• No results found

University of Groningen Improving treatment outcomes of tuberculosis Pradipta, Ivan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Improving treatment outcomes of tuberculosis Pradipta, Ivan"

Copied!
212
0
0

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

Hele tekst

(1)

Improving treatment outcomes of tuberculosis

Pradipta, Ivan

DOI:

10.33612/diss.113506043

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):

Pradipta, I. (2020). Improving treatment outcomes of tuberculosis: towards an antimicrobial stewardship

program. University of Groningen. https://doi.org/10.33612/diss.113506043

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)

1

Improving treatment outcomes of tuberculosis

Towards an antimicrobial stewardship program

(3)

ISBN : 978-94-034-2378-4 (printed book) ISBN : 978-94-034-2379-1 (electronic book) Author : Ivan Surya Pradipta

Cover : Ivan Surya Pradipta (concept) and Hartanto (graphic design) Lay-out : Lara Leijtens, persoonlijkproefschrift.nl

Printing : Ridderprint BV | www.ridderprint.nl

This thesis was conducted within the Groningen University Institute for Drug Exploration (GUIDE). Financial support for the printing of this thesis was kindly provided by the Graduate School of Sciences, University of Groningen, the Netherlands.

Ivan Surya Pradipta received a Ph.D. scholarship from the Indonesia Endowment Fund for Education or LPDP to conduct all studies in this book.

(4)

3

Improving treatment outcomes of

tuberculosis

Towards an antimicrobial stewardship program

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 Monday 24 February 2020 at 11.00 hours

by

Ivan Surya Pradipta

born on 20 June 1983 in Yogyakarta, Indonesia

(5)

Prof. J.W.C. Alffenaar

Assessment Committee

Prof. F.G.J. Cobelens

Prof. K. Taxis Prof. T.S. van der Werf

(6)

To my respected teachers, my beloved parents, brothers, wife and children.

(7)
(8)

TABLE OF CONTENTS

Chapter 1 General Introduction 8

Chapter 2 Risk factors of multidrug-resistant tuberculosis: a global systematic review and meta-analysis

Journal of Infection 2018 Dec;77(6):469-478.

18

Chapter 3 Predictors for treatment outcomes among patients with drug-susceptible tuberculosis in the Netherlands: a retrospective cohort study

Clinical Microbiology and Infection 2019 Jun;25(6):761.e1-761.e7.

48

Chapter 4 Treatment outcomes of drug-resistant tuberculosis in the Netherlands, 2005-2015

Antimicrobial Resistance and Infection Control 2019 Jul 12;8:115.

72

Chapter 5 A systematic review of measures to estimate adherence and persistence to multiple medications

Journal of Clinical Epidemiology 2019 Apr;108:44-53.

100

Chapter 6 Interventions to improve medication adherence in patients with latent and active tuberculosis infection: a systematic review of randomized controlled studies

Submitted

126

Chapter 7 Barriers and strategies to successful tuberculosis treatment in a high-burden tuberculosis setting: a qualitative study from the patient’s perspective

Submitted

150

Chapter 8 Discussion and future perspectives 178

Addendum Summary

Nederlandse Samenvatting Ringkasan

Acknowledgements About the Author PhD Portfolio 194 197 201 205 208 209

(9)
(10)

1

GENERAL INTRODUCTION

(11)

Tuberculosis (TB) remains a continuous global problem. TB, an infectious disease caused by Mycobacterium tuberculosis (M.tb), is an ancient disease that was described about 70,000 years ago.(1) The pathogen can easily transmit from a TB patient to healthy people by air transmission. Nowadays, TB has spread worldwide owing to its ability to establish a latent infection, the capability of long persistence in the host,(2) and variations in its sensitivity to antibiotics.(3)

Global burden of tuberculosis disease

Nowadays, TB is one of the top 10 causes of death.(4) The World Health Organization (WHO) estimated that 10 million people developed TB and 1.3 million patients died because of TB in 2017.(4) A global report estimated that a large proportion of the world population has a latent TB infection (an existing M.tb with no evidence of clinically manifest active tuberculosis).(4) In approximately 10% of the population infected with latent TB, active TB disease will develop during the lifetime.(4) As a consequence, an active TB status is associated with disease transmission and substantial morbidity, which is even more problematic in the case of drug-resistant tuberculosis (DR-TB).

Drug-resistant tuberculosis (DR-TB) is a resistance of M.tb to one or more anti-tuberculosis drugs. The resistance patterns can be classified into mono-, poly-, rifampicin-, multidrug- and extensive drug-resistance.(5) Mono-resistant TB is defined as resistance to one first-line anti-TB drug only, while poly-resistant TB is resistance to more than one first-first-line anti-TB drugs other than isoniazid and rifampicin.(5) The more extensive patterns of drug resistance are multi-drug resistant TB (MDR-TB) and extensive drug-resistant TB (XDR-TB). MDR-TB is defined as resistance to at least both of the most potent anti-TB drugs (isoniazid and rifampicin), while XDR-TB is resistance to any fluoroquinolone, and at least one of three second-line injectable drugs (capreomycin, kanamycin and amikacin) in addition to multidrug resistance.(5) As one of the most potent TB drugs, rifampicin resistance has been a great concern in TB treatment. Hence, the resistance of rifampicin has been included in the classification of DR-TB as rifampicin resistant-TB (RR-TB). Considering sensitivity to the anti-tuberculosis drugs, RR-TB is defined as resistance to rifampicin, with or without resistance to other anti-TB drugs, including any resistance to rifampicin, in the form of mono-resistance, poly-resistance, MDR or XDR.(5)

The WHO identified 558,000 people who developed RR-TB and among this group, 82% had MDR-TB.(4) MDR-TB was reported responsible for about a quarter of all deaths caused by antimicrobial-resistant infections(6) and contributed to an estimated 14% of TB deaths worldwide.(4) If it comes to the treatment of MDR-TB, such patients should take multiple drugs and the drug treatment can be up to 24 months.(7) Combination of prolonged therapy,

(12)

11 General Introduction optimal outcomes than DS-TB or mono-resistant TB,(4,8) with an overall low success rate for treatment of MDR-TB of 55%.(4)

The global target for TB elimination 2035: low versus high burden TB countries

The WHO has divided all countries into two general categories of TB countries, i.e. low and high burden countries. TB low burden countries are the countries with TB incidence below 10 per 100,000 population,(9) while high burden countries are the countries with the highest absolute number of estimated incident cases and those with the most severe burden in terms of incidence rate per capita.(10) Although the quantitative burden level is different among low and high burden countries, the global target defined by the WHO is to achieve a global incidence rate of TB below 10 per 100,000 population by the year 2035.(11) To achieve the strict targets, the WHO initiated a global strategy for TB prevention, care and control. The program, called the END-TB strategy, includes three main pillars, i.e., 1) integrated, patient-centered care and prevention, 2) strong policies and supportive systems, and 3) intensified research and innovation.(11) To respond to the global target, the specific target set in the low burden TB countries is to reduce the TB incidence to below 1 case per 100,000 population in 2035.(12) This challenging goal was followed up by the low burden TB countries. As an example, the Netherlands, a country with a total population of 17 million inhabitants in 2017, committed to reducing the TB case burden from 5.9 cases per 100,000 population in 2016 (13) to below 1 case per 100,000 population by 2035.(14) As an example for the high burden countries, Indonesia with a total estimated population of 264 million people, set a target from 254 cases per 100,000 population in 2017 (15) to 10 cases per 100,000 population by 2035.(16) More intensified research and health programs and policies are hence needed to achieve such strict targets within the coming 15 years.

Therapeutic failure is an essential problem for controlling drug-resistant tuberculosis

The mechanism of development of DR-TB can be classified into two main pathways: 1) primary drug-resistance which occurs when resistant strains are transmitted into a new host, and 2) secondary or acquired drug resistance which occurs through the acquisition of drug resistance mutations to one or more drugs.(17–19) Increasing the risk of primary drug resistance is higher in areas with a high prevalence of DR-TB. The transmission can be enhanced by environmental conditions such as crowding, poor ventilation and lack of infection control. On the other hand, secondary/acquired DR-TB is more affected by the low quality of drug treatment such as non-adherence to drugs and low drug exposure due to adverse drug reaction, drug interaction or inadequately dose.(5)

A history of previous TB treatment is one of the main risk factors for MDR-TB.(20) This evidence indicates that MDR-TB can be caused by secondary drug-resistance due to unsuccessful treatment from a prior treatment. A global report estimated that 3.5% of all

(13)

new TB cases had MDR or RR-TB, while 18% of previously treated cases had MDR or RR-TB in 2017.(4) The report provides evidence that unsuccessful TB treatment in the first episode of disease should be a concern for controlling DR-TB.

