Acessibilidade / Reportar erro

Brazilian guidelines for the pharmacological treatment of the pulmonary symptoms of cystic fibrosis. Official document of the Sociedade Brasileira de Pneumologia e Tisiologia (SBPT, Brazilian Thoracic Association)

ABSTRACT

Cystic fibrosis (CF) is a genetic disease that results in dysfunction of the CF transmembrane conductance regulator (CFTR) protein, which is a chloride and bicarbonate channel expressed in the apical portion of epithelial cells of various organs. Dysfunction of that protein results in diverse clinical manifestations, primarily involving the respiratory and gastrointestinal systems, impairing quality of life and reducing life expectancy. Although CF is still an incurable pathology, the therapeutic and prognostic perspectives are now totally different and much more favorable. The purpose of these guidelines is to define evidence-based recommendations regarding the use of pharmacological agents in the treatment of the pulmonary symptoms of CF in Brazil. Questions in the Patients of interest, Intervention to be studied, Comparison of interventions, and Outcome of interest (PICO) format were employed to address aspects related to the use of modulators of this protein (ivacaftor, lumacaftor+ivacaftor, and tezacaftor+ivacaftor), use of dornase alfa, eradication therapy and chronic suppression of Pseudomonas aeruginosa, and eradication of methicillin-resistant Staphylococcus aureus and Burkholderia cepacia complex. To formulate the PICO questions, a group of Brazilian specialists was assembled and a systematic review was carried out on the themes, with meta-analysis when applicable. The results obtained were analyzed in terms of the strength of the evidence compiled, the recommendations being devised by employing the GRADE approach. We believe that these guidelines represent a major advance to be incorporated into the approach to patients with CF, mainly aiming to favor the management of the disease, and could become an auxiliary tool in the definition of public policies related to CF.

Keywords:
Cystic fibrosis; GRADE approach; Cystic fibrosis/drug treatment; Clinical practice guide

RESUMO

A fibrose cística (FC) é uma doença genética que resulta em disfunção da proteína reguladora de condutância transmembrana da FC (CFTR), que é um canal de cloro e bicarbonato expresso na porção apical de células epiteliais de diversos órgãos. A disfunção dessa proteína resulta em manifestações clínicas diversas, envolvendo primariamente os sistemas respiratório e gastrointestinal com redução da qualidade e expectativa de vida. A FC ainda é uma patologia incurável, porém o horizonte terapêutico e prognóstico é hoje totalmente distinto e muito mais favorável. O objetivo destas diretrizes foi definir recomendações brasileiras baseadas em evidências em relação ao emprego de agentes farmacológicos no tratamento pulmonar da FC. As perguntas PICO (acrônimo baseado em perguntas referentes aos Pacientes de interesse, Intervenção a ser estudada, Comparação da intervenção e Outcome [desfecho] de interesse) abordaram aspectos relativos ao uso de moduladores de CFTR (ivacaftor, lumacaftor + ivacaftor e tezacaftor + ivacaftor), uso de dornase alfa, terapia de erradicação e supressão crônica de Pseudomonas aeruginosa, e erradicação de Staphylococcus aureus resistente a meticilina e do complexo Burkholderia cepacia. Para a formulação das perguntas, um grupo de especialistas brasileiros foi reunido e realizou-se uma revisão sistemática sobre os temas, com meta-análise quando aplicável. Os resultados encontrados foram analisados quanto à força das evidências compiladas, sendo concebidas recomendações seguindo a metodologia GRADE. Os autores acreditam que o presente documento represente um importante avanço a ser incorporado na abordagem de pacientes com FC, objetivando principalmente favorecer seu manejo, podendo se tornar uma ferramenta auxiliar na definição de políticas públicas relacionadas à FC.

Descritores:
Fibrose cística; Abordagem GRADE; Fibrose cística/tratamento farmacológico; Guia de prática clínica

INTRODUCTION

Cystic fibrosis (CF) is a genetic disease that results in dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein, which is a chloride and bicarbonate channel expressed in the apical membrane of epithelial cells in various organs of the human body.11 Ratjen F, Bell SC, Rowe SM, Goss CH, Quittner AL, Bush A. Cystic fibrosis. Nat Rev Dis Primers. 2015;1:15010. https://doi.org/10.1038/nrdp.2015.10
https://doi.org/10.1038/nrdp.2015.10...
Dysfunction of the CFTR protein results in multisystemic manifestations, impairing quality of life and reducing life expectancy.22 Bell SC, Mall MA, Gutierrez H, Macek M, Madge S, Davies JC, et al. The future of cystic fibrosis care: a global perspective [published correction appears in Lancet Respir Med. 2019 Dec;7(12):e40]. Lancet Respir Med. 2020;8(1):65-124. https://doi.org/10.1016/S2213-2600(19)30337-6
https://doi.org/10.1016/S2213-2600(19)30...

Although CF is still an incurable pathology, the therapeutic perspective is currently more favorable due to the discovery of CFTR modulators.33 Elborn JS. CFTR Modulators: Deciding What Is Best for Individuals in an Era of Precision Medicine. Ann Am Thorac Soc. 2018;15(3):298-300. https://doi.org/10.1513/AnnalsATS.201712-951ED
https://doi.org/10.1513/AnnalsATS.201712...
,44 Habib AR, Kajbafzadeh M, Desai S, Yang CL, Skolnik K, Quon BS. A Systematic Review of the Clinical Efficacy and Safety of CFTR Modulators in Cystic Fibrosis. Sci Rep. 2019;9(1):7234. https://doi.org/10.1038/s41598-019-43652-2
https://doi.org/10.1038/s41598-019-43652...
Historically, treatments for individuals with CF were developed to overcome deficiencies or to modify basic aspects of the pathophysiology of the disease.55 Flume PA, Robinson KA, O'Sullivan BP, Finder JD, Vender RL, Willey-Courand DB, et al. Cystic fibrosis pulmonary guidelines: airway clearance therapies. Respir Care. 2009;54(4):522-537.

6 Mogayzel PJ Jr, Naureckas ET, Robinson KA, Mueller G, Hadjiliadis D, Hoag JB, et al. Cystic fibrosis pulmonary guidelines. Chronic medications for maintenance of lung health. Am J Respir Crit Care Med. 2013;187(7):680-689. https://doi.org/10.1164/rccm.201207-1160OE
https://doi.org/10.1164/rccm.201207-1160...

7 Athanazio RA, Silva Filho LVRF, Vergara AA, Ribeiro AF, Riedi CA, Procianoy EDFA, et al. Brazilian guidelines for the diagnosis and treatment of cystic fibrosis. J Bras Pneumol. 2017;43(3):219-245. https://doi.org/10.1590/s1806-37562017000000065
https://doi.org/10.1590/s1806-3756201700...
-88 Castellani C, Duff AJA, Bell SC, Heijerman HGM, Munck A, Ratjen F, et al. ECFS best practice guidelines: the 2018 revision. J Cyst Fibros. 2018;17(2):153-178. https://doi.org/10.1016/j.jcf.2018.02.006
https://doi.org/10.1016/j.jcf.2018.02.00...

The emergence of CFTR modulators has prompted systematic reviews on the evidence for their beneficial effects on health outcomes for individuals with CF.33 Elborn JS. CFTR Modulators: Deciding What Is Best for Individuals in an Era of Precision Medicine. Ann Am Thorac Soc. 2018;15(3):298-300. https://doi.org/10.1513/AnnalsATS.201712-951ED
https://doi.org/10.1513/AnnalsATS.201712...
,44 Habib AR, Kajbafzadeh M, Desai S, Yang CL, Skolnik K, Quon BS. A Systematic Review of the Clinical Efficacy and Safety of CFTR Modulators in Cystic Fibrosis. Sci Rep. 2019;9(1):7234. https://doi.org/10.1038/s41598-019-43652-2
https://doi.org/10.1038/s41598-019-43652...
,99 Guimbellot J, Sharma J, Rowe SM. Toward inclusive therapy with CFTR modulators: Progress and challenges. Pediatr Pulmonol. 2017;52(S48):S4-S14. https://doi.org/10.1002/ppul.23773
https://doi.org/10.1002/ppul.23773...
,1010 Chaudary N. Triplet CFTR modulators: future prospects for treatment of cystic fibrosis. Ther Clin Risk Manag. 2018;14:2375-2383. https://doi.org/10.2147/TCRM.S147164
https://doi.org/10.2147/TCRM.S147164...
When these guidelines were first being devised, the CFTR modulators approved by the Brazilian Agência Nacional de Vigilância Sanitária (ANVISA, National Health Regulatory Agency) were ivacaftor1111 Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Drevinek P, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. https://doi.org/10.1056/NEJMoa1105185
https://doi.org/10.1056/NEJMoa1105185...
and two drug combinations: lumacaftor+ivacaftor1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
; and tezacaftor+ivacaftor.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
,1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
At the end of 2020, ivacaftor was incorporated into the Brazilian Unified Health Care System for use in individuals with CF ≥ 6 years of age and with genetic regulation (gating) mutations. In 2022, the Brazilian National Health Regulatory Agency approved triple therapy (elexacaftor+tezacaftor+ivacaftor), which proved to be highly effective for individuals with CF carrying the F508del genetic mutation, even for those who are heterozygous for that mutation.1515 Heijerman HGM, McKone EF, Downey DG, Van Braeckel E, Rowe SM, Tullis E, et al. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial [published correction appears in Lancet. 2020 May 30;395(10238):1694]. Lancet. 2019;394(10212):1940-1948. https://doi.org/10.1016/S0140-6736(19)32597-8
https://doi.org/10.1016/S0140-6736(19)32...
,1616 Middleton PG, Mall MA, Drevínek P, Lands LC, McKone EF, Polineni D, et al. Elexacaftor-Tezacaftor-Ivacaftor for Cystic Fibrosis with a Single Phe508del Allele. N Engl J Med. 2019;381(19):1809-1819. https://doi.org/10.1056/NEJMoa1908639
https://doi.org/10.1056/NEJMoa1908639...
Because that drug combination was approved so recently, it was not evaluated in these guidelines.

One of classical treatments for CF, dornase alfa,1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
has long been incorporated into the Brazilian Unified Health Care System. However, questions persist regarding its true impact on relevant outcomes, such as mortality and the frequency of pulmonary exacerbations.1818 Goss CH, MacNeill SJ, Quinton HB, Marshall BC, Elbert A, Knapp EA, et al. Children and young adults with CF in the USA have better lung function compared with the UK. Thorax. 2015;70(3):229-236. https://doi.org/10.1136/thoraxjnl-2014-205718
https://doi.org/10.1136/thoraxjnl-2014-2...

19 Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
https://doi.org/10.1002/14651858.CD00112...
-2020 Grupo Brasileiro de Estudos de Fibrose Cística (GBEFC) [homepage on the Internet]. Curitiba: GBEFC; c2023 [cited 2023 Feb 4]. Registro Brasileiro de Fibrose Cística 2019. [Adobe Acrobat document, 61p.]. Available from: http://portalgbefc.org.br/ckfinder/userfiles/files/REBRAFC_2019.pdf
http://portalgbefc.org.br/ckfinder/userf...

