from traditional, non-randomized, observational study designs is valuable for exploring such drug effects or toxicities within the field of pharmacoepidemiology. However, the evidence from observational studies is often inconsistent, and such designs have been criticized for their potential of bias (e.g., selection or information bias) and confounding (e.g. unmeasured confounding).49

(14)

General Introduction

1

Prescription sequence symmetry analysis (PSSA) is increasingly being used to detect adverse effects or events associated with medications. PSSA is a self-controlled study design in which genetic and other time-invariant confounding can be well controlled, it does not entail the abovementioned bias.50,51 It compares the symmetry in the sequence

of exposure medication and marker (outcome) medications as proxy for ADRs within a specific time window based on prescriptions or claims databases.52 The sequence

ratio (SR) reflects the association between exposure and outcome. However, PSSA is still sensitive to time-varying variables, notably if the follow-up time is long. The overall validity of PSSA study designs has not been fully evaluated by comparing its result with those from conventional observational parallel group study designs, and such comparisons are urgently required.

AIM OF THIS THESIS

In this thesis, we aim to develop a comprehensive profile on the effectiveness of antibiotic use for acute exacerbations of COPD both prescribed prophylactically and therapeutically, and to provide real-world data on neuropsychiatric safety of varenicline use for smoking cessation, particularly among high risk populations with COPD or psychiatric diseases.

OUTLINE OF THIS THESIS

In part I of this thesis, we present several studies on the role of antibiotics in acute exacerbations of COPD (AECOPD).

In Chapter 2, we report the results from a meta-analysis of RCTs focusing on the beneficial effects and side effects of prophylactic antibiotic therapy in COPD patients.

In Chapter 3, we demonstrate the real-world effects of doxycycline treatment on acute exacerbations among COPD outpatients based on data extracted from the University of Groningen’s prescription database (IADB.nl) and explored the possible influence of age on the clinical outcomes.

In Chapter 4, we further explored the real-world effects of several antibiotic drugs used for acute exacerbations of COPD patients based on a linked database between the Lifelines Cohort biobank with extensive clinical information and the University of Groningen’s prescription database (IADB.nl).

In Chapter 5, we present a systematic review of drug-drug interactions associated with frequently prescribed antibiotics among COPD patients based on causal evidence obtained from observational cohort studies, case-control studies and clinical studies, aimed at improving the safety of antibacterial prescriptions.

(15)

In part II of this thesis, we present studies on the role of varenicline for smoking cessation using different designs.

In Chapter 6, we present the results of a retrospective inception cohort study aimed at assessing the risk of neuropsychiatric adverse events (NPAEs) in starters with varenicline versus starters with nicotine replacement therapy (NRT) among both the general and COPD populations, with and without psychiatric disorders. This study was conducted using data extracted from the University of Groningen’s prescription database (IADB.nl). In Chapter 7, we further examine the association between varenicline use and the onset of NPAEs in a real-world setting using a prescription sequence symmetry analysis (PSSA) study design.

Furthermore, in Chapter 8 we systematically compared effect estimates derived from the PSSA study with effect estimates from conventional observational parallel group study designs, to assess the validity and constraints of the PSSA study design within epidemiological research.

Last, in Chapter 9, we summarized the main findings of this thesis, discussed these findings in detail and provided suggestions for future research.

(16)

General Introduction

1

REFERENCES

1. Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global Strategy for the Diagnosis, Management and Prevention of Chronic Obstructive Pulmonary Disease: 2020 Report. https://goldcopd.org/gold-reports/. Date last accessed: December 17, 2019. 2. Postma DS, Bush A, van den Berge

M. Risk factors and early origins of chronic obstructive pulmonary disease. Lancet. 2015;385(9971):899-909.

3. Lange P, Celli B, Agusti A, et al. Lung-Function Trajectories Leading to Chronic Obstructive Pulmonary Disease. N Engl J Med. 2015;373(2):111-122.

4. Stern DA, Morgan WJ, Wright AL, Guerra S, Martinez FD. Poor airway function in early infancy and lung function by age 22 years: a non-selective longitudinal cohort study. Lancet. 2007;370(9589):758-764.

5. Tashkin DP, Altose MD, Bleecker ER, et al. The lung health study: airway responsiveness to inhaled methacholine in smokers with mild to moderate airflow limitation. The Lung Health Study Research Group. Am Rev Respir Dis. 1992;145(2 Pt 1):301-310.

6. Adeloye D, Chua S, Lee C, et al. Global and regional estimates of COPD prevalence: Systematic review and meta-analysis. J Glob Health. 2015;5(2):020415.

7. Buist AS, McBurnie MA, Vollmer WM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007;370(9589):741-750.

8. Lozano R, Naghavi M, Foreman K, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095-2128.

9. Lopez AD, Shibuya K, Rao C, et al. Chronic obstructive pulmonary disease: current burden and future projections. Eur Respir J. 2006;27(2):397-412.

10. Rabe KF, Watz H. Chronic obstructive pulmonary disease. Lancet. 2017;389(10082):1931-1940. 11. Spruit MA, Singh SJ, Garvey C, et al. An

official American Thoracic Society/European Respiratory Society statement: key concepts and advances in pulmonary rehabilitation. Am J Respir Crit Care Med. 2013;188(8):e13-64. 12. Wedzicha JA, Seemungal TAR. COPD

exacerbations: defining their cause and prevention. Lancet. 2007;370(9589):786-796. 13. Soler-Cataluna JJ, Martinez-Garcia MA,

Roman Sanchez P, Salcedo E, Navarro M, Ochando R. Severe acute exacerbations and mortality in patients with chronic obstructive pulmonary disease. Thorax. 2005;60(11):925-931.

14. O’Reilly JF, Williams AE, Rice L. Health status impairment and costs associated with COPD exacerbation managed in hospital. Int J Clin Pract. 2007;61(7):1112-1120.