Besides TB case-finding, improving the rational use of anti-TB drugs has been an essential part of DR-TB control. Several studies have reported acquired drug-resistance due to therapeutic failures of TB treatment.(21–25) A study also demonstrated that TB patients with acquired drug resistance were at significantly increased risk for poor treatment outcome.(26) The global TB problem will become even more complicated when the trend of DR-TB transmission is shifting from acquired drug resistance to primary drug resistance. A dynamic modeling study in India showed that there is a predicted significant increasing transmission trend of primary drug-resistance from 15% in 2012 to 85% in 2032.(27) Therefore, emerging problems associated with TB drug treatment management should be primarily controlled to avoid the development of both primary and secondary DR-TB.

Antimicrobial stewardship program in TB disease

Antimicrobial stewardship is defined as “a coherent set of activities which promotes using antimicrobials in ways that ensure sustainable access to effective therapy for all who need them”.(28) The primary goal is to optimize clinical outcomes and minimize unintended consequences of antimicrobial use, including toxicity, the selection of pathogenic organisms and the emergence of resistance.(29) An effective program should be designed based on setting priorities.(30) Engagement of multiple actors such as health care providers, government, patients, and society are also needed to enhance the benefits of an antimicrobial stewardship program.(28)

Although the terminology of antimicrobial stewardship is uncommonly used in the research and management of TB disease, the principles are still very relevant to achieve the global target of TB elimination in 2035. Activities related to treatment supervision can be part of antimicrobial stewardship programs for controlling DR-TB. Scientific knowledge of anti-TB drug use is needed to determine priority areas and plans for intervention.(31) The development of an effective program requires a more personalized approach that takes into account the heterogeneity in terms of patient characteristics across geographical areas and health care systems. In the development of intervention programs, it should be explored which high-risk groups of patients should be targeted for therapeutic failures. Furthermore, since treatment adherence is widely known as the main problem in TB disease, it should be explored who should be monitored and which intervention strategies should be developed to maintain good treatment adherence. Therefore, a comprehensive picture of unsuccessful treatment risks, treatment barriers and treatment non-adherence in TB disease should be

(14)

13 General Introduction Unfortunately, comprehensive information on the current situation of high-risk groups for therapeutic failure and drug-related problems in TB disease is lacking. The controversial information is due to the differences in designs and settings of previous studies as well as the absence of more recent data, which is essential given the rapidly changing TB epidemiology. Updated studies and novel strategies are required for developing effective antimicrobial stewardship programs in TB disease. Therefore, this thesis attempted to provide a scientific knowledge base on the essential aspects to improve treatment outcomes of TB patients in the near future.

Thesis objectives and outlines

In this thesis, we aim to research both low and high TB endemic settings with the Netherlands as a low burden TB country and Indonesia as a high burden TB country. The empirical research will apply drug utilization and pharmacoepidemiological approaches to support potentially effective strategies to improve treatment outcomes as part of drug resistance control in TB disease. The specific objectives of this thesis can be listed as follows:

1. To analyze high-risk groups for therapeutic failure among TB patients.

2. To analyze potential interventions to improve treatment adherence in TB patients. 3. To analyze treatment barriers and potential strategies for successful TB treatment. In Chapter 2, we aim to identify risk factors for MDR-TB as a fundamental part of this thesis. We will present a global systematic review and meta-analysis study that will identify patients’ and geographical characteristics that are associated with the development of MDR-TB.

Since acquired resistant TB can develop after the therapeutic failure of drug-susceptible TB (DS-TB), predictors for unsuccessful TB treatment among DS-TB patients are studied in Chapter 3. Using a nationwide TB database from the Netherlands, a low TB incidence country, we aim to identify the incidence, high-risk groups and predictors for unsuccessful TB treatment outcomes in an adult DS-TB population.

In Chapter 4, the current situation of the high-risk DR-TB patients in the Netherlands is investigated. We will present the prevalence of different types of DR-TB cases and the characteristics of patients who were lost to follow-up for the treatment outcome. To gain more insights in the treatment outcomes, we will determine the incidence, high-risk groups and predictors for poor outcome of TB treatment among all types of DR-TB patients also including MDR-TB patients.

In Chapter 5, we will systematically review measures of adherence and persistence to multiple medications using a pharmacoepidemiological approach. Since there is a lack of

(15)

studies in adherence to TB medications using real-world prescription databases, an example of cardio-metabolic medications that represent chronic diseases treated with multiple medications will be studied in this chapter. This review will implicitly provide scientific considerations for measuring adherence in TB disease using a prescription database. Another TB drug adherence topic is continued in Chapter 6. This chapter will describe current studies on the impact of interventions to improve TB treatment adherence. In this chapter, we will also discuss several strategies to conduct valid intervention studies on TB treatment adherence.

In Chapter 7, we investigate treatment barriers among TB patients in a high burden TB setting. A qualitative study will be performed to determine treatment barriers from the patient perspective. In this chapter, we will discuss potential strategies to improve successful treatment among TB disease in Indonesia.

Finally, Chapter 8 will describe an overview, general discussion, implication and future perspectives related to the findings of the studies presented in this thesis.

(16)

15 General Introduction

REFERENCES

1. M Cristina G, Brisse S, Brosch R, Fabre M, Omaïs B, Marmiesse M, et al. Ancient origin and gene mosaicism of the progenitor of Mycobacterium tuberculosis. PLoS Pathog. 2005;1(1):55–61. 2. Chaves AS, Rodrigues MF, Mattos AMM,

Teixeira HC. Challenging Mycobacterium tuberculosis dormancy mechanisms and their immunodiagnostic potential. Brazilian J Infect Dis. 2015;19(6):636–42.

3. Aldridge BB, Fernandez-Suarez M, Heller D, Ambravaneswaran V, Irimia D, Toner M, et al. Asymmetry and aging of mycobacterial cells lead to variable growth and antibiotic susceptibility. Science (80- ). 2012;335(6064):100–4. 4. World Health Organization (WHO). Global

Tuberculosis Report 2018. Geneva: World health Organization; 2018.

5. WHO. Companion Handbook to the WHO Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis. Companion Handbook to the WHO Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis. 2014. 1–20 p.

6. O’Neill J. Tackling drug-resistant infections globally: final report and recomendations [Internet]. Welcome Trust and HM Governemnet. London; 2016. Available from: https://amr-review.org/sites/default/files/160518_Final paper_with cover.pdf

7. Pontali E, Raviglione MC, Migliori GB. Regimens to treat multidrug-resistant tuberculosis: past, present and future perspectives. Eur Respir Rev. 2019;28(152):1–7.

8. Pontali E, Visca D, Centis R, D’Ambrosio L, Spanevello A, Migliori GB. Multi and extensively drug-resistant pulmonary tuberculosis: Advances in diagnosis and management. Curr Opin Pulm Med. 2018;24(3):244–52.

9. Clancy L, Rieder HL, Enarson DA, Spinaci S. Tuberculosis elimination in the countries of Europe and other industrialized countries. Eur Respir J. 1991;4(10):1288–95.

10. WHO. Use of high burden country lists for TB by WHO in the post-2015 era. WHO Press. 2015;(April):19.

11. World Health Organization. Implementing the END TB strategy: the essentials. WHO. Geneva: World health Organization; 2015.

12. Lönnroth K, Migliori GB, Abubakar I, D’Ambrosio L, De Vries G, Diel R, et al. Towards tuberculosis elimination: An action framework for low-incidence countries. Eur Respir J. 2015;45(4):928–52.

13. ECDC. Tuberculosis surveillance and monitoring in Europe [Internet]. 2018th ed. WHO Regional Office for Europe (WHO/Europe) and the European Centre for Disease Prevention and Control (ECDC).; 2018. 206 p. Available from: https://ecdc.europa.eu/sites/portal/files/ documents/ecdc-tuberculosis-surveillance-monitoring-Europe-2018-19mar2018.pdf 14. de Vries G, Riesmeijer R. National Tuberculosis

Control Plan 2016-2020 : Towards elimination. Bilthoven: National Institute for Public Health and the Environment; 2016. 1–8 p.

15. Ministry of health Republic of Indonesia. INFODATIN Tuberkulosis (Temukan Obati Sampai Sembuh) [Internet]. Infodatin. 2018 [cited 2019 Oct 7]. Available from: http://www.depkes. go.id/folder/view/01/structure-publikasi-pusdatin-info-datin.html

16. Ministry of health Republic of Indonesia. Regulation of health minister RI no. 67 about management of tuberculosis disease. Ministry of Health, Republic of Indonesia. 2016. 17. Dookie N, Rambaran S, Padayatchi N, Mahomed

S, Naidoo K. Evolution of drug resistance in Mycobacterium tuberculosis: A review on the molecular determinants of resistance and implications for personalized care. J Antimicrob Chemother. 2018;73(5):1138–51.