Management strategies for respiratory infections in CF are quite heterogeneous. An evaluation of the evidence supporting eradication regimens for pathogens such as Pseudomonas aeruginosa,2121 Mogayzel PJ Jr, Naureckas ET, Robinson KA, Brady C, Guill M, Lahiri T, et al. Cystic Fibrosis Foundation pulmonary guideline. pharmacologic approaches to prevention and eradication of initial Pseudomonas aeruginosa infection. Ann Am Thorac Soc. 2014;11(10):1640-1650. https://doi.org/10.1513/AnnalsATS.201404-166OC
https://doi.org/10.1513/AnnalsATS.201404...
methicillin-resistant Staphylococcus aureus (MRSA),2222 Lo DK, Muhlebach MS, Smyth AR. Interventions for the eradication of meticillin-resistant Staphylococcus aureus (MRSA) in people with cystic fibrosis. Cochrane Database Syst Rev. 2018;7(7):CD009650. https://doi.org/10.1002/14651858.CD009650.pub4
https://doi.org/10.1002/14651858.CD00965...
and strains of the Burkholderia cepacia complex2323 Garcia BA, Carden JL, Goodwin DL, Smith TA, Gaggar A, Leon K, et al. Implementation of a successful eradication protocol for Burkholderia Cepacia complex in cystic fibrosis patients. BMC Pulm Med. 2018;18(1):35. https://doi.org/10.1186/s12890-018-0594-8
https://doi.org/10.1186/s12890-018-0594-...
could help clarify positions on the risks and benefits of such regimens. Another common practice in the treatment of individuals with CF is suppression therapy for chronic P. aeruginosa infection with inhaled antibiotic therapy. Given the impact of this treatment, which imposes long periods of nebulization,2424 Sawicki GS, Tiddens H. Managing treatment complexity in cystic fibrosis: challenges and opportunities. Pediatr Pulmonol. 2012;47(6):523-533. https://doi.org/10.1002/ppul.22546
https://doi.org/10.1002/ppul.22546...
the topic was also addressed in the guidelines presented here.

The aim of this special article is to carry out a systematic review and meta-analysis of data from the literature involving aspects of the treatment of individuals with CF regarding the use of CFTR modulators and dornase alfa, as well as strategies for the eradication and suppression of pathogens commonly associated with respiratory infections in such individuals.

METHODS

The steps for developing the guidelines followed the model proposed and approved by the Brazilian Thoracic Association, which employs the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach,2525 Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336(7650):924-926. https://doi.org/10.1136/bmj.39489.470347.AD
https://doi.org/10.1136/bmj.39489.470347...
together with questions in the Patients of interest, Intervention to be studied, Comparison of interventions, and Outcome of interest (PICO) format.2626 Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck-Ytter Y, Schünemann HJ; et al. What is "quality of evidence" and why is it important to clinicians?. BMJ. 2008;336(7651):995-998. https://doi.org/10.1136/bmj.39490.551019.BE
https://doi.org/10.1136/bmj.39490.551019...
On May 15, 2019, a virtual meeting, involving four coordinators (two CF specialists and two methodologists), one patient, and a committee of experts, was held for the approval of the methodology used. The experts formulated PICO questions about the pharmacological treatment of patients with CF. A vote was then taken to select the eight most relevant questions. The outcomes of interest for each question were defined a priori and classified as critical or important (Chart 1).

Chart 1
Questions and respective outcomes selected for the preparation of the guidelines.

The search for articles and the meta-analysis were carried out by a team of experienced methodologists, hired for the purpose of devising these guidelines. The project was registered on the International Prospective Register of Systematic Reviews (PROSPERO) platform (Protocol no. CRD42020173901). The searches were carried out in the MEDLINE and EMBASE databases. Clinical trials, case-control studies and cohort studies were included, with keywords pre-established by the specialist coordinators, with no date or language restrictions (Chart S1).

We first evaluated the articles on the basis of their titles and abstracts. Two methodologists, working independently, then performed a qualitative analysis of the full texts and selected articles to be included. Their selections were subsequently validated by the specialist coordinators. The reasons for inclusion or exclusion are presented in the supplementary material (Figures S1 to S8).

When appropriate, data on pharmacological interventions were pooled and meta-analyses were performed independently by the team of methodologists. For each of the eight PICO questions, the quality of evidence of each of the studies included in the meta-analyses was assessed by employing the GRADE approach, through the use of evidence tables, with the GRADEpro Guideline Development Tool (McMaster University, Hamilton, ON, Canada).

For any given study, the quality of evidence depends on the design, the implementation, and the risks of bias. As detailed in Charts 2 and 3, the quality of evidence can be classified as high, moderate, low, or very low.2525 Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336(7650):924-926. https://doi.org/10.1136/bmj.39489.470347.AD
https://doi.org/10.1136/bmj.39489.470347...

Chart 2
Interpretation of the quality of evidence employing the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach.
Chart 3
Factors that can affect the quality of evidence.a

In December of 2021, a working group met to review the evidence and make recommendations for each question according to the GRADE approach. The recommendations were classified as strong or conditional, according to degree of certainty regarding the strength and quality of the evidence. We use the term “recommend” for strong recommendations and “suggest” for conditional recommendations. Chart 4 shows the proposed interpretations of those recommendations,2626 Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck-Ytter Y, Schünemann HJ; et al. What is "quality of evidence" and why is it important to clinicians?. BMJ. 2008;336(7651):995-998. https://doi.org/10.1136/bmj.39490.551019.BE
https://doi.org/10.1136/bmj.39490.551019...
,2727 Guyatt GH, Oxman AD, Kunz R, Falck-Ytter Y, Vist GE, Liberati A, et al. Going from evidence to recommendations [published correction appears in BMJ. 2008 Jun 21;336(7658): doi:10.1136/bmj.a402]. BMJ. 2008;336(7652):1049-1051. https://doi.org/10.1136/bmj.a402 which are detailed in Tables S1 to S8.

Chart 4
Implications of the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach.

Question 1. Should we recommend treatment with ivacaftor in CF patients with class III (gating) or class IV (conduction) mutations in the CFTR gene?

Ivacaftor is a CFTR modulator. It acts as a CFTR potentiator, aimed at treating the dysfunction underlying this genetic alteration. It regulates the opening of the chlorine channel present in the cell membrane, restoring healthy mucus rheology in the airways.

In 2012, the US Food and Drug Administration approved the use of ivacaftor for CF patients ≥ 12 years of age.1111 Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Drevinek P, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. https://doi.org/10.1056/NEJMoa1105185
https://doi.org/10.1056/NEJMoa1105185...
It is the first drug approved for CF whose therapeutic action targets the basic problem of the disease, characterized by CFTR protein dysfunction. Various clinical trials were subsequently carried out, the results of which allowed the use of the drug to be extended to patients ≥ 6 years of age.

Evidence

Among the studies analyzed, there were 28 on the use of ivacaftor, for a variety of outcomes,1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
,2828 Abou Alaiwa MH, Launspach JL, Grogan B, Carter S, Zabner J, Stoltz DA, et al. Ivacaftor-induced sweat chloride reductions correlate with increases in airway surface liquid pH in cystic fibrosis. JCI Insight. 2018;3(15):e121468. https://doi.org/10.1172/jci.insight.121468
https://doi.org/10.1172/jci.insight.1214...

29 Accurso FJ, Rowe SM, Clancy JP, Boyle MP, Dunitz JM, Durie PR, et al. Effect of VX-770 in persons with cystic fibrosis and the G551D-CFTR mutation. N Engl J Med. 2010;363(21):1991-2003. https://doi.org/10.1056/NEJMoa0909825
https://doi.org/10.1056/NEJMoa0909825...

30 Barry PJ, Plant BJ, Nair A, Bicknell S, Simmonds NJ, Bell NJ, et al. Effects of ivacaftor in patients with cystic fibrosis who carry the G551D mutation and have severe lung disease. Chest. 2014;146(1):152-158. https://doi.org/10.1378/chest.13-2397
https://doi.org/10.1378/chest.13-2397...

31 Bessonova L, Volkova N, Higgins M, Bengtsson L, Tian S, Simard C, et al. Data from the US and UK cystic fibrosis registries support disease modification by CFTR modulation with ivacaftor. Thorax. 2018;73(8):731-740. https://doi.org/10.1136/thoraxjnl-2017-210394
https://doi.org/10.1136/thoraxjnl-2017-2...

32 Borowitz D, Lubarsky B, Wilschanski M, Munck A, Gelfond D, Bodewes F, et al. Nutritional Status Improved in Cystic Fibrosis Patients with the G551D Mutation After Treatment with Ivacaftor. Dig Dis Sci. 2016;61(1):198-207. https://doi.org/10.1007/s10620-015-3834-2
https://doi.org/10.1007/s10620-015-3834-...

33 Davies JC, Wainwright CE, Canny GJ, Chilvers MA, Howenstine MS, Munck A, et al. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med. 2013;187(11):1219-1225. https://doi.org/10.1164/rccm.201301-0153OC
https://doi.org/10.1164/rccm.201301-0153...

34 De Boeck K, Munck A, Walker S, Faro A, Hiatt P, Gilmartin G, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros. 2014;13(6):674-680. https://doi.org/10.1016/j.jcf.2014.09.005
https://doi.org/10.1016/j.jcf.2014.09.00...

35 Donaldson SH, Laube BL, Corcoran TE, Bhambhvani P, Zeman K, Ceppe A, et al. Effect of ivacaftor on mucociliary clearance and clinical outcomes in cystic fibrosis patients with G551D-CFTR. JCI Insight. 2018;3(24):e122695. https://doi.org/10.1172/jci.insight.122695
https://doi.org/10.1172/jci.insight.1226...

36 Edgeworth D, Keating D, Ellis M, Button B, Williams E, Clark D, et al. Improvement in exercise duration, lung function and well-being in G551D-cystic fibrosis patients: a double-blind, placebo-controlled, randomized, cross-over study with ivacaftor treatment. Clin Sci (Lond). 2017;131(15):2037-2045. https://doi.org/10.1042/CS20170995
https://doi.org/10.1042/CS20170995...

37 Gomez-Pastrana D, Nwokoro C, McLean M, Brown S, Christiansen N, Pao CS. Real-world effectiveness of ivacaftor in children with cystic fibrosis and the G551D mutation [Article in Spanish]. An Pediatr (Engl Ed). 2019;90(3):148-156. https://doi.org/10.1016/j.anpedi.2018.05.022
https://doi.org/10.1016/j.anpedi.2018.05...

38 Guerra L, D'Oria S, Favia M, Castellani S, Santostasi T, Polizzi AM, et al. CFTR-dependent chloride efflux in cystic fibrosis mononuclear cells is increased by ivacaftor therapy. Pediatr Pulmonol. 2017;52(7):900-908. https://doi.org/10.1002/ppul.23712
https://doi.org/10.1002/ppul.23712...

39 Guimbellot J, Solomon GM, Baines A, Heltshe SL, VanDalfsen J, Joseloff E, et al. Effectiveness of ivacaftor in cystic fibrosis patients with non-G551D gating mutations. J Cyst Fibros. 2019;18(1):102-109. https://doi.org/10.1016/j.jcf.2018.04.004
https://doi.org/10.1016/j.jcf.2018.04.00...

40 Harris JK, Wagner BD, Zemanick ET, Robertson CE, Stevens MJ, Heltshe SL, et al. Changes in Airway Microbiome and Inflammation with Ivacaftor Treatment in Patients with Cystic Fibrosis and the G551D Mutation. Ann Am Thorac Soc. 2020;17(2):212-220. https://doi.org/10.1513/AnnalsATS.201907-493OC
https://doi.org/10.1513/AnnalsATS.201907...

41 Hebestreit H, Sauer-Heilborn A, Fischer R, Käding M, Mainz JG. Effects of ivacaftor on severely ill patients with cystic fibrosis carrying a G551D mutation. J Cyst Fibros. 2013;12(6):599-603. https://doi.org/10.1016/j.jcf.2013.05.006
https://doi.org/10.1016/j.jcf.2013.05.00...

42 Hubert D, Dehillotte C, Munck A, David V, Baek J, Mely L, et al. Retrospective observational study of French patients with cystic fibrosis and a Gly551Asp-CFTR mutation after 1 and 2years of treatment with ivacaftor in a real-world setting. J Cyst Fibros. 2018;17(1):89-95. https://doi.org/10.1016/j.jcf.2017.07.001
https://doi.org/10.1016/j.jcf.2017.07.00...

43 McKone EF, Borowitz D, Drevinek P, Griese M, Konstan MW, Wainwright C, et al. Long-term safety and efficacy of ivacaftor in patients with cystic fibrosis who have the Gly551Asp-CFTR mutation: a phase 3, open-label extension study (PERSIST). Lancet Respir Med. 2014;2(11):902-910. https://doi.org/10.1016/S2213-2600(14)70218-8
https://doi.org/10.1016/S2213-2600(14)70...