15. Sethi S, Murphy TF. Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008;359(22):2355-2365.

16. Moghoofei M, Azimzadeh Jamalkandi S, Moein M, Salimian J, Ahmadi A. Bacterial infections in acute exacerbation of chronic obstructive pulmonary disease: a systematic review and meta-analysis. Infection. 2019. 17. Wilkinson TMA, Aris E, Bourne SC, et

al. Drivers of year-to-year variation in exacerbation frequency of COPD: analysis of the AERIS cohort. ERJ Open Res. 2019;5(1). 18. Monso E, Garcia-Aymerich J, Soler N, et al.

Bacterial infection in exacerbated COPD with changes in sputum characteristics. Epidemiol Infect. 2003;131(1):799-804. 19. Wedzicha JA, Calverley PMA, Albert RK, et al.

Prevention of COPD exacerbations: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2017;50(3). 20. Ni WT, Shao XD, Cai XJ, et al. Prophylactic Use

of Macrolide Antibiotics for the Prevention of Chronic Obstructive Pulmonary Disease

(17)

Exacerbation: A Meta-Analysis. Plos One. 2015;10(3).

21. Cameron EJ, McSharry C, Chaudhuri R, Farrow S, Thomson NC. Long-term macrolide treatment of chronic inflammatory airway diseases: risks, benefits and future developments. Clin Exp Allergy. 2012;42(9):1302-1312.

22. Walters JA, Tan DJ, White CJ, Gibson PG, Wood-Baker R, Walters EH. Systemic corticosteroids for acute exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2014(9):CD001288.

23. Vollenweider DJ, Jarrett H, Steurer-Stey CA, Garcia-Aymerich J, Puhan MA. Antibiotics for exacerbations of chronic obstructive pulmonary disease. Cochrane Db Syst Rev. 2012(12).

24. Vollenweider DJ, Frei A, Steurer-Stey CA, Garcia-Aymerich J, Puhan MA. Antibiotics for exacerbations of chronic obstructive pulmonary disease. Cochrane Db Syst Rev. 2018(10).

25. van Velzen P, Ter Riet G, Bresser P, et al. Doxycycline for outpatient-treated acute exacerbations of COPD: a randomised double-blind placebo-controlled trial. Lancet Respir Med. 2017;5(6):492-499. 26. Hassan WA, Shalan I, Elsobhy M. Impact of

antibiotics on acute exacerbations of COPD. Egypt J Chest Dis Tu. 2015;64(3):579-585. 27. Bathoorn E, Groenhof F, Hendrix R, et al.

Real-life data on antibiotic prescription and sputum culture diagnostics in acute exacerbations of COPD in primary care. Int J Chron Obstruct Pulmon Dis. 2017;12:285-290. 28. Roede BM, Bindels PJ, Brouwer HJ, Bresser

P, de Borgie CA, Prins JM. Antibiotics and steroids for exacerbations of COPD in primary care: compliance with Dutch guidelines. Br J Gen Pract. 2006;56(530):662-665.

29. Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, Mannino DM. Global burden of COPD: systematic review and meta-analysis. Eur Respir J. 2006;28(3):523-532.

30. Chetty U, McLean G, Morrison D, Agur K, Guthrie B, Mercer SW. Chronic obstructive pulmonary disease and comorbidities:

a large cross-sectional study in primary care. Br J Gen Pract. 2017;67(658):e321-e328. 31. Mangoni AA, Jackson SH. Age-related changes

in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6-14.

32. Hanlon P, Nicholl BI, Jani BD, et al. Examining patterns of multimorbidity, polypharmacy and risk of adverse drug reactions in chronic obstructive pulmonary disease: a cross-sectional UK Biobank study. BMJ Open. 2018;8(1):e018404.

33. Banks E, Joshy G, Korda RJ, et al. Tobacco smoking and risk of 36 cardiovascular disease subtypes: fatal and non-fatal outcomes in a large prospective Australian study. BMC Med. 2019;17(1):128.

34. Gaudet MM, Carter BD, Brinton LA, et al. Pooled analysis of active cigarette smoking and invasive breast cancer risk in 14 cohort studies. Int J Epidemiol. 2017;46(3):881-893. 35. Tjora T, Hetland J, Aaro LE, Wold B, Wiium

N, Overland S. The association between smoking and depression from adolescence to adulthood. Addiction. 2014;109(6):1022-1030. 36. Collaborators GBDT. Smoking prevalence and

attributable disease burden in 195 countries and territories, 1990-2015: a systematic analysis from the Global Burden of Disease Study 2015. Lancet. 2017;389(10082):1885-1906.

37. Thun MJ, Carter BD, Feskanich D, et al. 50-Year Trends in Smoking-Related Mortality in the United States. New Engl J Med. 2013;368(4):351-364.

38. The L. Progress towards a tobacco-free world. Lancet. 2018;392(10141):1.

39. World Health Organisation. WHO report on the global tobacco epidemic 2019: offer help to quit tobacco use. Geneva: World Health Organisation, 2019: 17-21.

40. Mehrotra R, Yadav A, Sinha DN, et al. Smokeless tobacco control in 180 countries across the globe: call to action for full implementation of WHO FCTC measures. Lancet Oncol. 2019;20(4):e208-e217.

41. Anthenelli RM, Benowitz NL, West R, et al. Neuropsychiatric safety and efficacy of

(18)

General Introduction

1

varenicline, bupropion, and nicotine patch

in smokers with and without psychiatric disorders (EAGLES): a double-blind, randomised, placebo-controlled clinical trial. Lancet. 2016;387(10037):2507-2520. 42. Moore TJ, Furberg CD, Glenmullen J,

Maltsberger JT, Singh S. Suicidal behavior and depression in smoking cessation treatments. Plos One. 2011;6(11):e27016. 43. Thomas KH, Martin RM, Knipe DW, Higgins

JP, Gunnell D. Risk of neuropsychiatric adverse events associated with varenicline: systematic review and meta-analysis. BMJ. 2015;350:h1109.

44. Lawrence D, Mitrou F, Zubrick SR. Smoking and mental illness: results from population surveys in Australia and the United States. Bmc Public Health. 2009;9.