18. Palomino JC, Martin A. Drug resistance mechanisms in Mycobacterium tuberculosis. Antibiotics. 2014;3(3):317–40.

19. da Silva PEA, Palomino JC. Molecular basis and mechanisms of drug resistance in Mycobacterium tuberculosis: Classical and new drugs. J Antimicrob Chemother. 2011;66(7):1417–30. 20. Dean AS, Cox H, Zignol M. Epidemiology of

drug-resistant tuberculosis. In: Gagneux S, editor. Strain Variation in the Mycobacterium tuberculosis Complex: Its Role in Biology, Epidemiology and Control. Springer, Cham; 2017. p. 209–20.

21. Chiang CY, Schaaf HS. Management of drug-resistant tuberculosis. Int J Tuberc Lung Dis. 2010;14(6):672–82.

22. Castillo-Chavez C, Feng Z. To treat or not to treat: The case of tuberculosis. J Math Biol. 1997;35(6):629–56.

23. Fodor T, Mitchison DA. How drug resistance emerges as a result of poor compliance. Int J Tuberc Lung Dis. 1999;3(2):174.

24. Gillespie SH. Evolution of drug resistance in Mycobacterium tuberculosis: Clinical and molecular perspective. Antimicrob Agents Chemother. 2002;46(2):267–74.

(17)

25. Ekaza E, N’Guessan RK, Kacou-N’Douba A, Aka N, Kouakou J, Le Vacon F, et al. Emergence in Western African countries of MDR-TB, focus on côte d’ivoire. Biomed Res Int. 2013;2013:1–9. 26. Kempker RR, Kipiani M, Mirtskhulava V,

Tukvadze N, Magee MJ, Blumberg HM. Acquired drug resistance in mycobacterium tuberculosis and poor outcomes among patients with multidrug-resistant tuberculosis. Emerg Infect Dis. 2015;21(6):992–1001.

27. Law S, Piatek AS, Vincent C, Oxlade O, Menzies D. Emergence of drug resistance in patients with tuberculosis cared for by the Indian health-care system: a dynamic modelling study. Lancet Public Heal. 2017;2(1):e47–55.

28. Dyar OJ, Huttner B, Schouten J, Pulcini C. What is antimicrobial stewardship? Clin Microbiol Infect. 2017;23(11):793–8.

29. Dellit TH. Summary of the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Infect Dis Clin Pract. 2007;15(4):263–4.

30. Doron S, Davidson LE. Antimicrobial Stewardship. Mayo Clin Proc [Internet]. 2011;86:1113– 23. Available from: http://10.0.15.225/ mcp.2011.0358%5Cnhttps://ezp.lib.unimelb. edu.au/login?url=https://search.ebscohost.com/ login.aspx?direct=true&db=edselp&AN=S0025 619611652026&site=eds-live&scope=site 31. Padayatchi N, Mahomed S, Loveday M,

Naidoo K. Antibiotic stewardship for drug resistant tuberculosis. Expert Opinion on Pharmacotherapy. 2016;17(15):1981–3.

(18)

17 General Introduction

(19)
(20)

2

RISK FACTORS OF

MULTIDRUG-RESISTANT TUBERCULOSIS:

A GLOBAL SYSTEMATIC REVIEW

AND META-ANALYSIS

Ivan S. Pradipta

Lina D. Forsman

Judith Bruchfeld

Eelko Hak

Jan-Willem C. Alffenaar

This chapter is based on the published manuscript:

Pradipta IS, Forsman LD, Bruchfeld J, Hak E, Alffenaar J-W. Risk factors of multidrug-resistant tuberculosis: A global systematic review and meta-analysis. J Infect. 2018;77:469–78.

(21)

ABSTRACT

Objectives: Since the risk of multidrug-resistant tuberculosis (MDR-TB) may depend on the

setting, we aimed to determine the associations of risk factors of MDR-TB across different regions.

Methods: A systematic review and meta-analysis was performed with Pubmed and Embase

databases. Information was retrieved on 37 pre-defined risk factors of MDR-TB. We estimated overall Mantel-Haenszel odds ratio as a measure of the association.

Results: Factors of previous TB disease and treatment are the most important risk factors

associated with MDR-TB. There was also a trend towards increased risk of MDR-TB for patients 40 years and older, unemployed, lacking health insurance, smear positive, with non-completion and failure of TB treatment, showing adverse drug reaction, non-adherent, HIV positive, with COPD and with M. Tuberculosis Beijing infection. Effect modification by geographical area was identified for several risk factors such as male gender, married patients, urban domicile, homelessness and history of imprisonment.

Conclusions: Assessment of risk factors of MDR-TB should be conducted regionally to

develop the most effective strategy for MDR-TB control. Across all regions, factors associated with previous TB disease and treatment are essential risk factors, indicating the appropriateness of diagnosis, treatment and monitoring are an important requirements.

(22)

21 Risk factors of MDR-TB

INTRODUCTION

According to the World Health Organization (WHO), tuberculosis (TB) remains a global problem with an increasing trend of new cases of TB from 6.1 million in 2015 to 6.3 million in 2016. (1) This global health problem has further worsened in recent years due to the increase in multidrug-resistant tuberculosis (MDR-TB, M. tuberculosis resistant to rifampicin and isoniazid), with an estimated 490 000 new patients in 2016.(1) From a health economics perspective, MDR-TB is a heavy burden on health care systems with treatment costs 20 times higher than the corresponding cost of drug-susceptible TB (DS-TB).(2)

The occurrence of drug-resistant tuberculosis (DR-TB) is not only determined by timely and correct diagnosis, adequate use of anti-TB drugs, patient factors commonly associated with drug adherence (beliefs, barriers, behavior), but also determined by microbiological factors. (3) Since spontaneous resistance mutation occurs for isoniazid and rifampicin, a combination of several TB-drugs is mandatory to avoid development of drug resistance. Although the combination of antibiotics in TB treatment can prevent acquired drug resistance to some extent, problems of adverse drug reactions (ADRs), potentially leading to treatment failure, remain a challenge worldwide.(4)

In 2014 WHO formulated globally applicable programmatic management guidelines for drug-resistant tuberculosis.(5) However, several studies reported conflicting results for some risk factors of MDR-TB.(6–11) Thus, identification of the risk factors and possible effect modification by region are needed for developing optimal intervention strategies for MDR-TB control.

Four systematic reviews and meta-analyses on risk factors for MDR-TB were performed prior to our study.(12–15) The findings of these studies were limited for several reasons. Firstly, the focus of the studies was restricted to one region and the geographical effect of the risk factors from a global perspective could not be assessed. Secondly, the risk factors were analysed from a specific perspective, either host- or pathogen related. To support global strategies to target MDR-TB effectively, we therefore conducted a comprehensive systematic review and meta-analysis in predictive studies to determine risk factors for MDR-TB across different regions. These studies had five different perspectives, including host characteristics, previous TB disease and treatment, comorbidities, lifestyle and environmental characteristics, as well as microbiological aspects.

(23)

MATERIAL AND METHODS

Search strategy and selection criteria

A systematic review and meta-analysis study following PRISMA guidelines(16) was performed. The study was registered in PROSPERO, number CRD42016038014. We included experimental and observational predictive study designs, without language restrictions, in which one or multiple risk factors for MDR-TB were analysed during the study, from January 1, 2010, to March 26, 2016. We excluded cross-sectional studies, case reports, case series, review articles as well as conference abstract papers.

The study domain was restricted to adult TB-patients, 18 years and older. For cohort studies we included adult DS-TB patients as the population at risk, with MDR-TB as the outcome. We compared the risk factors of adult DS-TB and MDR-TB patients in included case-control studies. DS-TB was defined as fully sensitive of all anti-tuberculosis drugs to the

Mycobacterium tuberculosis (M.tb) in a TB patient, while MDR-TB was defined as resistance

to the line TB drugs rifampicin and isoniazid, with or without resistance to other first-line TB drugs. Microbiological verification was needed to confirm resistance type of the patients in this study.

We excluded studies restricted to specific high-risk MDR-TB patient groups, such as TB patients with HIV, prior TB treatment, neoplastic disease or diabetes mellitus. We also excluded studies that only used clinical or histopathological information for defining the type of TB without microbiological confirmation. Six perspectives of risk factors, comprising a list of 37 pre-defined variables in total, were analysed. The perspectives and risk factors were developed from a conceptual framework of pathogen-host-environment interplay in the emerging infectious disease (17) as well as previously published studies(12–15) providing potential targets for controlling MDR-TB. The definition criteria for the risk factors can be found in the Appendix Table E1.

The outcome measure was MDR-TB defined as a resistance to the first-line TB drugs rifampicin and isoniazid, with or without resistance to other first-line TB drugs. MDR-TB status was verified by microbiological test using either phenotyping drug susceptibility test or polymerase chain reaction (PCR) based on the identification of mutations linked to resistance of M.tb.

Both Pubmed and Embase databases were used to find potentially eligible articles. We developed the search term and strategy together with a medical information specialist at the Central Medical Library, University of Groningen, resulting in selecting the following

(24)

23 Risk factors of MDR-TB MeSH terms for the PubMed database and Emtree for the Embase database. Duplicate studies from the two databases were removed using the RefWorks® program. The comprehensive search terms are provided in the Appendix Table E2.