44 Moss RB, Flume PA, Elborn JS, Cooke J, Rowe SM, McColley SA, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med. 2015;3(7):524-533. https://doi.org/10.1016/S2213-2600(15)00201-5
https://doi.org/10.1016/S2213-2600(15)00...

45 Quittner A, Suthoff E, Rendas-Baum R, Bayliss MS, Sermet-Gaudelus I, Castiglione B, et al. Effect of ivacaftor treatment in patients with cystic fibrosis and the G551D-CFTR mutation: patient-reported outcomes in the STRIVE randomized, controlled trial. Health Qual Life Outcomes. 2015;13:93. https://doi.org/10.1186/s12955-015-0293-6
https://doi.org/10.1186/s12955-015-0293-...

46 Ronan NJ, Einarsson GG, Twomey M, Mooney D, Mullane D, NiChroinin M, et al. CORK Study in Cystic Fibrosis: Sustained Improvements in Ultra-Low-Dose Chest CT Scores After CFTR Modulation With Ivacaftor. Chest. 2018;153(2):395-403. https://doi.org/10.1016/j.chest.2017.10.005
https://doi.org/10.1016/j.chest.2017.10....

47 Rosenfeld M, Wainwright CE, Higgins M, Wang LT, McKee C, Campbell D, et al. Ivacaftor treatment of cystic fibrosis in children aged 12 to <24 months and with a CFTR gating mutation (ARRIVAL): a phase 3 single-arm study [published correction appears in Lancet Respir Med. 2018 Jul;6(7):e35] [published correction appears in Lancet Respir Med. 2019 Apr;7(4):e15]. Lancet Respir Med. 2018;6(7):545-553. https://doi.org/10.1016/S2213-2600(18)30202-9
https://doi.org/10.1016/S2213-2600(18)30...

48 Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, et al. Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med. 2014;190(2):175-184. https://doi.org/10.1164/rccm.201404-0703OC
https://doi.org/10.1164/rccm.201404-0703...

49 Salvatore D, Carnovale V, Iacotucci P, Braggion C, Castellani C, Cimino G, et al. Effectivenesss of ivacaftor in severe cystic fibrosis patients and non-G551D gating mutations. Pediatr Pulmonol. 2019;54(9):1398-1403. https://doi.org/10.1002/ppul.24424
https://doi.org/10.1002/ppul.24424...

50 Sheikh SI, Long FR, McCoy KS, Johnson T, Ryan-Wenger NA, Hayes D Jr. Computed tomography correlates with improvement with ivacaftor in cystic fibrosis patients with G551D mutation. J Cyst Fibros. 2015;14(1):84-89. https://doi.org/10.1016/j.jcf.2014.06.011
https://doi.org/10.1016/j.jcf.2014.06.01...

51 Sheikh SI, Long FR, McCoy KS, Johnson T, Ryan-Wenger NA, Hayes D Jr. Ivacaftor improves appearance of sinus disease on computerised tomography in cystic fibrosis patients with G551D mutation. Clin Otolaryngol. 2015;40(1):16-21. https://doi.org/10.1111/coa.12310
https://doi.org/10.1111/coa.12310...

52 Stallings VA, Sainath N, Oberle M, Bertolaso C, Schall JI. Energy Balance and Mechanisms of Weight Gain with Ivacaftor Treatment of Cystic Fibrosis Gating Mutations. J Pediatr. 2018;201:229-237.e4. https://doi.org/10.1016/j.jpeds.2018.05.018
https://doi.org/10.1016/j.jpeds.2018.05....

53 Taylor-Cousar J, Niknian M, Gilmartin G, Pilewski JM; VX11-770-901 investigators. Effect of ivacaftor in patients with advanced cystic fibrosis and a G551D-CFTR mutation: Safety and efficacy in an expanded access program in the United States. J Cyst Fibros. 2016;15(1):116-122. https://doi.org/10.1016/j.jcf.2015.01.008
https://doi.org/10.1016/j.jcf.2015.01.00...
-5454 van de Peppel IP, Doktorova M, Berkers G, de Jonge HR, Houwen RHJ, Verkade HJ, et al. IVACAFTOR restores FGF19 regulated bile acid homeostasis in cystic fibrosis patients with an S1251N or a G551D gating mutation. J Cyst Fibros. 2019;18(2):286-293. https://doi.org/10.1016/j.jcf.2018.09.001
https://doi.org/10.1016/j.jcf.2018.09.00...
as shown in Figure S1 and Chart S2. Of those 28 studies, 8 were randomized controlled trials (RCTs).1111 Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Drevinek P, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. https://doi.org/10.1056/NEJMoa1105185
https://doi.org/10.1056/NEJMoa1105185...
,2929 Accurso FJ, Rowe SM, Clancy JP, Boyle MP, Dunitz JM, Durie PR, et al. Effect of VX-770 in persons with cystic fibrosis and the G551D-CFTR mutation. N Engl J Med. 2010;363(21):1991-2003. https://doi.org/10.1056/NEJMoa0909825
https://doi.org/10.1056/NEJMoa0909825...
,3232 Borowitz D, Lubarsky B, Wilschanski M, Munck A, Gelfond D, Bodewes F, et al. Nutritional Status Improved in Cystic Fibrosis Patients with the G551D Mutation After Treatment with Ivacaftor. Dig Dis Sci. 2016;61(1):198-207. https://doi.org/10.1007/s10620-015-3834-2
https://doi.org/10.1007/s10620-015-3834-...

33 Davies JC, Wainwright CE, Canny GJ, Chilvers MA, Howenstine MS, Munck A, et al. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med. 2013;187(11):1219-1225. https://doi.org/10.1164/rccm.201301-0153OC
https://doi.org/10.1164/rccm.201301-0153...
-3434 De Boeck K, Munck A, Walker S, Faro A, Hiatt P, Gilmartin G, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros. 2014;13(6):674-680. https://doi.org/10.1016/j.jcf.2014.09.005
https://doi.org/10.1016/j.jcf.2014.09.00...
,3636 Edgeworth D, Keating D, Ellis M, Button B, Williams E, Clark D, et al. Improvement in exercise duration, lung function and well-being in G551D-cystic fibrosis patients: a double-blind, placebo-controlled, randomized, cross-over study with ivacaftor treatment. Clin Sci (Lond). 2017;131(15):2037-2045. https://doi.org/10.1042/CS20170995
https://doi.org/10.1042/CS20170995...
,4444 Moss RB, Flume PA, Elborn JS, Cooke J, Rowe SM, McColley SA, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med. 2015;3(7):524-533. https://doi.org/10.1016/S2213-2600(15)00201-5
https://doi.org/10.1016/S2213-2600(15)00...
,4545 Quittner A, Suthoff E, Rendas-Baum R, Bayliss MS, Sermet-Gaudelus I, Castiglione B, et al. Effect of ivacaftor treatment in patients with cystic fibrosis and the G551D-CFTR mutation: patient-reported outcomes in the STRIVE randomized, controlled trial. Health Qual Life Outcomes. 2015;13:93. https://doi.org/10.1186/s12955-015-0293-6
https://doi.org/10.1186/s12955-015-0293-...
Although not all investigated the same sets of outcomes, the articles collectively demonstrated a beneficial therapeutic effect of ivacaftor in patients with class III (gating) or class IV (conduction) mutations.

A detailed description of the findings can be found in the Supplementary Material (Question S1). The quality of evidence of the selected articles for this question is summarized in Tables S1A and S1B.

In 2020, Volkova et al.5555 Volkova N, Moy K, Evans J, Campbell D, Tian S, Simard C, et al. Disease progression in patients with cystic fibrosis treated with ivacaftor: Data from national US and UK registries. J Cyst Fibros. 2020;19(1):68-79. https://doi.org/10.1016/j.jcf.2019.05.015
https://doi.org/10.1016/j.jcf.2019.05.01...
evaluated disease progression (real-life study) in CF patients treated with ivacaftor for 5 years. Patients enrolled in US and UK CF registries, which maintain a high degree of data integrity, were evaluated. The authors analyzed 635 cases versus 1,875 controls in the US registry and 247 cases versus 1,230 controls in the UK registry. They observed that, at the end of the 5-year follow-up period, the ivacaftor group patients had better lung function and better nutritional status, as well as a lower frequency of exacerbations and hospitalizations, when compared with their baseline values and with the values obtained for the standard therapy without ivacaftor (control) group.5555 Volkova N, Moy K, Evans J, Campbell D, Tian S, Simard C, et al. Disease progression in patients with cystic fibrosis treated with ivacaftor: Data from national US and UK registries. J Cyst Fibros. 2020;19(1):68-79. https://doi.org/10.1016/j.jcf.2019.05.015
https://doi.org/10.1016/j.jcf.2019.05.01...

Recommendation

For patients with CF and at least one class III (gating) or class IV (conduction) mutation, we suggest the use of ivacaftor (conditional recommendation, very low quality of evidence).

Comments

The results obtained in real-life studies of the use of ivacaftor are similar to those obtained in clinical studies, indicating that it has a positive effect as a modifying drug in the natural progression of CF. The very low quality of the evidence found is due to the high heterogeneity of the studies evaluated, given that our systematic review included clinical trials and observational studies. The studies evaluated included only patients ≥ 6 years of age, and it is not possible to extrapolate the recommendation to any younger age group.

Question 2. Should we recommend treatment with lumacaftor+ivacaftor in CF patients homozygous for the F508del mutation?

Lumacaftor and ivacaftor are both CFTR modulators. Lumacaftor is a CFTR corrector, which acts in the processing of the protein, correcting its format and consequently increasing in its quantity on the cell membrane.44 Habib AR, Kajbafzadeh M, Desai S, Yang CL, Skolnik K, Quon BS. A Systematic Review of the Clinical Efficacy and Safety of CFTR Modulators in Cystic Fibrosis. Sci Rep. 2019;9(1):7234. https://doi.org/10.1038/s41598-019-43652-2
https://doi.org/10.1038/s41598-019-43652...

Combination therapy was evaluated in CF patients aged 6 years or older who were homozygous for the F508del class II mutation. The results indicated a reduction in the number of pulmonary exacerbations, a slight increase in FEV1, improved nutritional status, better quality of life, and a reduction in sweat chloride levels.1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
,5656 Konstan MW, McKone EF, Moss RB, Marigowda G, Tian S, Waltz D, et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): a phase 3, extension study. Lancet Respir Med. 2017;5(2):107-118. https://doi.org/10.1016/S2213-2600(16)30427-1
https://doi.org/10.1016/S2213-2600(16)30...

57 Ratjen F, Hug C, Marigowda G, Tian S, Huang X, Stanojevic S, et al. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial [published correction appears in Lancet Respir Med. 2017 Aug;5(8):e28]. Lancet Respir Med. 2017;5(7):557-567. https://doi.org/10.1016/S2213-2600(17)30215-1
https://doi.org/10.1016/S2213-2600(17)30...

58 Burgel PR, Munck A, Durieu I, Chiron R, Mely L, Prevotat A, et al. Real-Life Safety and Real-Life Safety and Effectiveness of Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2020;201(2):188-197. https://doi.org/10.1164/rccm.201906-1227OC
https://doi.org/10.1164/rccm.201906-1227...

59 Diab-Cáceres L, Girón-Moreno RM, Pastor-Sanz MT, Quintana-Gallego E, Delgado-Pecellín I, Blanco-Aparicio M, et al. Compassionate Use of Lumacaftor/Ivacaftor in Cystic Fibrosis: Spanish Experience. Arch Bronconeumol (Engl Ed). 2018;54(12):614-618. https://doi.org/10.1016/j.arbr.2018.05.020
https://doi.org/10.1016/j.arbr.2018.05.0...
-6060 Graeber SY, Dopfer C, Naehrlich L, Gyulumyan L, Scheuermann H, Hirtz S, et al. Effects of Lumacaftor-Ivacaftor Therapy on Cystic Fibrosis Transmembrane Conductance Regulator Function in Phe508del Homozygous Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2018;197(11):1433-1442. https://doi.org/10.1164/rccm.201710-1983OC
https://doi.org/10.1164/rccm.201710-1983...