45. Garza D, Murphy M, Tseng LJ, Riordan HJ, Chatterjee A. A double-blind randomized placebo-controlled pilot study of neuropsychiatric adverse events in abstinent smokers treated with varenicline or placebo. Biol Psychiatry. 2011;69(11):1075-1082. 46. Tonstad S, Davies S, Flammer M, Russ C, Hughes

J. Psychiatric adverse events in randomized, double-blind, placebo-controlled clinical trials of varenicline: a pooled analysis. Drug Saf. 2010;33(4):289-301.

47. Kotz D, Viechtbauer W, Simpson CR, van Schayck OCP, West R, Sheikh A. Cardiovascular and neuropsychiatric risks of varenicline and bupropion in smokers with chronic obstructive pulmonary disease. Thorax. 2017;72(10):905-911.

48. Evins AE, Benowitz NL, West R, et al. Neuropsychiatric Safety and Efficacy of Varenicline, Bupropion, and Nicotine Patch in Smokers With Psychotic, Anxiety, and Mood Disorders in the EAGLES Trial. J Clin Psychopharmacol. 2019;39(2):108-116. 49. Boyko EJ. Observational

research--opportunities and limitations. J Diabetes Complications. 2013;27(6):642-648.

50. Wahab IA, Pratt NL, Wiese MD, Kalisch LM, Roughead EE. The validity of sequence symmetry analysis (SSA) for adverse drug reaction signal detection. Pharmacoepidemiol Drug Saf. 2013;22(5):496-502.

51. Lai ECC, Pratt N, Hsieh CY, et al. Sequence symmetry analysis in pharmacovigilance and pharmacoepidemiologic studies. European Journal of Epidemiology. 2017;32(7):567-582. 52. Hallas J. Evidence of depression

provoked by cardiovascular medication: a prescription sequence symmetry analysis. Epidemiology. 1996;7(5):478-484.

(19)
(20)

Effects of antibiotic use for COPD

exacerbations and potential DDIs during COPD

exacerbation management

(21)
(22)

Yuanyuan Wang

Tanja R. Zijp

Muh. Akbar Bahar

Janwillem W.H. Kocks

Bob Wilffert

Eelko Hak

Effects of Prophylactic Antibiotics on Patients

with Stable COPD: A Systematic Review and

Meta-Analysis of Randomized Controlled Trials

Published as: Wang Y, Zijp TR, Bahar MA, Kocks JWH, Wilffert B, Hak E. Effects of prophylactic antibiotics on patients with stable COPD: a systematic review and meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2018;73(12):3231–3243.

(23)

ABSTRACT

Background

As bacterial infections provoke exacerbations, COPD patients may benefit from prophylactic antibiotics. However, evidence regarding their overall benefit-risk is conflicting.

Objectives

To update previous evidence and systematically evaluate the beneficial and side effects of prophylactic antibiotics on stable COPD patients.

Methods

Several databases were searched up to April 26, 2017 for randomized controlled trials (RCTs) on prophylactic antibiotics in stable COPD patients. The Primary outcomes were exacerbations and quality of life. Duration and schedule of antibiotics were considered in sub-group analyses.

Results

Twelve RCTs involving 3,683 patients were included. Prophylactic antibiotics significantly reduced the frequency of exacerbations (risk ratio [RR] 0.74, 95% CI 0.60-0.92) and the number of patients with one or more exacerbations (RR 0.82, 95% CI 0.74-0.90). Erythromycin and azithromycin appeared the most effective with the number needed to treat ranging from four to seven. Quality of life was also significantly improved by prophylactic antibiotics (mean difference -1.55, 95% CI -2.59 to -0.51). Time to first exacerbation was prolonged in six studies with one conflicting result. Neither the rate of hospitalization nor the rate of adverse events was significantly changed. Furthermore, no significant changes were observed in lung function, bacterial load and airway inflammation. However, antibiotic resistant isolates were significantly increased (OR 4.49, 95% CI 2.48-8.12).

Conclusions

Prophylactic antibiotics were effective in preventing COPD exacerbations and improving quality of life among stable patients with moderate to severe COPD. The choice of prophylactic antibiotics should be analysed and considered case by case, especially for long and continuous use.

(24)

Effects of prophylactic antibiotics on COPD

2

INTRODUCTION

COPD is an inflammatory disease that is characterized by persistent respiratory symptoms and airflow limitation.1 At present, COPD is one of the leading causes

of chronic morbidity and mortality worldwide, its burden is predicted to increase in the coming decades due to continuous exposure to risk factors and aging of population globally.2 In the course of COPD, exacerbation as an acute worsening of respiratory

symptoms has a profound negative impact on health oucomes.3 A vicious circle of

infection and inflammation is thought as a key to trigger exacerbations of COPD, about 40-50% of exacerbations are caused by bacteria.4

The use of prophylactic antibiotic has been suggested to prevent exacerbations in COPD patients for a long time. However, a Cochrane review in 2003 concluded that antibiotics only contribute to a small 9% reduction of exacerbations and should not be part of routine treatment considering the risk of antibiotic resistance and adverse effects.5 Ten

years later in 2013, the review by Herath et al. concluded a clinically significant benefit in reducing COPD exacerbations from continuous use of prophylactic antibiotics, but not from intermittent way due to only one randomized controlled trial (RCT) included in this subgroup.6 Influence of different duration of antibiotic intervention were not explored

in this study. The most recent review by Ni et al. in 2015 focused on macrolides only and did not evaluate meaningful outcomes including the time to first exacerbation, change of lung function, bacterial load and airway inflammation.7 The latter outcome is

important to support the hypothetical mechanism behind the reduction of exacerbations by antibiotics.8

Current recommendation from guidelines about prophylactic antibiotic use in the management of COPD exacerbations is conditional and unspecific.1,9 At present,

the optimal regimen of prophylactic antibiotics for exacerbations has not been well established, and there are no advices for an appropriate schedule and duration of specific antibiotic intervention. To further enhance information on the public health benefit-risk associated with this intervention, we here aimed to provide a comprehensive overview of the positive and negative effects of prophylactic antibiotics on COPD patients.