Data abstraction and assessment of quality

Two reviewers (ISP, LDF) independently screened abstracts, full-text articles, and performed bias assessments. Disagreements between the two independent reviewers (ISP, LDF) were discussed and resolved by a third reviewer (EH). The level of disagreement was calculated using a percentage of agreement and reliability, Cohen’s Kappa.(18) Data were extracted by the first reviewer (ISP) from the included articles, evaluated by the second reviewer (LDF) and final evaluation was conducted by the third reviewer (EH). We attempted to contact study authors when more data were needed; however, if the information was not received, we assumed that data were missing. We conducted a risk of bias assessment using the Risk of Bias Assessment Tool for Non-randomized studies which is compatible with the Cochrane risk of bias tool and has an acceptable validity and reliability value.(19)

Statistical analysis

A dichotomous variable was applied for each factor that was analysed. We pooled all risk factors that had a similar definition using Mantel-Haenszel Odds Ratio (mhOR) with a 95% confidence interval (95% CI). The significance threshold was set at p-value < 0·05. If data about a risk factor were only available in one study, Odds Ratio (OR) instead of mhOR was calculated. The level of heterogeneity (I2 and p-value) was calculated to identify variation

in association measures across the studies. We defined considerable heterogeneity as I2

≥ 75%(20) and/or a p-value of heterogeneity < 0·05.(21) If the data were heterogeneous, we applied a random effects model to estimate the overall effect size. Furthermore, we performed a subgroup analysis to identify sources of heterogeneity. The geographic area of the study was used for stratification in subgroup analysis. Additionally, we performed sensitivity analysis for risk factors with heterogeneous data that excluded the high potential risk of bias studies, to identify the effect size of each risk factor. Heterogeneity level and direction of the effect size among the group were considered in defining the effect estimated in the sensitivity analysis. We used Review Manager version 5.3 to analyse the effect size of the study.

RESULTS

The search process found 644 original publications from Pubmed and 764 publications from Embase. A total of 1,056 abstracts were screened after duplications were removed, and 1,036 articles were excluded for several reasons (Figure 1). There were 47 discrepancies between the two independent reviewers in the title-abstract screening. The level of agreement was 96% (good), and the reliability according to Cohen’s Kappa was 0·78

(25)

(good). Furthermore, the disagreement arose in seven out of the 117 articles in the full-text screening, with a level of agreement of 94% (good), and reliability according to Cohen’s kappa was 0·84 (good). We found 20 studies fulfilling the inclusion criteria from the following continents; Asia (14), Africa (2), North America (1), South America (2) and Europe (1). The total number of patients included was 20 017, among which 1814 were MDR-TB patients and 18 203 DS-TB patients. Study characteristics are shown in Table 1.

(26)

25 Risk factors of MDR-TB Ta b le 1 . C h ar ac te ri sti cs o f t h e s tu d ie s in cl u d ed in t h e s ys te m ati c re vi ew a n d m et a-an al ys is . Au th o r (y ea r p u b lic at io n) C o u nt ry St u dy de si gn St u dy p er io d C as e (M D R -T B ) C o n tr o l (D S -T B ) R is k f ac to rs i d en ti fi ed A h m ad e t a l. ( 2 0 1 2) (2 2) P aki st an C as e-co n tro l 2 000 -2 00 2 50 75 M ar it al s ta tu s, g en d er , n o n -B C G v ac cin ati o n , p re vi o u s tre at m en t, s m o kin g, k n o w n c o n ta ct w it h T B p ati en t A n d re w e t a l. ( 2 0 1 0 )( 2 3 ) S o u th Afr ic a C as e-co n tro l 20 0 5 -20 0 7 1 23 11 6 G en d er , n o n -c o m p le ti o n a n d f ai lu re o f T B t re at m en t, H IV B ag h ae i e t a l. ( 2 0 0 9 )( 2 4 ) Ir an C as e-co n tro l 20 0 2 -20 0 5 48 23 4 G en d er , p re vi o u s t re at m en t, s m ea r p o si ti vi ty , s m o kin g, kn o w n c o n ta ct w it h T B p ati en t B al aj i e t a l ( 2 0 1 0 )( 2 5) In d ia C as e-co n tro l 20 0 2 -20 0 7 30 11 7 G en d er , l u n g c av it y, H IV C h u ch ot ta w o rn e t a l. ( 2 0 1 5) (8 ) T hai la n d C as e-co n tro l 20 0 7-20 1 3 14 5 14 5 A ge 4 0 y ea rs a n d o ld er , g en d er , n o n -c o m p le ti o n a n d fa ilu re o f T B t re at m en t, C V D , D M , H IV , l o w B M I D e S o uz a e t a l. ( 2 0 0 6 )( 2 6 ) B ra zi l C as e-co n tro l 2 000 -2 00 4 12 36 E m p lo ym en t s ta tu s, g en d er , p re vi o u s t re at m en t, s m ea r p o si ti vi ty , D M , H IV , k n o w n c o n ta ct w it h T B p ati en t, d ai ly al coh o l c on su mp ti on D ia n d e e t a l. ( 2 0 0 9 )( 1 0 ) B urki n a Fa so C as e-co n tro l 20 0 5 -20 0 6 56 30 4 A ge 4 0 y ea rs a n d o ld er , e m p lo ym en t s ta tu s, g en d er , p re vi o u s t re at m en t, H IV , k n o w n c o n ta ct w it h T B p ati en t, d ai ly a lco h o l co n su m p tio n E l S ah ly e t a l. ( 2 0 0 6 )( 2 7 ) The U n it ed St ate s C as e-co n tro l 19 9 5 -2 0 0 1 15 19 7 7 M ar it al s ta tu s, g en d er , p re vi o u s T B d is ea se , C O P D , H IV , h is to ry o f im p ri so n m en t, h is to ry h o m el es s, d ai ly a lc o h o l con su mp ti on E lm i e t a l. ( 2 0 1 5) (9 ) M al ay si a C as e-co n tro l 2 0 1 0 -2 0 1 4 10 5 2 09 G en d er , p re vi o u s t re at m en t, C O P D , M an to u x t es t p o si ti vi ty , l u n g c av it y, H IV , h is to ry o f im p ri so n m en t, sm o kin g, h is to ry o f h o m el es s, k n o w n c o n ta ct w it h T B p ati en t, d ai ly a lc o h o l c o n su m p ti o n F er ro e t a l ( 2 0 11 )( 2 8 ) Co lo m b ia C as e-co n tro l 20 0 7-20 0 8 76 84 M . t b B ei jin g g en ot yp e s tr ain F o x e t a l. ( 2 0 11 )( 7 ) Isra el C as e-co n tro l 20 0 2 -20 0 9 44 50 8 G en d er , D O T, n o n -c o m p le ti o n a n d f ai lu re o f T B tre at m en t, p re vi o u s T B d is ea se , h ep ati ti s, l u n g c av it y, H IV G ao e t a l ( 2 0 1 6 )( 2 9 ) C hi na C oh or t 20 0 8 -20 1 0 17 1 6 0 9 A ge 4 0 y ea rs a n d o ld er , g en d er , A D R s, p re vi o u s tre at m en t, l o w B M I H an g e t a l. ( 2 0 1 3 )( 6 ) V ietn am C as e-co n tro l 20 0 7-20 0 9 22 29 8 G en d er , H IV s m o kin g, M . t b B ei jin g g en ot yp e s tr ain

2

(27)