In view of the benefits it presents, the use of lumacaftor+ivacaftor has been approved by various international agencies, including the Brazilian ANVISA. In addition, that combination has been shown to have an acceptable safety profile, satisfactory patient adherence (the majority of patients completing the prescribed therapeutic regimen), and a low incidence of relevant adverse events.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
,5858 Burgel PR, Munck A, Durieu I, Chiron R, Mely L, Prevotat A, et al. Real-Life Safety and Real-Life Safety and Effectiveness of Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2020;201(2):188-197. https://doi.org/10.1164/rccm.201906-1227OC
https://doi.org/10.1164/rccm.201906-1227...

59 Diab-Cáceres L, Girón-Moreno RM, Pastor-Sanz MT, Quintana-Gallego E, Delgado-Pecellín I, Blanco-Aparicio M, et al. Compassionate Use of Lumacaftor/Ivacaftor in Cystic Fibrosis: Spanish Experience. Arch Bronconeumol (Engl Ed). 2018;54(12):614-618. https://doi.org/10.1016/j.arbr.2018.05.020
https://doi.org/10.1016/j.arbr.2018.05.0...
-6060 Graeber SY, Dopfer C, Naehrlich L, Gyulumyan L, Scheuermann H, Hirtz S, et al. Effects of Lumacaftor-Ivacaftor Therapy on Cystic Fibrosis Transmembrane Conductance Regulator Function in Phe508del Homozygous Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2018;197(11):1433-1442. https://doi.org/10.1164/rccm.201710-1983OC
https://doi.org/10.1164/rccm.201710-1983...

Evidence

Using the methodology described, we selected 16 articles.1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
,5656 Konstan MW, McKone EF, Moss RB, Marigowda G, Tian S, Waltz D, et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): a phase 3, extension study. Lancet Respir Med. 2017;5(2):107-118. https://doi.org/10.1016/S2213-2600(16)30427-1
https://doi.org/10.1016/S2213-2600(16)30...

57 Ratjen F, Hug C, Marigowda G, Tian S, Huang X, Stanojevic S, et al. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial [published correction appears in Lancet Respir Med. 2017 Aug;5(8):e28]. Lancet Respir Med. 2017;5(7):557-567. https://doi.org/10.1016/S2213-2600(17)30215-1
https://doi.org/10.1016/S2213-2600(17)30...

58 Burgel PR, Munck A, Durieu I, Chiron R, Mely L, Prevotat A, et al. Real-Life Safety and Real-Life Safety and Effectiveness of Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2020;201(2):188-197. https://doi.org/10.1164/rccm.201906-1227OC
https://doi.org/10.1164/rccm.201906-1227...

59 Diab-Cáceres L, Girón-Moreno RM, Pastor-Sanz MT, Quintana-Gallego E, Delgado-Pecellín I, Blanco-Aparicio M, et al. Compassionate Use of Lumacaftor/Ivacaftor in Cystic Fibrosis: Spanish Experience. Arch Bronconeumol (Engl Ed). 2018;54(12):614-618. https://doi.org/10.1016/j.arbr.2018.05.020
https://doi.org/10.1016/j.arbr.2018.05.0...

60 Graeber SY, Dopfer C, Naehrlich L, Gyulumyan L, Scheuermann H, Hirtz S, et al. Effects of Lumacaftor-Ivacaftor Therapy on Cystic Fibrosis Transmembrane Conductance Regulator Function in Phe508del Homozygous Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2018;197(11):1433-1442. https://doi.org/10.1164/rccm.201710-1983OC
https://doi.org/10.1164/rccm.201710-1983...

61 Hubert D, Chiron R, Camara B, Grenet D, Prévotat A, Bassinet L, et al. Real-life initiation of lumacaftor/ivacaftor combination in adults with cystic fibrosis homozygous for the Phe508del CFTR mutation and severe lung disease. J Cyst Fibros. 2017;16(3):388-391. https://doi.org/10.1016/j.jcf.2017.03.003
https://doi.org/10.1016/j.jcf.2017.03.00...

62 Jennings MT, Dezube R, Paranjape S, West NE, Hong G, Braun A, et al. An Observational Study of Outcomes and Tolerances in Patients with Cystic Fibrosis Initiated on Lumacaftor/Ivacaftor. Ann Am Thorac Soc. 2017;14(11):1662-1666. https://doi.org/10.1513/AnnalsATS.201701-058OC
https://doi.org/10.1513/AnnalsATS.201701...

63 Masson A, Schneider-Futschik EK, Baatallah N, Nguyen-Khoa T, Girodon E, Hatton A, et al. Predictive factors for lumacaftor/ivacaftor clinical response. J Cyst Fibros. 2019;18(3):368-374. https://doi.org/10.1016/j.jcf.2018.12.011
https://doi.org/10.1016/j.jcf.2018.12.01...

64 McNamara JJ, McColley SA, Marigowda G, Liu F, Tian S, Owen CA, et al. Safety, pharmacokinetics, and pharmacodynamics of lumacaftor and ivacaftor combination therapy in children aged 2-5 years with cystic fibrosis homozygous for F508del-CFTR: an open-label phase 3 study. Lancet Respir Med. 2019;7(4):325-335. https://doi.org/10.1016/S2213-2600(18)30460-0
https://doi.org/10.1016/S2213-2600(18)30...

65 Milla CE, Ratjen F, Marigowda G, Liu F, Waltz D, Rosenfeld M; et al. Lumacaftor/Ivacaftor in Patients Aged 6-11 Years with Cystic Fibrosis and Homozygous for F508del-CFTR. Am J Respir Crit Care Med. 2017;195(7):912-920. https://doi.org/10.1164/rccm.201608-1754OC
https://doi.org/10.1164/rccm.201608-1754...

66 Murer C, Huber LC, Kurowski T, Hirt A, Robinson CA, Bürgi U, et al. First experience in Switzerland in Phe508del homozygous cystic fibrosis patients with end-stage pulmonary disease enrolled in a lumacaftor-ivacaftor therapy trial - preliminary results. Swiss Med Wkly. 2018;148:w14593. https://doi.org/10.4414/smw.2018.14593
https://doi.org/10.4414/smw.2018.14593...

67 Taylor-Cousar JL, Jain M, Barto TL, Haddad T, Atkinson J, Tian S, et al. Lumacaftor/ivacaftor in patients with cystic fibrosis and advanced lung disease homozygous for F508del-CFTR. J Cyst Fibros. 2018;17(2):228-235. https://doi.org/10.1016/j.jcf.2017.09.012
https://doi.org/10.1016/j.jcf.2017.09.01...

68 Tesell MA, Alper CJ, Bacon R, Greenwood BC, Lenz K, Jeffrey PL, et al. Effect of Lumacaftor/Ivacaftor on Pulmonary Exacerbation Rates in Members with Cystic Fibrosis in a Medicaid Population. J Manag Care Spec Pharm. 2019;25(9):1021-1025. https://doi.org/10.18553/jmcp.2019.25.9.1021
https://doi.org/10.18553/jmcp.2019.25.9....
-6969 Wark PAB, Cookson K, Thiruchelvam T, Brannan J, Dorahy DJ. Lumacaftor/ Ivacaftor improves exercise tolerance in patients with Cystic Fibrosis and severe airflow obstruction. BMC Pulm Med. 2019;19(1):106. https://doi.org/10.1186/s12890-019-0866-y
https://doi.org/10.1186/s12890-019-0866-...
All of those articles were later included for reading, review, and synthesis of evidence (Figure S2 and Chart S3).

Of the 15 studies selected, only 2 were RCTs.1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
,5757 Ratjen F, Hug C, Marigowda G, Tian S, Huang X, Stanojevic S, et al. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial [published correction appears in Lancet Respir Med. 2017 Aug;5(8):e28]. Lancet Respir Med. 2017;5(7):557-567. https://doi.org/10.1016/S2213-2600(17)30215-1
https://doi.org/10.1016/S2213-2600(17)30...
Although not all investigated the same sets of outcomes, they collectively demonstrated that lumacaftor+ivacaftor has a beneficial therapeutic effect in patients homozygous for the F508del mutation.

A detailed description of the findings can be found in the Supplementary Material (Question S2). For each of the articles, the quality of evidence for this question is summarized in Tables S2A and S2B.

Recommendation

For CF patients with the F508del mutation, we do not suggest the use of lumacaftor+ivacaftor (conditional recommendation, very low quality of evidence).

Comments

The combination of a CFTR corrector and a CFTR potentiator can benefit patients homozygous for F508del, representing a differential in the treatment of approximately 45% of individuals with CF and that mutation. However, in the present systematic review, we identified no significant results regarding clinical outcomes that were considered critical. The positive findings obtained for the outcomes that were considered important were only marginal. Some patients may benefit from treatment. However, given the very low quality of evidence for most of the outcomes assessed, we do not suggest the use of lumacaftor+ivacaftor for the treatment of CF patients homozygous for the F508del mutation. It is important to emphasize that new classes/combinations of modulators, such as tezacaftor+ivacaftor1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
and, more recently, the triple combination (elexacaftor+tezacaftor+ivacaftor),1515 Heijerman HGM, McKone EF, Downey DG, Van Braeckel E, Rowe SM, Tullis E, et al. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial [published correction appears in Lancet. 2020 May 30;395(10238):1694]. Lancet. 2019;394(10212):1940-1948. https://doi.org/10.1016/S0140-6736(19)32597-8
https://doi.org/10.1016/S0140-6736(19)32...
have been approved and have been shown to have better efficacy and safety profiles in this population.

Question 3. Should we recommend treatment with tezacaftor+ivacaftor in CF patients homozygous for F508del or heterozygous for F508del and with residual function mutations?

Tezacaftor is a CFTR corrector that binds to the protein, improving its processing and trafficking through the cell to the cell membrane.99 Guimbellot J, Sharma J, Rowe SM. Toward inclusive therapy with CFTR modulators: Progress and challenges. Pediatr Pulmonol. 2017;52(S48):S4-S14. https://doi.org/10.1002/ppul.23773
https://doi.org/10.1002/ppul.23773...

The use of tezacaftor+ivacaftor was evaluated in phase III studies involving CF patients ≥ 12 years of age who were homozygous for the F508del mutation,1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
as well as for patients who were heterozygous for the F508del mutation and had a residual function mutation.1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
In those studies, the use of tezacaftor+ivacaftor was found to provide a significant improvement in lung function, a reduction in the number of exacerbations, and nutritional gain, with an adequate safety profile. Similar results were obtained in patients between 6 and 11 years of age.7070 Kosorok MR, Zeng L, West SE, Rock MJ, Splaingard ML, Laxova A, et al. Acceleration of lung disease in children with cystic fibrosis after Pseudomonas aeruginosa acquisition. Pediatr Pulmonol. 2001;32(4):277-287. https://doi.org/10.1002/ppul.2009.abs
https://doi.org/10.1002/ppul.2009.abs...

71 Blanchard AC, Horton E, Stanojevic S, Taylor L, Waters V, Ratjen F. Effectiveness of a stepwise Pseudomonas aeruginosa eradication protocol in children with cystic fibrosis. J Cyst Fibros. 2017;16(3):395-400. https://doi.org/10.1016/j.jcf.2017.01.007
https://doi.org/10.1016/j.jcf.2017.01.00...
-7272 Claude F, Rochat I, Hafen GM. No benefit of longer eradication therapy of Pseudomonas aeruginosa primoinfections in pediatric cystic fibrosis. BMC Res Notes. 2019;12(1):115. https://doi.org/10.1186/s13104-019-4157-8
https://doi.org/10.1186/s13104-019-4157-...
Therefore, tezacaftor+ivacaftor was approved for use in patients in several countries, including Brazil.