METHODS

Search strategy

We performed an update of the previous review by Herath et al. in 20136 according

to the PRISMA guidelines. Cochrane Central Register of Controlled Trials (CENTRAL), Medline, EMBASE, Web of Science, CINAHL, AMED and PsycINFO databases were systematically searched for relevant RCTs published from 29 August 2013 (when the review by Herath et al. ended) until 26 April 2017 using key elements of “COPD”,

(25)

“RCT” and “antibiotics” (details are presented in Table S1). References from identified studies and relevant review articles were also checked manually. No language restrictions were applied. For the final analysis, we included both the new studies from this searching strategy and previous studies from the review by Herath et al.

Selection criteria

Studies included in this review met the following criteria: (1) focus on the effects of prophylactic antibiotics in COPD patients; (2) study designs must be RCTs with placebo group; (3) COPD patients should be aged over 18 years and with a well-defined diagnosis of COPD and confirmed evidence of persistent airflow limitation (the presence of a post-bronchodilator FEV1/FVC < 0.7); (4) prophylactic antibiotics must be given for a minimum period of 12 weeks; (5) patients must be clinically stable without exacerbation for at least three weeks before enrolment. Studies that focused on combined antibiotics (≥ 2) and studies of patients with other respiratory disease (e.g. bronchiectasis, asthma) or related genetic diseases such as cystic fibrosis and primary ciliary dyskinesia were excluded.

Outcomes and data analysis

The Primary outcomes were: number of patients with exacerbations; frequency of exacerbation; health-related quality of life assessed by the St Georges Respiratory Questionnaire (SGRQ).10 The Secondary outcomes were: the median time to first

exacerbation; frequency of hospitalization; all-cause mortality; adverse events; antibiotic resistance; change in lung functions, bacteria load and airway inflammation. The influence of different schedules and durations of prophylactic antibiotic use on exacerbations and quality of life in COPD patients were explored. For the missing of standard deviation of SGRQ score change in two studies,11,12 we calculated it according

to Cochrane guideline (see Supplement data). All analyses were done in accordance with the intention-to-treat principle using Review Manager Version 5.3. Risk ratio (RR) or OR was calculated for binary outcomes, while mean difference (MD) was for continuous outcomes. Generic inverse variance (GIV) methods were used for non-standard types of both dichotomous and continuous data. Summary measures were pooled using random-effects models. If data could not be combined, we performed a descriptive analysis. Statistical heterogeneity among studies was assessed using conventional chi-squared (X2, or Chi2) test and I2 statistic of inconsistency. Sensitivity analysis was performed by

removing studies with a high risk for bias or deviation. A funnel plot was used to assess publication bias.

(26)

Effects of prophylactic antibiotics on COPD

2

Figure 1. Flow diagram of literature search and study selection.

data). All analyses were done in accordance with the intention‐to‐treat principle using Review  Manager Version 5.3. Risk ratio (RR) or OR was calculated for binary outcomes, while mean  difference (MD) was for continuous outcomes. Generic inverse variance (GIV) methods were  used for non‐standard types of both dichotomous and continuous data. Summary measures  were pooled using random‐effects models. If data could not be combined, we performed a  descriptive analysis. Statistical heterogeneity among studies was assessed using conventional 

chi‐squared  (X2,  or  Chi2)  test  and  I2  statistic  of  inconsistency.  Sensitivity  analysis  was 

Figure 1. Flow diagram of literature search and study selection.

RESULTS

Search results

From the 667 records generated by new search strategy, five new RCT studies were eligible and included (Figure 1). Together with the previous seven studies from the review by Herath et al.,6 a total of twelve RCTs were included for this systematic

review. However, of all twelve studies , one was a conference abstract,13 one was not

blinded,14 one did not report effect measures.15 In total, nine studies were qualified for

(27)

Table 1. Characteristics of included studies. Studies (1st author, year) Study design Country Patients (T/P) Age (year) (T/P) FEV1/FVC ratio (%) (T/P) Prophylactic Antibiotics (dose) Duration of

treatment & follow up

(months) Maintenance medication

Previous included studies

Albert, 2011 RCT US 570:572 65:66 42:43 Azithromycin, 250 mg daily 12 / 12 ICS, LABA, LAMA

He, 2010 RCT UK 18:18 68.8:69.3 46.9:48.6 Erythromycin, 125 mg, 3 times a day; 6 / 6 ICS,

Mygind, 2010 RCT Denmark 287:288 71 (Median) NA Azithromycin, 500 mg daily, 3 days a month 36 / 36 ICS, Theophylline, inhaled

anticholinergic, inhaled β-adrenergic

Sethi, 2010 RCT US 569:580 66.1:66.6 45.0:46.3 Moxifloxacin, 400 mg daily, 5 days every 8

weeks

12 / 18 LABA, LAMA, SABA, SAMA, ICS, theophylline;

Seemungal, 2008 RCT UK 53:56 66.6:67.8 48.9:50.9 Erythromycin, 250 mg twice daily 12 / 12 LABA, LAMA, theophylline

Banerjee, 2005 RCT UK 31:36 65.1:68.1 43.8:45.5 Clarithromycin, 500 mg once daily 3 / 3 ICS

Suzuki, 2001 RCT Japan 55:54 69.1:71.7 NA Erythromycin, 200-400 mg daily 12 / 12 Inhaled anticholinergic,

theophylline New included studies

*Brill, 2015 RCT UK (25:25:25):24 (70.9:70.4:67.9): 68.7 (51:51:45):51 T

1: Moxifloxacin, 400 mg, 5 times every 4 weeks; T2: Doxycycline, 100 mg daily;

T3: Azithromycin, 250 mg, 3 times a week;

3.25 /3.25 ICS

Shafuddin, 2015 RCT New Zealand 97:94 67.6:66.7 41.5:43.7 Roxithromycin, 300 mg daily 3 / 12 Not available