lic at io n) C o u nt ry St u dy de si gn St u dy p er io d C as e (M D R -T B ) C o n tr o l (D S -T B ) R is k f ac to rs i d en ti fi ed 0 11 )( 3 0 ) C hi na C as e-co n tro l 20 0 7-20 0 9 10 0 97 N o n -c o ve ra ge h ea lt h in su ra n ce , g en d er , p re vi o u s tre at m en t, l u n g c av it y, k n o w n c o n ta ct w it h T B p ati en t, M . tb B ei jin g g en ot yp e s tr ain ri f e t a l. ( 2 0 1 6 ) M al ay si a C as e-co n tro l 2 0 1 3 -2 0 1 4 30 120 M ar it al s ta tu s, g en d er , n o n -a d h ere n ce , h ig h er e d u ca ti o n , p re vi o u s t re at m en t, s m ea r p o si ti ve , D M , H IV , h is to ry o f im p ri so n m en t, s m o kin g, u rb an a re a, k n o w n c o n ta ct w it h T B p ati en t, d ai ly a lc o h o l c o n su m p ti o n 2 0 1 4 )( 3 2) Isra el C as e-co n tro l 1999 -2 0 0 0 2 07 31 0 7 G en d er , p re vi o u s t re at m en t, s m ea r p o si ti vi ty , H IV , h is to ry o f i mp ri sonm en t t a l. ( 2 0 0 8 )( 3 3 ) En gla n d C as e-co n tro l 19 8 2 -2 0 0 4 42 84 G en d er , p re vi o u s T B d is ea se , s m ea r p o si ti vi ty , k n o w n co n ta ct w it h T B p ati en t 2 0 0 9 )( 3 4 ) C hi na C as e-co n tro l 2 000 -2 00 6 333 7 0 1 8 G en d er , p re vi o u s t re at m en t, s m ea r p o si ti vi ty , l u n g c av it y, u rb an a re a l. ( 2 0 11 )( 11 ) In d ia C as e-co n tro l 2 0 09 1 8 4 56 G en d er , p re vi o u s T B d is ea se , l u n g c av it y, H IV 2 0 1 2) (3 5) C hi na C as e-co n tro l 20 0 4 -20 0 5 1 75 2 0 09 A ge 4 0 y ea rs a n d o ld er , g en d er , p re vi o u s T B t re at m en t n : M D R -T B : m ul ti d ru g-re si st an t tu b er cul o si s; D S -T B : d ru g-su sc ep ti b le tu b er cul o si s; B C G : B ac ill e C alm et te -G u ér in ; H IV : h u m an im m u n o d efi ci en cy : c ard io va sc ul ar ; D M : d ia b et es m el lit u s; B M I: b o d y m as s in d ex ; C O P D : c h ro n ic o b st ru cti ve p ulm o n ar y d is ea se ; M .t b : M yc o b ac te ri u m t u b er cul o si s; y o b se rv ed t re at m en t; A D R s: a d ve rs e d ru g re ac ti o n s. n ti n u ed ). C h ar ac te ri sti cs o f t h e s tu d ie s in cl u d ed in t h e s ys te m ati c re vi ew a n d m et a-an al ys is .

(28)

27 Risk factors of MDR-TB Potential bias was analysed for the 20 included studies. Thirty percent of all included studies displayed a high potential for bias in the measurement of exposure. Interview bias, recall bias, self-reported data, and unclear definition of the exposure were identified as common sources of bias. However, the overall risk of bias was low (see Appendix Fig. E1,

E2). Not all pre-defined perspectives could be analysed due to lack of availability of the

data in the included articles. We were not able to analyse risk factors from a health services perspective. Therefore five of the six different perspectives of risk factors, comprising 29 specific factors from the included studies, were analysed in this study. Additional data were received upon request for one study.(6) We identified significant risk factors of MDR-TB (p<0.05) from four perspectives, namely patient characteristics (i.e. unemployed, lacking health insurance coverage, smear positive, mantoux test positive and lung cavity), TB history and treatment (i.e. previous TB disease, previous TB treatment, non-completion and failure of TB treatment, adverse drug reaction, non-BCG vaccination, non-adherence), comorbidity (i.e. Chronic Obstructive Pulmonary Disease, COPD) and strain (i.e. M.tb Beijing strain). However, several risk factors of MDR-TB showed heterogeneous results (I2 ≥ 75% or p-value

heterogeneity < 0·05), i.e. age 40 years and older, male gender, married patients, lung cavity, previous TB disease, previous TB treatment, HIV, known contact with TB patients, low BMI, urban domicile, homelessness, and history of imprisonment. The pooled effect estimated for all risk factors can be found in Table 2.

Subgroup analysis was performed for factors with heterogeneous results to identify the effect of geographical area. When stratifying patients by setting, homogenous results appeared within subgroups for variables of gender, marital status, previous TB disease, domicile area, nature of abode, and history of imprisonment status (Fig. 2, 3), while heterogeneous results appeared within subgroups for variables of age, BMI, status of lung cavity, previous TB treatment, HIV and known contact with TB patients (see Appendix

Figure E3, E4). Subgroup analysis indicated variations dependent on setting for several risk

factors of MDR-TB, such as male gender, married patients, urban domicile, homelessness, having a previous TB disease and a history of imprisonment. For example, pooled effect estimates of studies in America (Brazil and USA)(26,27) showed female patients and unmarried patients were more likely to be diagnosed with MDR-TB than DS-TB. On the contrary, effect estimates from studies in Western Asia (Iran and Israel)(7,24,32) revealed that males were more prone to MDR-TB and marital status was not a risk factor for MDR-TB in Asia (Fig. 2A, 2B).(22,31) Likewise, studies from North America(27) described a protective effect of MDR-TB for subjects who had a history of imprisonment, whereas several Asian studies failed to prove any association with history of imprisonment and MDR-TB (Fig. 3C). (9,31,32)

(29)

ec t e stim at es f o r r is k f ac to rs o f M ul ti d ru g-R es is ta n t T u b er cul o si s (M D R -T B) rs N u m b er o f St u d ie s P ar ti ci p ant s E ff ec t E st im at ed O d d s R at io ( 9 5 % C I) H et er og en ei ty I 2 (p -v alu e) ra ct er is ti cs rs a n d o ld er 4( 8 ,1 0, 2 9, 3 5 ) 4 4 60 1· 3 4 ( 0 .7 5 -2 .3 9 ) 76% (0 ·0 0 6) † er 19 (6 –1 0 ,2 2– 2 7,2 9 –3 6 ) 19 8 5 6 1· 0 7 ( 0 ·8 5 -1 .3 6 ) 6 7 % ( < 0 ·0 0 1 ) † u ca ti o n 1 (3 1 ) 15 0 1· 6 9 ( 0 ·7 3 -3 ·8 7 ) n /a en t 2 (1 0 ,2 6 ) 4 08 3 ·0 0 ( 1· 6 9 -5 ·3 0 ) *† 6 9 % ( 0 ·0 7 ) lt h in su ra n ce c o ve ra ge 1 (3 0 ) 19 7 1· 9 9 (1· 1 2 -3 ·5 4 ) † n /a ati en t 3 (2 2 ,2 7, 3 1 ) 2 2 67 0 ·6 4 (0 ·1 3 -3 ·1 1 ) 8 7 % ( < 0 ·0 0 1 ) † ti ve 6 (2 4 ,2 6 ,3 1– 3 4 ) 11 1 61 1· 7 2 ( 1· 4 0 -2 ·1 2) *† 41 % ( 0 ·1 3 ) es t p o si ti ve 1 (9 ) 10 3 3 ·3 8 ( 1· 4 5 -7 ·8 9 ) † n /a y 7 (7 –9 ,2 5 ,3 0 ,3 4 ,3 6 ) 8 82 5 1· 9 2 ( 1· 0 2 -3 ·6 2) † 8 9 % ( < 0 ·0 0 1 ) † re at m en t f p re vi o u s T B d is ea se 4 (7 ,2 7, 33 ,36 ) 2 9 07 4 ·42 (1 ·4 6 -1 3 ·3 7 ) † 8 6 % ( < 0 ·0 0 1 ) † f p re vi o u s T B t re at m en t 11 (9 ,1 0 ,2 2 ,2 4 ,2 6 ,2 9 –3 2 ,3 4 ,3 5 ) 1 5 6 57 7· 2 4 ( 4 ·0 6 -1 2 ·8 9 ) † 8 8 % ( < 0 ·0 0 1 ) † le ti o n a n d f ai lu re o f T B t re at m en t ‡ 3 (7, 8 ,2 3 ) 13 5 4 5 ·6 0 ( 3 ·3 6 -9 ·3 2) *† 0 % ; ( 0 ·3 7 ) am 1 (7 ) 5 52 1· 3 6 ( 0 ·4 7-3 ·9 5) n /a f a d ve rs e D ru g R ea cti o n 1 (2 9 ) 5 52 2 ·3 1 (1 ·1 4-4 ·6 9 ) † n /a ac cin ati o n 1 (2 2 ) 125 2 ·7 9 ( 1· 1 3 -6 ·8 5) † n /a en ce 1 (3 1 ) 15 0 4 ·5 0 ( 1· 7 1-11 ·8 2) † n /a c om or bi di ty ve 11 (6 –1 0 ,2 3 ,2 5 ,2 7,3 1 ,3 2 ,3 6 ) 10 7 3 6 1· 4 9 ( 0 ·7 3 -3 ·0 6 ) 8 1 % ( < 0 ·0 0 1 ) † elli tu s 4 (8 ,9, 2 6 ,3 1 ) 8 02 1· 3 0 (0 ·9 1-1· 8 6 ) * 4 4% ( 0 ·1 5) la r d ise ase 1 (8 ) 29 0 0 ·7 5 ( 0 ·3 6 -1 ·5 8 ) n /a 2 (9, 2 7 ) 23 0 6 2 ·5 3 ( 1· 0 5 -6 ·1 4 ) *† 4 0 % ( 0 ·2 0 ) 1 (7 ) 5 52 0 ·4 2 (0 ·1 3 -1 ·40 ) n /a

(30)