Evidence

As shown in Figure S3 and Chart S4, 5 articles were selected by using the methodology described: 4 RCTs and 1 observational study.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
,1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
,7070 Kosorok MR, Zeng L, West SE, Rock MJ, Splaingard ML, Laxova A, et al. Acceleration of lung disease in children with cystic fibrosis after Pseudomonas aeruginosa acquisition. Pediatr Pulmonol. 2001;32(4):277-287. https://doi.org/10.1002/ppul.2009.abs
https://doi.org/10.1002/ppul.2009.abs...

71 Blanchard AC, Horton E, Stanojevic S, Taylor L, Waters V, Ratjen F. Effectiveness of a stepwise Pseudomonas aeruginosa eradication protocol in children with cystic fibrosis. J Cyst Fibros. 2017;16(3):395-400. https://doi.org/10.1016/j.jcf.2017.01.007
https://doi.org/10.1016/j.jcf.2017.01.00...
-7272 Claude F, Rochat I, Hafen GM. No benefit of longer eradication therapy of Pseudomonas aeruginosa primoinfections in pediatric cystic fibrosis. BMC Res Notes. 2019;12(1):115. https://doi.org/10.1186/s13104-019-4157-8
https://doi.org/10.1186/s13104-019-4157-...
All of the studies assessed more than one outcome, including BMI, quality of life, adverse events, occurrence of exacerbations, lung function, and mortality.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
,1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
,7070 Kosorok MR, Zeng L, West SE, Rock MJ, Splaingard ML, Laxova A, et al. Acceleration of lung disease in children with cystic fibrosis after Pseudomonas aeruginosa acquisition. Pediatr Pulmonol. 2001;32(4):277-287. https://doi.org/10.1002/ppul.2009.abs
https://doi.org/10.1002/ppul.2009.abs...

71 Blanchard AC, Horton E, Stanojevic S, Taylor L, Waters V, Ratjen F. Effectiveness of a stepwise Pseudomonas aeruginosa eradication protocol in children with cystic fibrosis. J Cyst Fibros. 2017;16(3):395-400. https://doi.org/10.1016/j.jcf.2017.01.007
https://doi.org/10.1016/j.jcf.2017.01.00...
-7272 Claude F, Rochat I, Hafen GM. No benefit of longer eradication therapy of Pseudomonas aeruginosa primoinfections in pediatric cystic fibrosis. BMC Res Notes. 2019;12(1):115. https://doi.org/10.1186/s13104-019-4157-8
https://doi.org/10.1186/s13104-019-4157-...

For each of the articles, the quality of evidence for this question is summarized in Table S3. A detailed description of the findings can be found in the Supplementary Material (Question S3).

Recommendation

For CF patients homozygous for F508del or heterozygous for F508del and with a residual function mutation, we suggest using tezacaftor+ivacaftor (conditional recommendation, very low quality of evidence).

Comments

The use of tezacaftor+ivacaftor was evaluated in CF patients ≥ 6 years of age who were homozygous for F508del or heterozygous for F508del and with a residual function mutation. The main effect of the drug was on lung function. The gain in FEV1, albeit modest, seems significant when considered in the context of a disease that leads to a progressive decline in lung function. It is important to point out that the clinical benefits obtained in patients with a residual function mutation seem to be greater than those found in patients homozygous for F508del. In all of the studies evaluated, there was no difference between the control and tezacaftor+ivacaftor groups in terms of the occurrence of adverse events. Most of the adverse events observed were mild and did not lead to discontinuation of the treatment. In addition, many of the events reported (such as increased pulmonary secretion and increased coughing) can be attributed to the disease itself, which attests to the adequate safety profile of the drug.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
,1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
,7070 Kosorok MR, Zeng L, West SE, Rock MJ, Splaingard ML, Laxova A, et al. Acceleration of lung disease in children with cystic fibrosis after Pseudomonas aeruginosa acquisition. Pediatr Pulmonol. 2001;32(4):277-287. https://doi.org/10.1002/ppul.2009.abs
https://doi.org/10.1002/ppul.2009.abs...

71 Blanchard AC, Horton E, Stanojevic S, Taylor L, Waters V, Ratjen F. Effectiveness of a stepwise Pseudomonas aeruginosa eradication protocol in children with cystic fibrosis. J Cyst Fibros. 2017;16(3):395-400. https://doi.org/10.1016/j.jcf.2017.01.007
https://doi.org/10.1016/j.jcf.2017.01.00...
-7272 Claude F, Rochat I, Hafen GM. No benefit of longer eradication therapy of Pseudomonas aeruginosa primoinfections in pediatric cystic fibrosis. BMC Res Notes. 2019;12(1):115. https://doi.org/10.1186/s13104-019-4157-8
https://doi.org/10.1186/s13104-019-4157-...

Question 4. Should we recommend eradicating P. aeruginosa infection in individuals with CF?

Respiratory tract infections are common in individuals with CF, who are more susceptible to infection with certain microorganisms, including P. aeruginosa. Infection with this pathogen is considered a major predictor of morbidity and mortality from CF,7373 Davies JC, Sermet-Gaudelus I, Naehrlich L, Harris RS, Campbell D, Ahluwalia N, et al. A phase 3, double-blind, parallel-group study to evaluate the efficacy and safety of tezacaftor in combination with ivacaftor in participants 6 through 11 years of age with cystic fibrosis homozygous for F508del or heterozygous for the F508del-CFTR mutation and a residual function mutation. J Cyst Fibros. 2021;20(1):68-77. https://doi.org/10.1016/j.jcf.2020.07.023
https://doi.org/10.1016/j.jcf.2020.07.02...
as well as of a severe loss of lung function.7474 Donaldson SH, Pilewski JM, Griese M, Cooke J, Viswanathan L, Tullis E, et al. Tezacaftor/Ivacaftor in Subjects with Cystic Fibrosis and F508del/F508del-CFTR or F508del/G551D-CFTR. Am J Respir Crit Care Med. 2018;197(2):214-224. https://doi.org/10.1164/rccm.201704-0717OC
https://doi.org/10.1164/rccm.201704-0717...
Since the 1990s, CF referral centers have used regimens for early eradication of P. aeruginosa, aiming to delay the progression to chronic infection and its unfavorable outcomes.

Evidence

Using the methodology described, we selected 17 articles, conducted between 1980 and 2019 and published between 1991 and 2020,7575 Walker S, Flume P, McNamara J, Solomon M, Chilvers M, Chmiel J, et al. A phase 3 study of tezacaftor in combination with ivacaftor in children aged 6 through 11 years with cystic fibrosis. J Cyst Fibros. 2019;18(5):708-713. https://doi.org/10.1016/j.jcf.2019.06.009
https://doi.org/10.1016/j.jcf.2019.06.00...

76 Emerson J, Rosenfeld M, McNamara S, Ramsey B, Gibson RL. Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis. Pediatr Pulmonol. 2002;34(2):91-100. https://doi.org/10.1002/ppul.10127
https://doi.org/10.1002/ppul.10127...

77 Cohen-Cymberknoh M, Gilead N, Gartner S, Rovira S, Blau H, Mussaffi H, et al. Eradication failure of newly acquired Pseudomonas aeruginosa isolates in cystic fibrosis. J Cyst Fibros. 2016;15(6):776-782. https://doi.org/10.1016/j.jcf.2016.04.006
https://doi.org/10.1016/j.jcf.2016.04.00...

78 Emiralioglu N, Yalcin E, Meral A, Sener B, Dogru D, Ozcelik U, et al. The success of the different eradication therapy regimens for Pseudomonas aeruginosa in cystic fibrosis. J Clin Pharm Ther. 2016;41(4):419-423. https://doi.org/10.1111/jcpt.12407
https://doi.org/10.1111/jcpt.12407...

79 Frederiksen B, Koch C, Høiby N. Antibiotic treatment of initial colonization with Pseudomonas aeruginosa postpones chronic infection and prevents deterioration of pulmonary function in cystic fibrosis. Pediatr Pulmonol. 1997;23(5):330-335. https://doi.org/10.1002/(SICI)1099-0496(199705)23:5<330::AID-PPUL4>3.0.CO;2-O
https://doi.org/10.1002/(SICI)1099-0496(...

80 Giugno H, Castaños C, Lubatti A, Pinheiro JL, Hernández C, González Pena H. Early antibiotic treatment for eradication of initial infection by Pseudomonas aeruginosa in patients with cystic fibrosis. Arch Argent Pediatr. 2010;108(2):141-147. https://doi.org/10.1590/S0325-00752010000200009
https://doi.org/10.1590/S0325-0075201000...

81 Hewer SCL, Smyth AR, Brown M, Jones AP, Hickey H, Kenna D, et al. Intravenous versus oral antibiotics for eradication of Pseudomonas aeruginosa in cystic fibrosis (TORPEDO-CF): a randomised controlled trial. Lancet Respir Med. 2020;8(10):975-986. https://doi.org/10.1016/S2213-2600(20)30331-3
https://doi.org/10.1016/S2213-2600(20)30...

82 Kenny SL, Shaw TD, Downey DG, Moore JE, Rendall JC, Elborn JS. Eradication of Pseudomonas aeruginosa in adults with cystic fibrosis. BMJ Open Respir Res. 2014;1(1):e000021. https://doi.org/10.1136/bmjresp-2014-000021
https://doi.org/10.1136/bmjresp-2014-000...

83 Mayer-Hamblett N, Kloster M, Rosenfeld M, Gibson RL, Retsch-Bogart GZ, Emerson J, et al. Impact of Sustained Eradication of New Pseudomonas aeruginosa Infection on Long-term Outcomes in Cystic Fibrosis. Clin Infect Dis. 2015;61(5):707-715. https://doi.org/10.1093/cid/civ377
https://doi.org/10.1093/cid/civ377...

84 Proesmans M, Vermeulen F, Boulanger L, Verhaegen J, De Boeck K. Comparison of two treatment regimens for eradication of Pseudomonas aeruginosa infection in children with cystic fibrosis. J Cyst Fibros. 2013;12(1):29-34. https://doi.org/10.1016/j.jcf.2012.06.001
https://doi.org/10.1016/j.jcf.2012.06.00...

85 Ratjen F, Munck A, Kho P, Angyalosi G; ELITE Study Group. Treatment of early Pseudomonas aeruginosa infection in patients with cystic fibrosis: the ELITE trial. Thorax. 2010;65(4):286-291. https://doi.org/10.1136/thx.2009.121657
https://doi.org/10.1136/thx.2009.121657...

86 Ratjen F, Moeller A, McKinney ML, Asherova I, Alon N, Maykut R, et al. Eradication of early P. aeruginosa infection in children <7 years of age with cystic fibrosis: The early study. J Cyst Fibros. 2019;18(1):78-85. https://doi.org/10.1016/j.jcf.2018.04.002
https://doi.org/10.1016/j.jcf.2018.04.00...

87 Taccetti G, Campana S, Festini F, Mascherini M, Döring G. Early eradication therapy against Pseudomonas aeruginosa in cystic fibrosis patients. Eur Respir J. 2005;26(3):458-461. https://doi.org/10.1183/09031936.05.00009605
https://doi.org/10.1183/09031936.05.0000...

88 Taccetti G, Bianchini E, Cariani L, Buzzetti R, Costantini D, Trevisan F, et al. Early antibiotic treatment for Pseudomonas aeruginosa eradication in patients with cystic fibrosis: a randomised multicentre study comparing two different protocols. Thorax. 2012;67(10):853-859. https://doi.org/10.1136/thoraxjnl-2011-200832
https://doi.org/10.1136/thoraxjnl-2011-2...

89 Treggiari MM, Retsch-Bogart G, Mayer-Hamblett N, Khan U, Kulich M, Kronmal R, et al. Comparative efficacy and safety of 4 randomized regimens to treat early Pseudomonas aeruginosa infection in children with cystic fibrosis. Arch Pediatr Adolesc Med. 2011;165(9):847-856. https://doi.org/10.1001/archpediatrics.2011.136
https://doi.org/10.1001/archpediatrics.2...