Simpson, 2014 RCT Australia 15:15 71.7:69.9 52.3:51.3 Azithromycin, 250 mg daily 3 / 6 ICS

Uzun, 2014 RCT Netherlands 47:45 64.7:64.9 38.0:40.3 Azithromycin, 500 mg, 3 times a week 12 / 12 LABA, LAMA, SABA, ICS,

Prednisolone

Berkhof, 2013 RCT Netherlands 42:42 67:68 42.2:43.2 Azithromycin, 250 mg, 3 times a week 3 / 4.5 LABA, LAMA, ICS

T/P: Treatment group versus Placebo group; ICS: inhaled corticosteroid; LABA: acting beta-2 agonists; LAMA: long-acting muscarinic antagonist; SABA: short-long-acting beta-2 agonists; SAMA: short-long-acting muscarinic antagonist; NA: data were not available; *This study designed

3 different treatment arms with one common placebo arm; †The study included 2 treatment arms, according to preset criteria, we only include the arm about single antibiotic use, the other arm in this study about combined antibiotic treatment is excluded;

Characteristics of included studies

The characteristics of twelve included studies are shown in Table 1, other specific baseline characteristics about COPD severity and exacerbation history were summarized in Table S2. All these studies were conducted over the last seventeen years involving 3,683 stable COPD patients, with 2932 patients involved in the meta-analysis. All included studies focused on one antibiotic arm with one placebo arm except the study by Brill et al.,16 which compared three antibiotics with one

common placebo and we treated this study as three independent RCTs (Trial 1-3: T1, T2, T3). In all, six antibiotics were investigated in this review: azithromycin,11,13,16-19

erythromycin,12,14,20 moxifloxacin,16,21 clarithromycin,15 roxithromycin22 and doxycycline.16

(28)

Effects of prophylactic antibiotics on COPD

2

Table 1. Characteristics of included studies.

Studies (1st author, year) Study design Country Patients (T/P) Age (year) (T/P) FEV1/FVC ratio (%) (T/P) Prophylactic Antibiotics (dose) Duration of

treatment & follow up

(months) Maintenance medication

Previous included studies

Albert, 2011 RCT US 570:572 65:66 42:43 Azithromycin, 250 mg daily 12 / 12 ICS, LABA, LAMA

He, 2010 RCT UK 18:18 68.8:69.3 46.9:48.6 Erythromycin, 125 mg, 3 times a day; 6 / 6 ICS,

Mygind, 2010 RCT Denmark 287:288 71 (Median) NA Azithromycin, 500 mg daily, 3 days a month 36 / 36 ICS, Theophylline, inhaled

anticholinergic, inhaled β-adrenergic

Sethi, 2010 RCT US 569:580 66.1:66.6 45.0:46.3 Moxifloxacin, 400 mg daily, 5 days every 8

weeks

12 / 18 LABA, LAMA, SABA, SAMA, ICS, theophylline;

Seemungal, 2008 RCT UK 53:56 66.6:67.8 48.9:50.9 Erythromycin, 250 mg twice daily 12 / 12 LABA, LAMA, theophylline

Banerjee, 2005 RCT UK 31:36 65.1:68.1 43.8:45.5 Clarithromycin, 500 mg once daily 3 / 3 ICS

Suzuki, 2001 RCT Japan 55:54 69.1:71.7 NA Erythromycin, 200-400 mg daily 12 / 12 Inhaled anticholinergic,

theophylline New included studies

*Brill, 2015 RCT UK (25:25:25):24 (70.9:70.4:67.9): 68.7 (51:51:45):51 T

1: Moxifloxacin, 400 mg, 5 times every 4 weeks; T2: Doxycycline, 100 mg daily;

T3: Azithromycin, 250 mg, 3 times a week;

3.25 /3.25 ICS

Shafuddin, 2015 RCT New Zealand 97:94 67.6:66.7 41.5:43.7 Roxithromycin, 300 mg daily 3 / 12 Not available

Simpson, 2014 RCT Australia 15:15 71.7:69.9 52.3:51.3 Azithromycin, 250 mg daily 3 / 6 ICS

Uzun, 2014 RCT Netherlands 47:45 64.7:64.9 38.0:40.3 Azithromycin, 500 mg, 3 times a week 12 / 12 LABA, LAMA, SABA, ICS,

Prednisolone

Berkhof, 2013 RCT Netherlands 42:42 67:68 42.2:43.2 Azithromycin, 250 mg, 3 times a week 3 / 4.5 LABA, LAMA, ICS

T/P: Treatment group versus Placebo group; ICS: inhaled corticosteroid; LABA: acting beta-2 agonists; LAMA: long-acting muscarinic antagonist; SABA: short-long-acting beta-2 agonists; SAMA: short-long-acting muscarinic antagonist; NA: data were not available; *This study designed

3 different treatment arms with one common placebo arm; †The study included 2 treatment arms, according to preset criteria, we only include the arm about single antibiotic use, the other arm in this study about combined antibiotic treatment is excluded;

Quality assessment

The review authors’ judgment about each risk of bias item in each study can be seen in Figure S1.1. The risk of bias items presented as percentage across all included studies were presented in Figure S1.2. There was no reporting bias in all included studies; only 2 studies14,16 have potential high risk in the blinding process. For the remaining of bias

items, only a small proportion of unclear bias exists. Overall, low risk of bias dominates in all domains of bias.

Primary outcomes

Seven studies involving 2,642 participants11,12,17-21 reported the number of patients

(29)

0.74-which requires further confirmation. The risk difference (RD) between antibiotic and placebo  

Figure 2. Forest plot of risk ratio (antibiotics versus placebo) for total number of patients with  one  or  more  exacerbations  stratified  by  (a)  schedule  of  prophylactic  antibiotics  and  (b) 

duration of prophylactic antibiotics. M‐H: Mantel‐Haenszel; *Studies reviewed by Herath et al. 

in 2013.