29 Risk factors of MDR-TB R isk fa ct o rs N u m b er o f St u d ie s P ar ti ci p ant s E ff ec t E st im at ed O d d s R at io ( 9 5 % C I) H et er og en ei ty I 2 (p -v alu e) Li fe s ty le & E nvi ro nm en ta l K n o w n c o n ta ct w it h T B p ati en t 8 (9 ,1 0 ,2 2 ,2 4 ,2 6 ,3 0 ,3 1 ,3 3 ) 14 5 3 1· 3 0 ( 0 ·7 4 -2 ·2 9 ) 6 7 % ( 0 ·0 0 4 ) † Sm o ke r 5 (6 ,9 ,2 2 ,24 ,3 1 ) 11 8 9 0 ·9 0 ( 0 ·6 6 -1 ·2 2) * 2 1 % ( 0 ·2 8 ) Lo w B M I* * 2(8 ,2 9 ) 1 8 65 0 ·8 6 ( 0 ·1 7-4 ·2 7 ) 8 2 % ( 0 ·0 2) † U rb an dom ic ile 2 (3 1 ,3 4 ) 75 0 1 0 ·8 8 ( 0 ·2 0 -3 ·8 9 ) 9 1 % ( < 0 ·0 0 1 ) † D ai ly a lco h o l co n su m p tio n 5 (9 ,1 0 ,26 ,27 ,3 1 ) 27 2 0 0 ·8 0 ( 0 ·4 9 -1 ·3 0 ) * 4 9 % ( 0 ·1 0 ) H o m el ess n ess 2 (9, 2 7 ) 23 0 6 2 ·7 3 ( 0 ·1 8 -4 0 ·9 5) 8 7 % ( 0 ·0 0 6 ) † H is tor y o f i mp ri sonm en t 4 (9 ,2 7,3 1 ,3 2 ) 57 7 0 0 ·8 6 ( 0 ·2 7-2 ·7 8 ) 6 3 % ( 0 ·0 4 ) † St ra in B ei jin g s tr ain 3 (6 ,2 8 ,3 0 ) 6 65 5 ·5 8 ( 1· 6 6 -1 8 ·7 6 ) *† 6 6 % ( 0 ·0 5) In fo rm ati o n : * F ix ed e ff ec t m o d el ; †Si gn ifi ca n t va lu e (p < 0 .0 5) ; ‡in cl u d in g n o n -c u re , n o n -c o m p le ti o n , d ef aul t an d fa ilu re tre at m en t; ** B o d y M as s In d ex (B M I) < 1 8 k g /m 2; n /a : n ot a p p lic ab le ; C O P D : C h ro n ic o b st ru cti ve p ulm o n ar y d is ea se ; H IV : H u m an I m m u n o d efi ci en cy V ir u s; D O T: D ire ct O b se rv ed T re at m en t. Ta b le 2 ( C o n ti n u ed ). E ff ec t e stim at es f o r r is k f ac to rs o f M ul ti d ru g-R es is ta n t T u b er cul o si s (M D R -T B)

2

(31)

A. Gender (female vs. male)

Figure 2. Homogeneous effect estimated within the subgroup of gender, marital status and

previ-ous tuberculosis disease, stratified by area of study. Notes: reference group in each of factors: (A) female, (B) unmarried (C) non-previous TB disease

(32)

31 Risk factors of MDR-TB

B. Marital status (unmarried vs. married)

C. Previous TB disease (non-previous TB disease vs. previous TB disease)

Figure 2 (Continued). Homogeneous effect estimated within the subgroup of gender, marital status

and previous tuberculosis disease, stratified by area of study. Notes: reference group in each of factors: (A) female, (B) unmarried (C) non-previous TB disease

(33)

A. Domicile area (rural vs. urban)

B. Nature of abode (non-homelessness vs. homelessness)

(34)

33 Risk factors of MDR-TB Regarding variables of previous TB disease status and domicile area, we analysed that having a previous TB disease remained a significant risk factor of MDR-TB in the pooled estimate (p< 0·001; OR 4·42; 95%CI 1·46-13·37). Although risk factors of previous TB disease showed heterogeneous result (I2: 86%), the forest plot described the same directions

for a risk factor of MDR-TB in the all subgroups of variable previous TB disease (Fig. 2C). On the contrary, the risk factor of urban domicile differed significantly depending on the setting, where a Malaysian study indicated a protective effect of urban dwelling (p=0·03; OR 0·39; 95%CI 0·16-0·93) whereas a study in China showed an increased risk (p= 0·001; OR 1·77; 95%CI 1·42-2·21) (Fig. 3A).

Since heterogeneity in several variables, such as age, BMI and status of lung cavity, previous TB treatment, HIV, known contact with TB remained high (see Supplementary Fig. E3,

E4), we therefore conducted a sensitivity analysis of these variables by excluded studies

with high risk of bias. The studies that were exluded in the sensitivity analysis, i.e. studies assessing age (three studies(8,10,35)), lung cavity (five studies(8,9,25,34,36)), previous TB treatment(eight studies(9,10,22,24,26,31,34,35)), HIV (six studies(8–10,25,27,31)), known contact with TB (six studies(9,10,22,24,26,31)) and BMI (one study(8)). The sensitivity analysis showed being HIV positive, previous TB treatment and age 40 years and older to be risk factors of MDR-TB (Table 3). However, the variables ‘previous TB treatment’ and ‘lung cavity status’ displayed a heterogenous association with the risk of MDR-TB and should therefore be interpreted carefully. Regarding previous TB treatment, despite heterogeneity all effect estimates of the studies were of the same nature as risk factors of MDR-TB, while the presence of lung cavity cannot be interpreted as a risk factor for MDR-TB since effect estimates across studies showed conflicting results (Appendix Fig. E4B).

DISCUSSION

We identified an effect modification by geographic area for several risk factors of MDR-TB, such as male gender, married patient, urban domicile, homelessness and having a history of imprisonment. Our results confirm prior reviews that having a previous TB disease and treatment are the most influential risk factors for developing MDR-TB, independent of the setting. Furthermore, patients 40 years and older, lacking health insurance, unemployed, non-adherent, ADRs, with a history of non-completion or failure of TB treatment, without BCG vaccination, HIV positive, with COPD, with infection with M. tb Beijing strain, smear and mantoux test positivite, show significant risk factors for developing MDR-TB. On the contrary, other risk factors identified in prior studies, such as, low education status, non-Directly Observed Treatment (DOT), diabetes mellitus, cardiovascular diseases, hepatitis, known contact with TB patients, smoking, low BMI and daily alcohol intake, did not show a clear association with MDR-TB in our study.

(35)

Table 3. Sensitivity analysis of heterogeneous’ factors

No Risk factors Pre-sensitivity analysis Post-sensitivity analysis Number of Studies Odd Ratio (95% CI) I2 (p-value) Number of Studies Odd Ratio (95% CI) I2 (p-value) 1 Age 40 years and older 4 1·34 (0·75-2·39) 76% ( 0·006)† 1 14·18 (1·88-107·18) † n/a 2 Lung cavity 7 1·92 (1·02-3·62)† 89% (< 0·001) † 2 1·10 (0·40-3·02) 82% (0·02) † 3 Presence of previous TB treatment 11 7·24 (4·06-12·89) † 88% (< 0·001) † 3 5·38 (1·67-13·37)† 80% (0·007)† 4 HIV positive 11 1·49 (0·73-3·06) 81% (< 0·001) † 5 3·04 (1·60-5·77) † 55% (0·08) 5 Known contact with TB patient 8 1·30 (0·74-2·29) 67% (0·004) † 2 0·80 (0·22-2·85) 58% (0·12) 6 Low BMI 2 0·86 (0·17-4·27) 82% (0·02) † 1 0·34 (0·10-1·19) n/a

Information : I2: heterogeneity; Significant value; Low body mass index (BMI) : BMI < 18 kg/m2; 95%CI

: 95% confidence interval; HIV: Human Immunodeficiency Virus; TB: tuberculosis

In terms of microbiological aspect, our study was supported by other studies. Beijing M. tb strains are more likely to be MDR-TB than non-Beijing M. tb strains, according to studies from Indonesia,(37) Vietnam,(38) and Russia,(39) linking the M. tb Beijing genotype strain with a history of previous TB treatment and treatment failure. Animal studies have shown Beijing M. tb strains to be more virulent with more extensive tissue destruction, rapid outgrowth, and increased mortality.(40) Suggested hypotheses for this association regard differences in cell wall structure, leading to suboptimal intracellular drug concentrations, as well as a higher virulence per se, resulting in longer persistent infection.(41)

Regarding comorbidities, it is a matter of debate whether HIV is a risk factor for MDR-TB. A previous systematic review showed no association between HIV and primary or secondary MDR-TB.(42) However, our study indicated that HIV is a risk factor for MDR-TB after sensitivity analysis was performed. This can be explained by both immune status and drug-related factors. Immunosuppression can lead to reactivation of latent TB, increased risk of re-infection recurrence due to new M.tb infection and rapid progression to active TB.(43) Furthermore, problems relating to drug interactions, overlapping drug toxicities, high pill burden, drug malabsorption and immune reconstitution inflammatory syndrome (IRIS) can potentially lead to the development of drug resistance and therapeutic failure in co-infected TB-HIV patients.(44) Hence, there is biological plausibility for HIV being a risk

(36)

35 Risk factors of MDR-TB Another comorbidity, COPD, has also been discussed as a risk factor of MDR-TB. A prospective study of pulmonary tuberculosis (PTB) patients aged ≥ 40 years with concomitant COPD had an increased risk of developing MDR-TB. (45) There is also evidence of an inverse relationship; TB patients can develop COPD as a result of long-term damage of structural and functional of the lung.(46,47) In our study, we analysed two case-control studies from Malaysia and USA, with 120 MDR-TB patients as cases and 2,186 DS-TB patients as controls. Our study indicated that COPD patients were more likely to have MDR-TB than patients without COPD, with a pooled estimate 2.5 times higher for COPD patients than non-COPD patients.