90 Valerius NH, Koch C, Høiby N. Prevention of chronic Pseudomonas aeruginosa colonisation in cystic fibrosis by early treatment. Lancet. 1991;338(8769):725-726. https://doi.org/10.1016/0140-6736(91)91446-2
https://doi.org/10.1016/0140-6736(91)914...
-9191 Wiesemann HG, Steinkamp G, Ratjen F, Bauernfeind A, Przyklenk B, Döring G, et al. Placebo-controlled, double-blind, randomized study of aerosolized tobramycin for early treatment of Pseudomonas aeruginosa colonization in cystic fibrosis. Pediatr Pulmonol. 1998;25(2):88-92. https://doi.org/10.1002/(SICI)1099-0496(199802)25:2<88::AID-PPUL3>3.0.CO;2-J
https://doi.org/10.1002/(SICI)...
of which 10 were observational studies7575 Walker S, Flume P, McNamara J, Solomon M, Chilvers M, Chmiel J, et al. A phase 3 study of tezacaftor in combination with ivacaftor in children aged 6 through 11 years with cystic fibrosis. J Cyst Fibros. 2019;18(5):708-713. https://doi.org/10.1016/j.jcf.2019.06.009
https://doi.org/10.1016/j.jcf.2019.06.00...

76 Emerson J, Rosenfeld M, McNamara S, Ramsey B, Gibson RL. Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis. Pediatr Pulmonol. 2002;34(2):91-100. https://doi.org/10.1002/ppul.10127
https://doi.org/10.1002/ppul.10127...

77 Cohen-Cymberknoh M, Gilead N, Gartner S, Rovira S, Blau H, Mussaffi H, et al. Eradication failure of newly acquired Pseudomonas aeruginosa isolates in cystic fibrosis. J Cyst Fibros. 2016;15(6):776-782. https://doi.org/10.1016/j.jcf.2016.04.006
https://doi.org/10.1016/j.jcf.2016.04.00...

78 Emiralioglu N, Yalcin E, Meral A, Sener B, Dogru D, Ozcelik U, et al. The success of the different eradication therapy regimens for Pseudomonas aeruginosa in cystic fibrosis. J Clin Pharm Ther. 2016;41(4):419-423. https://doi.org/10.1111/jcpt.12407
https://doi.org/10.1111/jcpt.12407...

79 Frederiksen B, Koch C, Høiby N. Antibiotic treatment of initial colonization with Pseudomonas aeruginosa postpones chronic infection and prevents deterioration of pulmonary function in cystic fibrosis. Pediatr Pulmonol. 1997;23(5):330-335. https://doi.org/10.1002/(SICI)1099-0496(199705)23:5<330::AID-PPUL4>3.0.CO;2-O
https://doi.org/10.1002/(SICI)1099-0496(...
-8080 Giugno H, Castaños C, Lubatti A, Pinheiro JL, Hernández C, González Pena H. Early antibiotic treatment for eradication of initial infection by Pseudomonas aeruginosa in patients with cystic fibrosis. Arch Argent Pediatr. 2010;108(2):141-147. https://doi.org/10.1590/S0325-00752010000200009
https://doi.org/10.1590/S0325-0075201000...
,8282 Kenny SL, Shaw TD, Downey DG, Moore JE, Rendall JC, Elborn JS. Eradication of Pseudomonas aeruginosa in adults with cystic fibrosis. BMJ Open Respir Res. 2014;1(1):e000021. https://doi.org/10.1136/bmjresp-2014-000021
https://doi.org/10.1136/bmjresp-2014-000...
,8383 Mayer-Hamblett N, Kloster M, Rosenfeld M, Gibson RL, Retsch-Bogart GZ, Emerson J, et al. Impact of Sustained Eradication of New Pseudomonas aeruginosa Infection on Long-term Outcomes in Cystic Fibrosis. Clin Infect Dis. 2015;61(5):707-715. https://doi.org/10.1093/cid/civ377
https://doi.org/10.1093/cid/civ377...
,8686 Ratjen F, Moeller A, McKinney ML, Asherova I, Alon N, Maykut R, et al. Eradication of early P. aeruginosa infection in children <7 years of age with cystic fibrosis: The early study. J Cyst Fibros. 2019;18(1):78-85. https://doi.org/10.1016/j.jcf.2018.04.002
https://doi.org/10.1016/j.jcf.2018.04.00...
,8787 Taccetti G, Campana S, Festini F, Mascherini M, Döring G. Early eradication therapy against Pseudomonas aeruginosa in cystic fibrosis patients. Eur Respir J. 2005;26(3):458-461. https://doi.org/10.1183/09031936.05.00009605
https://doi.org/10.1183/09031936.05.0000...
and 7 were RCTs.8181 Hewer SCL, Smyth AR, Brown M, Jones AP, Hickey H, Kenna D, et al. Intravenous versus oral antibiotics for eradication of Pseudomonas aeruginosa in cystic fibrosis (TORPEDO-CF): a randomised controlled trial. Lancet Respir Med. 2020;8(10):975-986. https://doi.org/10.1016/S2213-2600(20)30331-3
https://doi.org/10.1016/S2213-2600(20)30...
,8484 Proesmans M, Vermeulen F, Boulanger L, Verhaegen J, De Boeck K. Comparison of two treatment regimens for eradication of Pseudomonas aeruginosa infection in children with cystic fibrosis. J Cyst Fibros. 2013;12(1):29-34. https://doi.org/10.1016/j.jcf.2012.06.001
https://doi.org/10.1016/j.jcf.2012.06.00...
,8585 Ratjen F, Munck A, Kho P, Angyalosi G; ELITE Study Group. Treatment of early Pseudomonas aeruginosa infection in patients with cystic fibrosis: the ELITE trial. Thorax. 2010;65(4):286-291. https://doi.org/10.1136/thx.2009.121657
https://doi.org/10.1136/thx.2009.121657...
,8888 Taccetti G, Bianchini E, Cariani L, Buzzetti R, Costantini D, Trevisan F, et al. Early antibiotic treatment for Pseudomonas aeruginosa eradication in patients with cystic fibrosis: a randomised multicentre study comparing two different protocols. Thorax. 2012;67(10):853-859. https://doi.org/10.1136/thoraxjnl-2011-200832
https://doi.org/10.1136/thoraxjnl-2011-2...

89 Treggiari MM, Retsch-Bogart G, Mayer-Hamblett N, Khan U, Kulich M, Kronmal R, et al. Comparative efficacy and safety of 4 randomized regimens to treat early Pseudomonas aeruginosa infection in children with cystic fibrosis. Arch Pediatr Adolesc Med. 2011;165(9):847-856. https://doi.org/10.1001/archpediatrics.2011.136
https://doi.org/10.1001/archpediatrics.2...

90 Valerius NH, Koch C, Høiby N. Prevention of chronic Pseudomonas aeruginosa colonisation in cystic fibrosis by early treatment. Lancet. 1991;338(8769):725-726. https://doi.org/10.1016/0140-6736(91)91446-2
https://doi.org/10.1016/0140-6736(91)914...
-9191 Wiesemann HG, Steinkamp G, Ratjen F, Bauernfeind A, Przyklenk B, Döring G, et al. Placebo-controlled, double-blind, randomized study of aerosolized tobramycin for early treatment of Pseudomonas aeruginosa colonization in cystic fibrosis. Pediatr Pulmonol. 1998;25(2):88-92. https://doi.org/10.1002/(SICI)1099-0496(199802)25:2<88::AID-PPUL3>3.0.CO;2-J
https://doi.org/10.1002/(SICI)...
The number of individuals included in the studies ranged from 11 to 304, and 12 of the studies had a sample size of less than 200 (Figure S4 and Chart S5).

For each of the articles, the quality of evidence for this question is summarized in Table S4. A detailed description of the findings can be found in the Supplementary Material (Question S4).

Recommendation

For individuals with CF, we do not have sufficient evidence to recommend or not recommend the use of P. aeruginosa eradication therapy.

Comments

The association between P. aeruginosa infection and poor CF outcomes is well established. In addition, eradication therapy has already been incorporated into the routine of referral centers, making it difficult to carry out new studies on the subject. The literature review made it possible to include a limited number of studies, most of which were observational in nature and, in general, had small sample sizes. Such characteristics can lead to inconsistent results, making it impossible to make an appropriate recommendation on the subject. Despite being a practice recommended in several national and international guidelines, further studies are needed to determine the efficacy and safety of P. aeruginosa eradication therapy, especially in the era of CFTR modulator use.

Question 5. Should we recommend treatment with inhaled antimicrobials in patients with CF and chronic P. aeruginosa infection?

Through complex mechanisms, P. aeruginosa adapts to and can remain in the airways of CF patients for long periods. Chronic infection in CF is defined as detection of the pathogen in more than 50% of respiratory secretion samples over a 12-month period.9292 Lee TW, Brownlee KG, Conway SP, Denton M, Littlewood JM. Evaluation of a new definition for chronic Pseudomonas aeruginosa infection in cystic fibrosis patients. J Cyst Fibros. 2003;2(1):29-34. https://doi.org/10.1016/S1569-1993(02)00141-8
https://doi.org/10.1016/S1569-1993(02)00...
Chronic infection with P. aeruginosa can affect up to 60% of patients in adult life and is associated with progression of lung disease and higher mortality.2121 Mogayzel PJ Jr, Naureckas ET, Robinson KA, Brady C, Guill M, Lahiri T, et al. Cystic Fibrosis Foundation pulmonary guideline. pharmacologic approaches to prevention and eradication of initial Pseudomonas aeruginosa infection. Ann Am Thorac Soc. 2014;11(10):1640-1650. https://doi.org/10.1513/AnnalsATS.201404-166OC
https://doi.org/10.1513/AnnalsATS.201404...
,9393 Lund-Palau H, Turnbull AR, Bush A, Bardin E, Cameron L, Soren O, et al. Pseudomonas aeruginosa infection in cystic fibrosis: pathophysiological mechanisms and therapeutic approaches. Expert Rev Respir Med. 2016;10(6):685-697. https://doi.org/10.1080/17476348.2016.1177460
https://doi.org/10.1080/17476348.2016.11...
,9494 Flume PA, VanDevanter DR, Morgan EE, Dudley MN, Loutit JS, Bell SC, et al. A phase 3, multi-center, multinational, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of levofloxacin inhalation solution (APT-1026) in stable cystic fibrosis patients. J Cyst Fibros. 2016;15(4):495-502. https://doi.org/10.1016/j.jcf.2015.12.004
https://doi.org/10.1016/j.jcf.2015.12.00...
Inhaled antimicrobials are widely used for the suppression of P. aeruginosa in patients with chronic infection, and their use is aimed at reducing the consequences of the presence of the pathogen in the airways. Inhaled drug options for such treatment classically include colistimethate, tobramycin, and, more recently, aztreonam.9393 Lund-Palau H, Turnbull AR, Bush A, Bardin E, Cameron L, Soren O, et al. Pseudomonas aeruginosa infection in cystic fibrosis: pathophysiological mechanisms and therapeutic approaches. Expert Rev Respir Med. 2016;10(6):685-697. https://doi.org/10.1080/17476348.2016.1177460
https://doi.org/10.1080/17476348.2016.11...

Evidence

Using the methodology described, we selected 25 studies carried out between 1995 and 2008.9494 Flume PA, VanDevanter DR, Morgan EE, Dudley MN, Loutit JS, Bell SC, et al. A phase 3, multi-center, multinational, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of levofloxacin inhalation solution (APT-1026) in stable cystic fibrosis patients. J Cyst Fibros. 2016;15(4):495-502. https://doi.org/10.1016/j.jcf.2015.12.004
https://doi.org/10.1016/j.jcf.2015.12.00...