Figure 2. Forest plot of risk ratio (antibiotics versus placebo) for total number of patients with one or more exacerbations stratified by (a) schedule of prophylactic antibiotics and (b) duration of prophylactic antibiotics. M-H: Mantel-Haenszel; *Studies reviewed by Herath et al. in 2013.

and intermittent subgroups. However, the difference between other subgroups with a distinct trend toward significance (p = 0.07) suggested that using antibiotics ≤ 6 months may achieve better treatment effects (RR 0.59, 95% CI 0.40-0.86) than longer time (RR 0.84, 95% CI 0.77-0.93), which requires further confirmation. The risk difference (RD) between antibiotic and placebo groups is presented in Figure 3, for erythromycin, the RD is substantial (RD -0.24, 95% CI -0.39 to -0.08), the corresponding number needed to treat (NNT) was 4; for azithromycin, the RD was moderate (RD -0.14, 95% CI -0.20 to -0.08), the NNT was 7; no statistically significant effect for moxifloxacin intervention.

(30)

Effects of prophylactic antibiotics on COPD

2

Figure 3. Forest plot of risk difference (antibiotics versus placebo) for total number of patients  with one or more exacerbations stratified by types of antibiotics. M‐H: Mantel‐Haenszel test;  *Studies reviewed by Herath et al. in 2013.  groups is presented in Figure 3, for erythromycin, the RD is substantial (RD ‐0.24, 95% CI ‐0.39  to ‐0.08), the corresponding number needed to treat (NNT) was 4; for azithromycin, the RD  was moderate (RD ‐0.14, 95% CI ‐0.20 to ‐0.08), the NNT was 7; no statistically significant effect  for moxifloxacin intervention.  Use of prophylactic antibiotic was also associated with a significant reduction in the frequency 

of  exacerbations  (RR  0.74,  95%  CI  0.60‐0.92,  Figure  4).  As  the  study  by  Brill  et  al.16  has  a 

potential risk of bias in the blinding process, a sensitivity analysis was done with the other 6  studies,  which  resulted  in  a  31%  RR  reduction  of  exacerbations  among  patients  taking  prophylactic antibiotics (RR 0.69, 95% CI 0.58‐0.82). In subgroup analysis showed in Figure 4,  macrolides  (azithromycin,  erythromycin  and  roxithromycin)  showed  beneficial  effects  on  frequency reduction of exacerbations, the benefits from both azithromycin and erythromycin  were  of  clinical  significance.  However,  this  beneficial  effect  was  not  seen  in  the  use  of 

Figure 3. Forest plot of risk difference (antibiotics versus placebo) for total number of patients with one or more exacerbations stratified by types of antibiotics. M-H: Mantel-Haenszel test; *Studies reviewed by Herath et al. in 2013.

Use of prophylactic antibiotic was also associated with a significant reduction in the frequency of exacerbations (RR 0.74, 95% CI 0.60-0.92, Figure 4). As the study by Brill et al.16 has a potential risk of bias in the blinding process, a sensitivity analysis was done

with the other 6 studies, which resulted in a 31% RR reduction of exacerbations among patients taking prophylactic antibiotics (RR 0.69, 95% CI 0.58-0.82). In subgroup analysis showed in Figure 4, macrolides (azithromycin, erythromycin and roxithromycin) showed beneficial effects on frequency reduction of exacerbations, the benefits from both azithromycin and erythromycin were of clinical significance. However, this beneficial effect was not seen in the use of moxifloxacin and doxycycline. These subgroup differences for frequency of exacerbations were of statistical significance (p = 0.02). Health-related quality of life using SGRQ was measured in seven studies.11,12,16-19,21 When

we performed a sensitivity analysis by removing the study by Berkhof et al.,18 which

was very different from the other data, the heterogeneity reduced sharply (I2 changed

from 92% to 0%). Hence, only the remaining 6 studies were included for the final meta-analysis. The pooled result indicated that prophylactic antibiotics led to a significant improvement in the total SGRQ score (MD -1.55, 95% CI -2.59 to -0.51, Figure 5). In subgroup analysis, the improvement of SGRQ score was not seen in both continuous and intermittent antibiotics. However, another subgroup result indicated that the total

(31)

Chapter 2

Health‐related quality of life using SGRQ was measured in seven studies.11,12,16‐19,21 When we 

performed a  sensitivity  analysis  by  removing  the  study  by  Berkhof  et al.,18  which  was  very 

different from the other data, the heterogeneity reduced sharply (I2 changed from 92% to 0%). 

Hence,  only  the  remaining  6  studies  were  included  for  the  final  meta‐analysis.  The  pooled  result  indicated  that  prophylactic  antibiotics  led  to  a  significant  improvement  in  the  total  SGRQ score (MD ‐1.55, 95% CI ‐2.59 to ‐0.51, Figure 5). In subgroup analysis, the improvement  of SGRQ score was not seen in both continuous and intermittent antibiotics. However, another  

Figure 4. Forest plot of risk ratio (antibiotics versus placebo) for frequency of exacerbations 

stratified by types of antibiotics. SE: standard error; IV: inverse variance; *Studies reviewed by 

Herath et al. in 2013; T1‐3: three independent RCTs in study by Brill et al. 

Figure 4. Forest plot of risk ratio (antibiotics versus placebo) for frequency of exacerbations stratified by types of antibiotics. SE: standard error; IV: inverse variance; *Studies reviewed by Herath et al. in 2013; T1-3: three independent RCTs in study by Brill et al.

SGRQ score significantly changed by long-term intervention (MD -1.70, 95% CI% -2.81 to -0.60), although it was not changed by short-term (≤ 6) intervention (MD -0.34, 95% CI -3.43 to 2.75).