Our study demonstrated that failed TB treatment is a considerable risk factor for MDR-TB. Although non-adherence to treatment is believed to be a cause of treatment failure in TB patients, a pre-clinical study showed that non-adherence alone was not sufficient for the development of MDR-TB, but in-between patient pharmacokinetic variability was necessary.(48) Similarly, a meta-analysis identified pharmacokinetic variability of isoniazid to be associated with therapeutic failure and acquired drug resistance.(49) Another meta-analysis analysed genetic factors such as rate of acetylation, where patients who have rapid acetylation of isoniazid were more likely to have microbial failure, acquire drug resistance and relapse than patients with slow acetylation.(49) On the other hand, patients with a slow isoniazid acetylation profile were more prone to hepatotoxicity than patients with a rapid acetylation profile.(50) It is apparent that pharmacogenetics variation plays an important role in therapeutic response and ADRs, besides inter-individual variability of pharmacokinetics profile.(50)

Our study corroborated the results of prior meta-analyses showing that previous TB disease and treatment were essential risk factors of MDR-TB, while alcohol abuse and low education were not.(12–14)Moreover, meta-analyses in China pointed out pulmonary cavity and living in rural area as risk factors of MDR-TB,(12,13) while studies in Europe showed that male gender, homelessness and urban domicile to be risk factors of MDR-TB.(14) As described in the aforementioned studies, the impact of risk factors can differ according to geographical area.

Our study suggests that identifying risk factors of MDR-TB regionally is important in developing strategies for MDR-TB control as a result of regional differences in the risk factors due to variation of healthcare quality, socio-behavioral and poor living conditions. Since unemployment and lack of health insurance coverage are risk factors of MDR-TB in our study, government support is crucial to organise universal health coverage that will cover not only drug cost but also diagnosis, treatment and monitoring for TB patients. In addition, enhancing access to health facilities and laboratories, including qualified drug

(37)

susceptible tests, are required for appropriate diagnosis and treatment as well as for correct surveillance of the MDR-TB epidemic.

The fact that we identified non-adherence, previous and failed TB treatment as considerable risk factors of MDR-TB in our study, indicates that variation of adherence, pharmacokinetics and pharmacogenetics profile among TB patients is a factor that should be considered to avoid development of MDR-TB. Antibiotic stewardship program for drug-resistant tuberculosis is required to be established at an institution level, specifically in high-burden areas of TB. The collaborative team should include physicians, pharmacists, microbiologists, nurses and administrators, all with a common goal to improve diagnosis, treatment and monitoring of TB patients. Personalised treatment could be a promising approach for controlling MDR-TB, especially in patients at high risk of MDR-TB. Therapeutic drug monitoring and intervention with individual non-adherence can be implemented as a program to achieve treatment success.(51) However, since personalised treatment needs advanced resources, free consultation of TB experts should be widely available for health care providers to make rapid decisions on the management of complex TB cases, particularly in an area with limited resources.

There are several limitations in our study. Firstly, most of our included studies were case-control studies where recall bias may have occurred. Secondly, not all countries and included risk factors could be assessed due to unavailability of data. Thirdly, since the majority of studies were predictive studies, the causality of risk factors and outcome should be explored further using an appropriate study design. Finally, the power of the study was low for some risk factors of MDR-TB, such as non-BCG vaccination and positive Mantoux test. We noticed a potential information bias due to missing data in the only included study which analysed positive Mantoux test as a risk factor for MDR-TB. The study showed a high proportion of participants who had unknown information of Mantoux test results in the MDR-TB group (58.8%). The multivariate analysis indicated that Mantoux test positivity and non-BCG vaccine status were not significant risk factors for MDR-TB. (p≥0·05).(9,22) Hence, there is no clear support of an association of Mantoux test and BCG vaccination with MDR-TB. On the other hand, we performed a thorough full-text screening, excluding studies with a high level of bias in the sensitivity analysis. We also assessed statistical heterogeneity and biological plausibility from the current evidence. Furthermore, we attempted to contact study authors to obtain more comprehensive data in our study.

In conclusion, factors of previous TB disease and treatment are the major risk factors for MDR-TB across all settings. Subsequently, we identified patients age 40 years and older,

(38)

37 Risk factors of MDR-TB COPD and with M. Tuberculosis Beijing infection who should be carefully monitored during their TB treatment to avoid development of MDR-TB. Equally important, risk factors of MDR-TB related to male gender, married patient, urban domicile, homelessness and having a history of imprisonment can vary depending on the setting. Therefore, assessment of risk factors of MDR-TB should be conducted regionally to develop the most effective strategy for MDR-TB control.

Funding

This work was supported by Indonesia Endowment Fund for Education or LPDP in the form of a Ph.D. scholarship to ISP.

Acknowledgment

We thank Prof. Naoto Keicho, The Research Institute of Tuberculosis and Japan Anti-tuberculosis Association, and Nguyen Thi Le Hang, MD, PhD for providing additional information. We also thank Brian Davies for language correction.

Conflict of interests

ISP, LDF, JB, EH and JWA have no competing financial or non-financial interests in this work

(39)

REFERENCES

1. WHO. WHO Global tuberculosis report 2017 [Internet]. World Health Organization Press. 2017. Available from: http://www.who.int/tb/ publications/global_report/en/

2. WHO. WHO | Global tuberculosis report 2016 [Internet]. WHO 2016. 2015. Available from: http://www.who.int/tb/publications/global_ report/en/

3. Muller B, Borrell S, Rose G, Gagneux S. The heterogeneous evolution of multidrug-resistant Mycobacterium tuberculosis. Trends Genet. 2013;29(3):160–9.

4. Singh A, Prasad R, Balasubramanian V, Gupta N, Gupta P. Prevalence of adverse drug reaction with first-line drugs among patients treated for pulmonary tuberculosis. Clin Epidemiol Glob Heal. 2015;3:S80–90.

5. WHO. Companion Handbook to the WHO Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis. Companion Handbook to the WHO Guidelines for the Programmatic Management of Drug-Resistant Tuberculosis. 2014. 1–20 p.

6. Hang NT Le, Maeda S, Lien LT, Thuong PH, Van Hung N, Thuy TB, et al. Primary Drug-Resistant Tuberculosis in Hanoi, Viet Nam: Present Status and Risk Factors. PLoS One. 2013;8(8):1–9. 7. Fox L, Kramer MR, Haim I, Priess R, Metvachuk A,

Shitrit D. Comparison of isoniazid monoresistant tuberculosis with drug-Susceptible tuberculosis and multidrug-Resistant tuberculosis. Eur J Clin Microbiol Infect Dis. 2011;30(7):863–7. 8. Chuchottaworn C, Thanachartwet V, Sangsayunh

P, Than TZM, Sahassananda D, Surabotsophon M, et al. Risk factors for multidrug-resistant tuberculosis among patients with pulmonary tuberculosis at the central chest institute of Thailand. PLoS One. 2015;10(10):1–17. 9. Elmi OS, Hasan H, Abdullah S, Mat Jeab MZ,

Bin Alwi Z, Naing NN. Multidrug-resistant tuberculosis and risk factors associated with its development: a retrospective study. J Infect Dev Ctries. 2015;9(10):1076–85.

10. Diandé S, Sangaré L, Kouanda S, Dingtoumda BI, Mourfou A, Ouédraogo F, et al. Risk Factors for Multidrug-Resistant Tuberculosis in Four Centers in Burkina Faso, West Africa. Microb Drug Resist. 2009;15(3):217–21.

11. Vadwai V, Shetty A, Soman R, Rodrigues C. Determination of risk factors for isoniazid monoresistance and multidrug-resistant

12. Zhao P, Li XJ, Zhang SF, Wang XS, Liu CY. Social behaviour risk factors for drug resistant tuberculosis in mainland China: a meta-analysis. J Int Med Res. 2012;40(2):436–45.