95 Assael BM, Pressler T, Bilton D, Fayon M, Fischer R, Chiron R, et al. Inhaled aztreonam lysine vs. inhaled tobramycin in cystic fibrosis: a comparative efficacy trial. J Cyst Fibros. 2013;12(2):130-140. https://doi.org/10.1016/j.jcf.2012.07.006
https://doi.org/10.1016/j.jcf.2012.07.00...

96 Chuchalin A, Csiszér E, Gyurkovics K, Bartnicka MT, Sands D, Kapranov N, et al. A formulation of aerosolized tobramycin (Bramitob) in the treatment of patients with cystic fibrosis and Pseudomonas aeruginosa infection: a double-blind, placebo-controlled, multicenter study. Paediatr Drugs. 2007;9 Suppl 1:21-31. https://doi.org/10.2165/00148581-200709001-00004
https://doi.org/10.2165/00148581-2007090...

97 Geller DE, Flume PA, Staab D, Fischer R, Loutit JS, Conrad DJ; et al. Levofloxacin inhalation solution (MP-376) in patients with cystic fibrosis with Pseudomonas aeruginosa. Am J Respir Crit Care Med. 2011;183(11):1510-1516. https://doi.org/10.1164/rccm.201008-1293OC
https://doi.org/10.1164/rccm.201008-1293...

98 Hodson ME, Penketh AR, Batten JC. Aerosol carbenicillin and gentamicin treatment of Pseudomonas aeruginosa infection in patients with cystic fibrosis. Lancet. 1981;2(8256):1137-1139. https://doi.org/10.1016/S0140-6736(81)90588-2
https://doi.org/10.1016/S0140-6736(81)90...

99 Hodson ME, Gallagher CG, Govan JR. A randomised clinical trial of nebulised tobramycin or colistin in cystic fibrosis. Eur Respir J. 2002;20(3):658-664. https://doi.org/10.1183/09031936.02.00248102
https://doi.org/10.1183/09031936.02.0024...

100 Jensen T, Pedersen SS, Garne S, Heilmann C, Høiby N, Koch C. Colistin inhalation therapy in cystic fibrosis patients with chronic Pseudomonas aeruginosa lung infection. J Antimicrob Chemother. 1987;19(6):831-838. https://doi.org/10.1093/jac/19.6.831
https://doi.org/10.1093/jac/19.6.831...

101 Konstan MW, Geller DE, Minic P, Brockhaus F, Zhang J, Angyalosi G. Tobramycin inhalation powder for P. aeruginosa infection in cystic fibrosis: the EVOLVE trial. Pediatr Pulmonol. 2011;46(3):230-238. https://doi.org/10.1002/ppul.21356
https://doi.org/10.1002/ppul.21356...

102 Konstan MW, Flume PA, Kappler M, Chiron R, Higgins M, Brockhaus F, et al. Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: The EAGER trial. J Cyst Fibros. 2011;10(1):54-61. https://doi.org/10.1016/j.jcf.2010.10.003
https://doi.org/10.1016/j.jcf.2010.10.00...

103 Kun P, Landau LI, Phelan PD. Nebulized gentamicin in children and adolescents with cystic fibrosis. Aust Paediatr J. 1984;20(1):43-45. https://doi.org/10.1111/j.1440-1754.1984.tb00035.x
https://doi.org/10.1111/j.1440-1754.1984...

104 Lenoir G, Antypkin YG, Miano A, Moretti P, Zanda M, Varoli G, et al. Efficacy, safety, and local pharmacokinetics of highly concentrated nebulized tobramycin in patients with cystic fibrosis colonized with Pseudomonas aeruginosa. Paediatr Drugs. 2007;9 Suppl 1:11-20. https://doi.org/10.2165/00148581-200709001-00003
https://doi.org/10.2165/00148581-2007090...

105 MacLusky IB, Gold R, Corey M, Levison H. Long-term effects of inhaled tobramycin in patients with cystic fibrosis colonized with Pseudomonas aeruginosa. Pediatr Pulmonol. 1989;7(1):42-48. https://doi.org/10.1002/ppul.1950070110
https://doi.org/10.1002/ppul.1950070110...

106 McCoy KS, Quittner AL, Oermann CM, Gibson RL, Retsch-Bogart GZ, Montgomery AB. Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis. Am J Respir Crit Care Med. 2008;178(9):921-928. https://doi.org/10.1164/rccm.200712-1804OC
https://doi.org/10.1164/rccm.200712-1804...

107 Moss RB. Administration of aerosolized antibiotics in cystic fibrosis patients. Chest. 2001;120(3 Suppl):107S-113S. https://doi.org/10.1378/chest.120.3_suppl.107S
https://doi.org/10.1378/chest.120.3_supp...

108 Murphy TD, Anbar RD, Lester LA, Nasr SZ, Nickerson B, VanDevanter DR, et al. Treatment with tobramycin solution for inhalation reduces hospitalizations in young CF subjects with mild lung disease. Pediatr Pulmonol. 2004;38(4):314-320. https://doi.org/10.1002/ppul.20097
https://doi.org/10.1002/ppul.20097...

109 Nasr SZ, Sakmar E, Christodoulou E, Eckhardt BP, Streetman DS, Strouse PJ. The use of high resolution computerized tomography (HRCT) of the chest in evaluating the effect of tobramycin solution for inhalation in cystic fibrosis lung disease. Pediatr Pulmonol. 2010;45(5):440-449. https://doi.org/10.1002/ppul.21188
https://doi.org/10.1002/ppul.21188...

110 Nikolaizik WH, Vietzke D, Ratjen F. A pilot study to compare tobramycin 80 mg injectable preparation with 300 mg solution for inhalation in cystic fibrosis patients. Can Respir J. 2008;15(5):259-262. https://doi.org/10.1155/2008/202464
https://doi.org/10.1155/2008/202464...

111 Quintana-Gallego E, Lopez-Campos JL, Calero C, Dapena FJ. Nebulized colistin versus tobramycin in the treatment of chronic Pseudomonas colonization in cystic fibrosis patients [Article in Spanish]. Med Clin (Barc). 2014;142(2):59-63. https://doi.org/10.1016/j.medcli.2012.11.026
https://doi.org/10.1016/j.medcli.2012.11...

112 Ramsey BW, Pepe MS, Quan JM, Otto KL, Montgomery AB, Williams-Warren J, et al. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group. N Engl J Med. 1999;340(1):23-30. https://doi.org/10.1056/NEJM199901073400104
https://doi.org/10.1056/NEJM199901073400...

113 Retsch-Bogart GZ, Quittner AL, Gibson RL, Oermann CM, McCoy KS, Montgomery AB, et al. Efficacy and safety of inhaled aztreonam lysine for airway pseudomonas in cystic fibrosis. Chest. 2009;135(5):1223-1232. https://doi.org/10.1378/chest.08-1421
https://doi.org/10.1378/chest.08-1421...

114 Schuster A, Haliburn C, Döring G, Goldman MH; Freedom Study Group. Safety, efficacy and convenience of colistimethate sodium dry powder for inhalation (Colobreathe DPI) in patients with cystic fibrosis: a randomised study. Thorax. 2013;68(4):344-350. https://doi.org/10.1136/thoraxjnl-2012-202059
https://doi.org/10.1136/thoraxjnl-2012-2...

115 Stead RJ, Hodson ME, Batten JC. Inhaled ceftazidime compared with gentamicin and carbenicillin in older patients with cystic fibrosis infected with Pseudomonas aeruginosa. Br J Dis Chest. 1987;81(3):272-279. https://doi.org/10.1016/0007-0971(87)90161-6
https://doi.org/10.1016/0007-0971(87)901...

116 Stuart Elborn J, Geller DE, Conrad D, Aaron SD, Smyth AR, Fischer R, et al. A phase 3, open-label, randomized trial to evaluate the safety and efficacy of levofloxacin inhalation solution (APT-1026) versus tobramycin inhalation solution in stable cystic fibrosis patients. J Cyst Fibros. 2015;14(4):507-514. https://doi.org/10.1016/j.jcf.2014.12.013
https://doi.org/10.1016/j.jcf.2014.12.01...

117 Toukan Y, Gur M, Hakim FH, Geffen Y, Bar-Yoseph R, Nir V, et al. Intermittent inhaled tobramycin and systemic cytokines response in CF patients with Pseudomonas aeruginosa. Clin Invest Med. 2018;41(3):E136-E143. https://doi.org/10.25011/cim.v41i3.30792
https://doi.org/10.25011/cim.v41i3.30792...
-118118 Westerman EM, Le Brun PP, Touw DJ, Frijlink HW, Heijerman HG. Effect of nebulized colistin sulphate and colistin sulphomethate on lung function in patients with cystic fibrosis: a pilot study. J Cyst Fibros. 2004;3(1):23-28. https://doi.org/10.1016/j.jcf.2003.12.005
https://doi.org/10.1016/j.jcf.2003.12.00...
Five were observational studies101101 Konstan MW, Geller DE, Minic P, Brockhaus F, Zhang J, Angyalosi G. Tobramycin inhalation powder for P. aeruginosa infection in cystic fibrosis: the EVOLVE trial. Pediatr Pulmonol. 2011;46(3):230-238. https://doi.org/10.1002/ppul.21356
https://doi.org/10.1002/ppul.21356...

102 Konstan MW, Flume PA, Kappler M, Chiron R, Higgins M, Brockhaus F, et al. Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: The EAGER trial. J Cyst Fibros. 2011;10(1):54-61. https://doi.org/10.1016/j.jcf.2010.10.003
https://doi.org/10.1016/j.jcf.2010.10.00...
-103103 Kun P, Landau LI, Phelan PD. Nebulized gentamicin in children and adolescents with cystic fibrosis. Aust Paediatr J. 1984;20(1):43-45. https://doi.org/10.1111/j.1440-1754.1984.tb00035.x
https://doi.org/10.1111/j.1440-1754.1984...
,109109 Nasr SZ, Sakmar E, Christodoulou E, Eckhardt BP, Streetman DS, Strouse PJ. The use of high resolution computerized tomography (HRCT) of the chest in evaluating the effect of tobramycin solution for inhalation in cystic fibrosis lung disease. Pediatr Pulmonol. 2010;45(5):440-449. https://doi.org/10.1002/ppul.21188
https://doi.org/10.1002/ppul.21188...
,117117 Toukan Y, Gur M, Hakim FH, Geffen Y, Bar-Yoseph R, Nir V, et al. Intermittent inhaled tobramycin and systemic cytokines response in CF patients with Pseudomonas aeruginosa. Clin Invest Med. 2018;41(3):E136-E143. https://doi.org/10.25011/cim.v41i3.30792
https://doi.org/10.25011/cim.v41i3.30792...
and the others were classified as RCTs. Samples sizes were over 200 in 9 of the studies, of which 2 included more than 500 individuals (Figure S5 and Chart S6).

For each of the articles, the quality of evidence for this question is summarized in Table S5. A detailed description of the findings can be found in the Supplementary Material (Question S5).

Recommendation

For CF patients with chronic P. aeruginosa colonization, we suggest chronic suppression therapy with inhaled antibiotics (conditional recommendation, very low quality of evidence).

Comments

Although not all of the studies analyzed exactly the same sets of outcomes, as a whole, they point in favor of the use of inhalation treatment of patients with chronic P. aeruginosa colonization, because such treatment can result in functional improvement, better quality of life, and lower mortality in those patients. It should be borne in mind that, despite the potential benefits, there is heterogeneity among the studies evaluated, resulting in a low quality of evidence.

Question 6. Should we recommend antimicrobial eradication treatment in CF patients with MRSA colonization of the airways?