Four studies12,17,19,21 also reported the component scores of SGRQ (Figure S2). Both

the symptom (MD -3.89, 95% CI -5.48 to -2.31) and impact (MD -1.32, 95% CI -2.61 to -0.03) scores were improved with prophylactic antibiotics. However, the activity score did not show any significant improvement. Of note, none of these improvements mentioned above in SGRQ score reached the hypothesized clinically beneficial level (> 4-unit reduction).10

(32)

Effects of prophylactic antibiotics on COPD

2

Secondary outcomes

Seven studies involving 2,803 patients reported the median time to first exacerbation (Table S3). Four studies indicated that using prophylactic antibiotics lengthened the median time to first exacerbation signficantly.12,17,19,20 Two other studies found

a similar trend, but without statistical significance.18,21 Only one study showed

the opposite result in antibiotic and placebo arms.22

Figure 5. Forest plot of mean difference (antibiotics versus placebo) of quality of life by SGRQ stratified by (a) schedule of prophylactic antibiotics and (b) duration of prophylactic antibiotics. SGRQ: St Georges Respiratory Questionnaire; IV: inverse variance; SE: standard error; *Studies reviewed by Herath et al. in 2013; T1-3: three independent RCTs in study by Brill et al.

Figure 5. Forest plot of mean difference (antibiotics versus placebo) of quality of life by SGRQ  stratified  by  (a)  schedule  of  prophylactic  antibiotics  and  (b)  duration  of  prophylactic 

antibiotics.  SGRQ: St  Georges  Respiratory  Questionnaire;  IV:  inverse  variance;  SE:  standard 

error; *Studies reviewed by Herath et al. in 2013; T

1‐3: three independent RCTs in study by Brill 

et al. 

subgroup  result  indicated  that  the  total  SGRQ  score  significantly  changed  by  long‐term  intervention (MD ‐1.70, 95% CI% ‐2.81 to ‐0.60), although it was not changed by short‐term (   6) intervention (MD ‐0.34, 95% CI ‐3.43 to 2.75). 

(33)

Chapter 2

32

Figure 7. Forest plot of risk ratio (antibiotics versus placebo) for adverse events. M‐H: Mantel‐

Haenszel test; *Studies reviewed by Herath et al. in 2013.

show  significant  difference  between  two  arms  (RR  0.93,  95%  CI  0.83‐1.05,  I2  =  2%).  In 

subgroups,  gastrointestinal  disorders  were  more  frequent  in  the  intervention  group  than  control group (RR 1.87, 95 CI 0.98‐3.59, Figure S4) with a boundary statistical significance (p =  0.06).  However,  no  statistical  significant  difference  for  respiratory  and  cardiovascular  disorders were found. 

Eight studies had the bacteriological assessments (see Table S4),12,15‐21 however, only three 

studies with five RCTs reported the quantitative results for antibiotic resistance.16,17,19 Due to 

the different definition about bacterial resistant outcome in study by Uzun et al, only the other  homogeneous studies involving four RCTs were included for pooled results (OR 4.49, 95% CI  2.48‐8.12,  Figure  8),  long‐term  (versus  short‐term)  and  continuous  (versus  intermittent)  antibiotic intervention seems to cause more antibiotic resistance, although these subgroup  difference did not reach statistical significant level. Antibiotic resistance appeared in all types  of  antibiotics  involved  (Figure  9),  although  the  result  from  moxifloxacin  did  not  reach  statistical  significance.  No  subgroup  differences  about  this  outcome  were  seen  among  azithromycin, moxifloxacin and doxycycline (p = 0.63).  

Eight studies11,13,16‐18,20‐22 provided the data on changes of lung function (Table S5). However, 

no study found significant increase by antibiotic intervention compared with placebo. Mygind 

et al. did not compare the lung function change directly, but measured and compared the lung 

function  in  both  groups  at  enrolment  and  endpoint  separately,  they  also  did  not  find  any 

significant difference.13 

Figure 6. Forest plot of risk ratio (antibiotics versus placebo) for frequency of hospitalization. M-H: Mantel-Haenszel test; *Studies reviewed by Herath et al. in 2013.

were improved with prophylactic antibiotics. However, the activity score did not show any  significant  improvement.  Of  note,  none  of  these  improvements  mentioned  above  in  SGRQ 

score reached the hypothesized clinically beneficial level (> 4‐unit reduction).10  Secondary outcomes  Seven studies involving 2,803 patients reported the median time to first exacerbation (Table  S3). Four studies indicated that using prophylactic antibiotics lengthened the median time to  first exacerbation signficantly.12,17,19,20 Two other studies found a similar trend, but without  statistical significance.18,21 Only one study showed the opposite result in antibiotic and placebo  arms.22 

The  frequency  of  hospitalization  related  to  COPD  was  pooled  from  five  studies  with  2,576 

participants,17‐21  no  significant  difference  was  observed  between  antibiotic  and  placebo 

groups  (RR  0.94,  95%  CI  0.83‐1.06,  Figure  6).  Also,  no  difference  in  the  rate  of  all‐cause  mortality were found between the two arms (Figure S3).  

Eight  studies  involving  2,833  participants  reported  adverse  events  related  to  antibiotic  use.11,12,17‐22 Overall, there was no significant difference between two comparison arms in the  rate of adverse events (RR 1.09, 95% CI 0.84‐1.42, Figure 7). As there was a lack of uniform  definition about adverse events, the heterogeneity was substantial (I2 = 73%). Considering that  the result by Shafuddin et al.22 was deviant from the other seven studies, a sensitivity analysis  was, therefore, performed after removal of this study. The homogeneous result also did not  Figure 6. Forest plot of risk ratio (antibiotics versus placebo) for frequency of hospitalization.  M‐H: Mantel‐Haenszel test; *Studies reviewed by Herath et al. in 2013.

Figure 7. Forest plot of risk ratio (antibiotics versus placebo) for adverse events. M-H: Mantel-Haenszel test; *Studies reviewed by Herath et al. in 2013.

The frequency of hospitalization related to COPD was pooled from five studies with 2,576 participants,17-21 no significant difference was observed between antibiotic and

placebo groups (RR 0.94, 95% CI 0.83-1.06, Figure 6). Also, no difference in the rate of all-cause mortality were found between the two arms (Figure S3).