13. Cai X, Zhang D, Yan Y, Tan D, Xu Y. Meta-analysis on risk factors of multidrug resitant tuberculosis in China. Chinese J Epidemiol. 2015;36(12):1424–9.

14. Faustini A, Hall AJ, Perucci CA. Risk factors for multidrug resistant tuberculosis in Europe: a systematic review. Thorax. 2006;61:158–63. 15. Lukoye D, Ssengooba W, Musisi K, Kasule

GW, Cobelens FGJ, Joloba M, et al. Variation and risk factors of drug resistant tuberculosis in sub-Saharan Africa: a systematic review and meta-analysis. BioMed Cent [Internet]. 2015;15(291):1/13. Available from: http://www. biomedcentral.com/1471-2458/15/1/ 16. Moher D, Liberati A, Tetzlaff J, Altman DG.

Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med [Internet]. 2009 Jul 21 [cited 2016 Aug 27];6(7):e1000097. Available from: http:// dx.doi.org/10.1371/journal.pmed.1000097 17. Engering A, Hogerwerf L, Slingenbergh J.

Pathogen-host-environment interplay and disease emergence. Emerging Microbes and Infections. 2013.

18. Sim J, Wright CC. The kappa statistic in reliability studies: use, interpretation, and sample size requirements. Phys Ther. 2005;85(3):257–68. 19. Kim SY, Park JE, Lee YJ, Seo HJ, Sheen SS,

Hahn S, et al. Testing a tool for assessing the risk of bias for nonrandomized studies showed moderate reliability and promising validity. J Clin Epidemiol. 2013;66(4):408–14.

20. Higgins J, Green S. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. Cochrane Collab [Internet]. 2011;0(March):10–1. Available from: http://www.cochrane.org/training/cochrane-handbook

21. Sedgwick P. Meta-analyses: heterogeneity and subgroup analysis. Bmj [Internet]. 2013;346(June):1–3. Available from: http:// www.bmj.com/cgi/doi/10.1136/bmj.f4040 22. Ahmad AM, Akhtar S, Hasan R, Khan JA, Hussain

SF, Rizvi N. Risk factors for multidrug-resistant tuberculosis in urban Pakistan: A multicenter case-control study. Int J Mycobacteriology [Internet]. 2012;1(3):137–42. Available from:

(40)

39 Risk factors of MDR-TB

23. Andrews JR, Shah NS, Weissman D, Moll AP, Friedland G, Gandhi NR. Predictors of multidrug-and extensively drug-resistant tuberculosis in a high HIV prevalence community. PLoS One. 2010;5(12):1–6.

24. Baghaei P, Tabarsi P, Chitsaz E, Novin A, Alipanah N, Kazempour M, et al. Risk factors associated with multidrug-resistant tuberculosis. Tanaffos. 2009;8(3):17–21.

25. Balaji V, Daley P, Anand AA, Sudarsanam T, Michael JS, Sahni RD, et al. Risk factors for MDR and XDR-TB in a tertiary referral hospital in India. PLoS One. 2010;5(3):1–6.

26. de Souza M, Antunes C, Garcia G. Multidrug-resistant Mycobacterium tuberculosis at a referral center for infectious diseases in the state of Minas Gerais, Brazil: sensitivity profile and related risk factors. J Bras Pneumol. 2006;32(5):430–7.

27. El Sahly HM, Teeter LD, Pawlak RR, Musser JM, Graviss EA. Drug-resistant tuberculosis: A disease of target populations in Houston, Texas. J Infect. 2006;53(1):5–11.

28. Ferro BE, Nieto LM, Rozo JC, Forero L, van Soolingen D. Multidrug-resistant Mycobacterium tuberculosis, southwestern Colombia. Emerg Infect Dis. 2011;17(7):1259–62.

29. Gao J, Ma Y, Du J, Zhu G, Tan S, Fu Y, et al. Later emergence of acquired drug resistance and its effect on treatment outcome in patients treated with Standard Short-Course Chemotherapy for tuberculosis. BMC Pulm Med [Internet]. 2016;16:26. Available from: http://www. pubmedcentral.nih.gov/articlerender.fcgi?artid= 474333 0&tool=pmcentrez&render t ype= abstract

30. He GX, Wang HY, Borgdorff MW, van Soolingen D, van der Werf MJ, Liu ZM, et al. Multidrug-resistant Tuberculosis, People’s Republic of China, 2007-2009. Emerg Infect Dis. 2011;17(10):1831–8.

31. Mohd Shariff N, Shah SA, Kamaludin F. Previous treatment, sputum-smear nonconversion, and suburban living: The risk factors of multidrug-resistant tuberculosis among Malaysians. Int J Mycobacteriology [Internet]. 2016;5(1):51–8. Available from: http://dx.doi.org/10.1016/j. ijmyco.2015.11.001

32. Mor Z, Goldblatt D, Kaidar-Shwartz H, Cedar N, Rorman E, Chemtob D. Drug-resistant tuberculosis in Israel: Risk factors and treatment outcomes. Int J Tuberc Lung Dis. 2014;18(10):1195–201.

33. O’Riordan P, Schwab U, Logan S, Cooke G, Wilkinson RJ, Davidson RN, et al. Rapid molecular detection of rifampicin resistance facilitates early diagnosis and treatment of multi-drug resistant tuberculosis: Case control study. PLoS One. 2008;3(9):1–7.

34. Shen X, DeRiemer K, Yuan ZA, Shen M, Xia Z, Gui X, et al. Drug-resistant tuberculosis in Shanghai, China, 2000-2006: Prevalence, trends and risk factors. Int J Tuberc Lung Dis. 2009;13(2):253–9. 35. Zhao Y, Xu S, Wang L, Chin DP, Wang S,

Jiang G, et al. National Survey of Drug-Resistant Tuberculosis in China. N Engl J Med. 2012;366(23):2161–70.

36. Vadwai V, Shetty A, Soman R, Rodrigues C. Determination of risk factors for isoniazid monoresistance and multidrug-resistant tuberculosis in treatment failure patients. Scand J Infect Dis [Internet]. 2012;44(1):48–50. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/21923626

37. Parwati I, Alisjahbana B, Apriani L, Soetikno RD, Ottenhoff TH, van der Zanden AGM, et al. Mycobacterium tuberculosis Beijing genotype is an independent risk factor for tuberculosis treatment failure in Indonesia. J Infect Dis. 2010;201(4):553–7.

38. Lan NTN, Lien HTK, Tung LB, Borgdorff MW, Kremer K, Van Soolingen D. Mycobacterium tuberculosis Beijing Genotype and Risk for Treatment Failure and Relapse, Vietnam. Vol. 9, Emerging Infectious Diseases. 2003. p. 1633–5. 39. Drobniewski F, Balabanova Y, Nikolayevsky

V, Ruddy M, Kuznetzov S, Zakharova S, et al. Drug-resistant tuberculosis, clinical virulence, and the dominance of the Beijing strain family in Russia. Jama [Internet]. 2005;293(22):2726–31. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/15941801

40. Dormans J, Burger M, Aguilar D, Hernandez-Pando R, Kremer K, Roholl P, et al. Correlation of virulence, lung pathology, bacterial load and delayed type hypersensitivity responses after infection with different Mycobacterium tuberculosis genotypes in a BALB/c mouse model. Clin Exp Immunol. 2004;137(3):460–8. 41. Parwati I, van Crevel R, van Soolingen D.

Possible underlying mechanisms for successful emergence of the Mycobacterium tuberculosis Beijing genotype strains. Vol. 10, The Lancet Infectious Diseases. 2010. p. 103–11.

42. Suchindran S, Brouwer ES, Van Rie A. Is HIV infection a risk factor for multi-drug resistant tuberculosis? A systematic review. Vol. 4, PLoS ONE. 2009.

Referenties

GERELATEERDE DOCUMENTEN

Publication of this thesis was financially supported by University of Groningen, Univer- sity Medical Center Groningen, Graduate School of Medical Sciences, Eric Bleumink Fund,

Multidrug-resistant tuberculosis treatment outcomes in relation to treatment and initial versus acquired second-line drug resistance. Management of patients

Ultimately, 37 articles were included describing in vitro susceptibility testing of levofloxacin against MDR-TB strains, and clinical studies on pharmacokinetic monitoring

Studies that used community-based DOT interventions showed opposite effect in Iraq(21) and Tanzania.(28) Family DOT(20) and community DOT(28) were not superior in the improvement

The stigma originating from close family generates discrimination and isolation in TB patients. In the present study, several patients reported to have been left alone by their

(40,41) Moreover, educational programs and drug monitoring provided by a pharmacist demonstrated a significant improvement of adherence to TB treatment among TB and

Beberapa intervensi yang diketahui memiliki efektivitas dalam meningkatkan kepatuhan penggunaan obat dan memperbaiki luaran terapi pada kelompok pasien TB aktif, antara lain

In the development of an antimicrobial stewardship program for TB in Indonesia, problems related to socio-economy, TB drug treatment, knowledge and perception