Eight studies involving 2,833 participants reported adverse events related to antibiotic use.11,12,17-22 Overall, there was no significant difference between two comparison

arms in the rate of adverse events (RR 1.09, 95% CI 0.84-1.42, Figure 7). As there was a lack of uniform definition about adverse events, the heterogeneity was substantial (I2 = 73%). Considering that the result by Shafuddin et al.22 was deviant from the other

seven studies, a sensitivity analysis was, therefore, performed after removal of this study. The homogeneous result also did not show significant difference between two arms (RR 0.93, 95% CI 0.83-1.05, I2 = 2%). In subgroups, gastrointestinal disorders were

(34)

Effects of prophylactic antibiotics on COPD

2

Three  studies  reported  the  change  of  bacterial  load.11,15,16  Although  both  Brill  et  al.  and 

Simpson  et  al.  have  found  the  more  reduction  of  bacterial  load  by  prophylactic  antibiotic  compared with placebo, the results did not reach the level of statistical significance, even both  quantitative culture and 16S qPCR methods were used by Brill et al. Benerjee et al. also did  not find a significant difference between pre‐and post‐ sputum cfu numbers/bacterial (PPM)  isolates in two arms. 

Figure  8.  Forest  plot  of  odds  ratio  (antibiotics  versus  placebo)  for  antibiotic  resistance  stratified  by  (a)  schedule  of  prophylactic  antibiotics  and  (b)  duration  of  prophylactic 

antibiotics.  IV:  inverse  variance; *Studies  reviewed  by  Herath  et  al.  in  2013;  T

1‐3:  three 

independent RCTs in study by Brill et al.

Figure 8. Forest plot of odds ratio (antibiotics versus placebo) for antibiotic resistance stratified by (a) schedule of prophylactic antibiotics and (b) duration of prophylactic antibiotics. IV: inverse variance; *Studies reviewed by Herath et al. in 2013; T

1-3: three independent RCTs in study by Brill et al.

Figure S4) with a boundary statistical significance (p = 0.06). However, no statistical significant difference for respiratory and cardiovascular disorders were found.

Eight studies had the bacteriological assessments (see Table S4),12,15-21 however, only three

studies with five RCTs reported the quantitative results for antibiotic resistance.16,17,19 Due

to the different definition about bacterial resistant outcome in study by Uzun et al, only the other homogeneous studies involving four RCTs were included for pooled results (OR 4.49, 95% CI 2.48-8.12, Figure 8), long-term (versus short-term) and continuous (versus intermittent) antibiotic intervention seems to cause more antibiotic resistance, although these subgroup difference did not reach statistical significant level. Antibiotic resistance appeared in all types of antibiotics involved (Figure 9), although the result

(35)

Figure  9:  Forest  plot  of  odds  ratio  (antibiotics  versus  placebo)  for  antibiotic  resistance  stratified by types of antibiotics. SE: standard error; IV: inverse variance; *Studies reviewed by  Herath et al. in 2013; T1‐3: three independent RCTs in study by Brill et al.  The change of airway inflammation was only reported in two studies.11,16 The study by Brill et  al. showed that no significant changes were seen in cytokines IL‐6, IL‐8 and IL‐1  in any of  three antibiotic arms compared with placebo.16 Similarly, Simpson et al. also did not report a 

significant  reduction  in  sputum  neutrophil  proportion  level  of  IL‐8  in  those  who  received 

azithromycin compared to placebo group.11 

   

Figure 9. Forest plot of odds ratio (antibiotics versus placebo) for antibiotic resistance stratified by types of antibiotics. SE: standard error; IV: inverse variance; *Studies reviewed by Herath et al. in 2013; T1-3: three independent RCTs in study by Brill et al.

from moxifloxacin did not reach statistical significance. No subgroup differences about this outcome were seen among azithromycin, moxifloxacin and doxycycline (p = 0.63). Eight studies11,13,16-18,20-22 provided the data on changes of lung function (Table S5).

However, no study found significant increase by antibiotic intervention compared with placebo. Mygind et al. did not compare the lung function change directly, but measured and compared the lung function in both groups at enrolment and endpoint separately, they also did not find any significant difference.13

Three studies reported the change of bacterial load.11,15,16 Although both Brill et al.

and Simpson et al. have found the more reduction of bacterial load by prophylactic antibiotic compared with placebo, the results did not reach the level of statistical significance, even both quantitative culture and 16S qPCR methods were used by Brill et al. Benerjee et al. also did not find a significant difference between pre-and post- sputum cfu numbers/bacterial (PPM) isolates in two arms.

The change of airway inflammation was only reported in two studies.11,16 The study by

Brill et al. showed that no significant changes were seen in cytokines IL-6, IL-8 and IL-1β

in any of three antibiotic arms compared with placebo.16 Similarly, Simpson et al. also

did not report a significant reduction in sputum neutrophil proportion level of IL-8 in those who received azithromycin compared to placebo group.11

Referenties

GERELATEERDE DOCUMENTEN

In this thesis, we aim to develop a comprehensive profile on the effectiveness of antibiotic use for acute exacerbations of COPD both prescribed prophylactically

This updated systematic review confirms the benefit of prophylactic antibiotics in preventing exacerbations in stable patients with moderate to severe COPD, this benefit

Among patients with a first identified AECOPD, during their treatment period of oral prednisone or prednisolone (3 days before till 7 days after the index date), those who were

Real-world short- and long-term effects of antibiotic therapy on acute exacerbations of COPD in outpatients: a cohort study under the PharmLines Initiative.. Submitted

The impact of comorbidities on quality of life in COPD patients are well reported, however, potential drug interactions between drugs for these comorbidities and ABs

Within 24 weeks following initiation of varenicline treatment, we found no significantly increased risk of NPAEs in both the general and COPD population compared with

5-7 However, subsequent post-marketing reports related to neuropsychiatric adverse events (NPAEs), such as depression, anxiety, sleep disorder but also suicide,

This review indicates that, even though PSSA has a lot of potential, the effect estimates generated by the PSSA are usually lower than the effect estimates generated by parallel