Chronic MRSA infection is associated with worse clinical outcomes in CF patients.119119 Dasenbrook EC, Checkley W, Merlo CA, Konstan MW, Lechtzin N, Boyle MP. Association between respiratory tract methicillin-resistant Staphylococcus aureus and survival in cystic fibrosis. JAMA. 2010;303(23):2386-2392. https://doi.org/10.1001/jama.2010.791
https://doi.org/10.1001/jama.2010.791...
There are various antimicrobial regimens for eradicating this pathogen, including combinations of oral, topical, and inhaled drugs. There is also great variability regarding treatment time, and some authors argue that combined treatment is more effective than is monotherapy.120120 Hall H, Gadhok R, Alshafi K, Bilton D, Simmonds NJ. Eradication of respiratory tract MRSA at a large adult cystic fibrosis centre. Respir Med. 2015;109(3):357-363. https://doi.org/10.1016/j.rmed.2015.01.013
https://doi.org/10.1016/j.rmed.2015.01.0...
,121121 Vallières E, Rendall JC, Moore JE, McCaughan J, Hoeritzauer AI, Tunney MM, et al. MRSA eradication of newly acquired lower respiratory tract infection in cystic fibrosis. ERJ Open Res. 2016;2(1):00064-2015. https://doi.org/10.1183/23120541.00064-2015
https://doi.org/10.1183/23120541.00064-2...
However, there is still no consensus in the literature and it is questionable whether there is robust scientific evidence that MRSA eradication is beneficial for CF patients.122122 Vanderhelst E, De Wachter E, Willekens J, Schuermans D, Vincken W, Malfroot A, et al. Increase in ventilated air spaces after eradication of chronic methicillin-resistant Staphylococcus aureus infection in cystic fibrosis patients. Acta Clin Belg. 2015;70(1):30-33. https://doi.org/10.1179/2295333714Y.0000000079
https://doi.org/10.1179/2295333714Y.0000...
,123123 Dolce D, Neri S, Grisotto L, Campana S, Ravenni N, Miselli F, et al. Methicillin-resistant Staphylococcus aureus eradication in cystic fibrosis patients: A randomized multicenter study. PLoS One. 2019;14(3):e0213497. https://doi.org/10.1371/journal.pone.0213497
https://doi.org/10.1371/journal.pone.021...

Evidence

Using the methodology described, we selected 8 studies (Figure S6 and Chart S7).120120 Hall H, Gadhok R, Alshafi K, Bilton D, Simmonds NJ. Eradication of respiratory tract MRSA at a large adult cystic fibrosis centre. Respir Med. 2015;109(3):357-363. https://doi.org/10.1016/j.rmed.2015.01.013
https://doi.org/10.1016/j.rmed.2015.01.0...

121 Vallières E, Rendall JC, Moore JE, McCaughan J, Hoeritzauer AI, Tunney MM, et al. MRSA eradication of newly acquired lower respiratory tract infection in cystic fibrosis. ERJ Open Res. 2016;2(1):00064-2015. https://doi.org/10.1183/23120541.00064-2015
https://doi.org/10.1183/23120541.00064-2...

122 Vanderhelst E, De Wachter E, Willekens J, Schuermans D, Vincken W, Malfroot A, et al. Increase in ventilated air spaces after eradication of chronic methicillin-resistant Staphylococcus aureus infection in cystic fibrosis patients. Acta Clin Belg. 2015;70(1):30-33. https://doi.org/10.1179/2295333714Y.0000000079
https://doi.org/10.1179/2295333714Y.0000...

123 Dolce D, Neri S, Grisotto L, Campana S, Ravenni N, Miselli F, et al. Methicillin-resistant Staphylococcus aureus eradication in cystic fibrosis patients: A randomized multicenter study. PLoS One. 2019;14(3):e0213497. https://doi.org/10.1371/journal.pone.0213497
https://doi.org/10.1371/journal.pone.021...

124 Garske LA, Kidd TJ, Gan R, Bunting JP, Franks CA, Coulter C, et al. Rifampicin and sodium fusidate reduces the frequency of methicillin-resistant Staphylococcus aureus (MRSA) isolation in adults with cystic fibrosis and chronic MRSA infection. J Hosp Infect. 2004;56(3):208-214. https://doi.org/10.1016/j.jhin.2003.12.003
https://doi.org/10.1016/j.jhin.2003.12.0...

125 Kappler M, Nagel F, Feilcke M, Kröner C, Pawlita I, Naehrig S, et al. Eradication of methicillin resistant Staphylococcus aureus detected for the first time in cystic fibrosis: A single center observational study. Pediatr Pulmonol. 2016;51(10):1010-1019. https://doi.org/10.1002/ppul.23519
https://doi.org/10.1002/ppul.23519...

126 Muhlebach MS, Beckett V, Popowitch E, Miller MB, Baines A, Mayer-Hamblett N, et al. Microbiological efficacy of early MRSA treatment in cystic fibrosis in a randomised controlled trial. Thorax. 2017;72(4):318-326. https://doi.org/10.1136/thoraxjnl-2016-208949
https://doi.org/10.1136/thoraxjnl-2016-2...
-127127 Solís A, Brown D, Hughes J, Van Saene HK, Heaf DP. Methicillin-resistant Staphylococcus aureus in children with cystic fibrosis: An eradication protocol. Pediatr Pulmonol. 2003;36(3):189-195. https://doi.org/10.1002/ppul.10231
https://doi.org/10.1002/ppul.10231...
Of those, only 2 are RCTs123123 Dolce D, Neri S, Grisotto L, Campana S, Ravenni N, Miselli F, et al. Methicillin-resistant Staphylococcus aureus eradication in cystic fibrosis patients: A randomized multicenter study. PLoS One. 2019;14(3):e0213497. https://doi.org/10.1371/journal.pone.0213497
https://doi.org/10.1371/journal.pone.021...
,126126 Muhlebach MS, Beckett V, Popowitch E, Miller MB, Baines A, Mayer-Hamblett N, et al. Microbiological efficacy of early MRSA treatment in cystic fibrosis in a randomised controlled trial. Thorax. 2017;72(4):318-326. https://doi.org/10.1136/thoraxjnl-2016-208949
https://doi.org/10.1136/thoraxjnl-2016-2...
and the other 6 are observational studies.120120 Hall H, Gadhok R, Alshafi K, Bilton D, Simmonds NJ. Eradication of respiratory tract MRSA at a large adult cystic fibrosis centre. Respir Med. 2015;109(3):357-363. https://doi.org/10.1016/j.rmed.2015.01.013
https://doi.org/10.1016/j.rmed.2015.01.0...

121 Vallières E, Rendall JC, Moore JE, McCaughan J, Hoeritzauer AI, Tunney MM, et al. MRSA eradication of newly acquired lower respiratory tract infection in cystic fibrosis. ERJ Open Res. 2016;2(1):00064-2015. https://doi.org/10.1183/23120541.00064-2015
https://doi.org/10.1183/23120541.00064-2...
-122122 Vanderhelst E, De Wachter E, Willekens J, Schuermans D, Vincken W, Malfroot A, et al. Increase in ventilated air spaces after eradication of chronic methicillin-resistant Staphylococcus aureus infection in cystic fibrosis patients. Acta Clin Belg. 2015;70(1):30-33. https://doi.org/10.1179/2295333714Y.0000000079
https://doi.org/10.1179/2295333714Y.0000...
,124124 Garske LA, Kidd TJ, Gan R, Bunting JP, Franks CA, Coulter C, et al. Rifampicin and sodium fusidate reduces the frequency of methicillin-resistant Staphylococcus aureus (MRSA) isolation in adults with cystic fibrosis and chronic MRSA infection. J Hosp Infect. 2004;56(3):208-214. https://doi.org/10.1016/j.jhin.2003.12.003
https://doi.org/10.1016/j.jhin.2003.12.0...
,125125 Kappler M, Nagel F, Feilcke M, Kröner C, Pawlita I, Naehrig S, et al. Eradication of methicillin resistant Staphylococcus aureus detected for the first time in cystic fibrosis: A single center observational study. Pediatr Pulmonol. 2016;51(10):1010-1019. https://doi.org/10.1002/ppul.23519
https://doi.org/10.1002/ppul.23519...
,127127 Solís A, Brown D, Hughes J, Van Saene HK, Heaf DP. Methicillin-resistant Staphylococcus aureus in children with cystic fibrosis: An eradication protocol. Pediatr Pulmonol. 2003;36(3):189-195. https://doi.org/10.1002/ppul.10231
https://doi.org/10.1002/ppul.10231...

For each of the articles, the quality of evidence for this question is summarized in Tables S6A and S6B. A detailed description of the findings can be found in the Supplementary Material (Question S6).

Recommendation

For CF patients, we do not have enough evidence to recommend or not recommend the use of MRSA eradication therapy.

Comments

Although all of the studies assessed the eradication rate, they employed different treatment protocols, as well as different means of assessing eradication (short- vs. long-term follow-up). All of the studies evaluated had sample sizes of less than 70 individuals. It is possible that further studies, especially studies with larger patient samples, will result in a change in the confidence level of this recommendation.

Question 7. Should we recommend nebulized dornase alfa for CF patients ≥ 6 years of age?

In CF patients, chronic inflammation and infection results in extracellular DNA from leukocytes being constantly released into the airways and accumulating in lung secretions.1818 Goss CH, MacNeill SJ, Quinton HB, Marshall BC, Elbert A, Knapp EA, et al. Children and young adults with CF in the USA have better lung function compared with the UK. Thorax. 2015;70(3):229-236. https://doi.org/10.1136/thoraxjnl-2014-205718
https://doi.org/10.1136/thoraxjnl-2014-2...
Consequently, there is an increase in the viscosity and adhesion of mucus. Dornase alfa is an enzyme capable of cleaving the extracellular DNA contained in mucus, reducing its viscosity and promoting greater clearance of secretions.128128 Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
https://doi.org/10.1002/14651858.CD00112...

Dornase alfa is administered by inhalation, at the usual dose of 2.5 mg once a day, and should be used in conjunction with other airway clearance techniques.1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,128128 Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
https://doi.org/10.1002/14651858.CD00112...

In phase I and II studies, the use of nebulized dornase alfa in CF patients proved to be safe and led to an increase in FEV1 in the short term, together with improvements in symptoms and quality of life.129129 Aitken ML, Burke W, McDonald G, Shak S, Montgomery AB, Smith A. Recombinant human DNase inhalation in normal subjects and patients with cystic fibrosis. A phase 1 study. JAMA. 1992;267(14):1947-1951. https://doi.org/10.1001/jama.1992.03480140073036
https://doi.org/10.1001/jama.1992.034801...
,130130 Hubbard RC, McElvaney NG, Birrer P, Shak S, Robinson WW, Jolley C, et al. A preliminary study of aerosolized recombinant human deoxyribonuclease I in the treatment of cystic fibrosis. N Engl J Med. 1992;326(12):812-815. https://doi.org/10.1056/NEJM199203193261207
https://doi.org/10.1056/NEJM199203193261...
Subsequent studies demonstrated the maintenance of benefits in the long term, with sustained improvement in FEV1, a reduced risk of exacerbations, and a good safety profile.128128 Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
https://doi.org/10.1002/14651858.CD00112...

Evidence

We selected 32 studies,1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,131131 Amin R, Subbarao P, Lou W, Jabar A, Balkovec S, Jensen R, et al. The effect of dornase alfa on ventilation inhomogeneity in patients with cystic fibrosis. Eur Respir J. 2011;37(4):806-812. https://doi.org/10.1183/09031936.00072510
https://doi.org/10.1183/09031936.0007251...

132 Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, et al. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011;38(6):1328-1335. https://doi.org/10.1183/09031936.00006211
https://doi.org/10.1183/09031936.0000621...

133 Barker M, Franke E, Böhle M, Pfannenstiel C, Heimann G. Effect of DNase on exercise capacity in cystic fibrosis. Pediatr Pulmonol. 2004;38(1):70-74. https://doi.org/10.1002/ppul.20048
https://doi.org/10.1002/ppul.20048...

134 Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, Assael BM, Merkus PJ, Tiddens HA. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011 Dec;38(6):1328-35. https://doi.org/10.1183/09031936.00006211
https://doi.org/10.1183/09031936.0000621...

135 Böllert FG, Paton JY, Marshall TG, Calvert J, Greening AP, Innes JA. Recombinant DNase in cystic fibrosis: a protocol for targeted introduction through n-of-1 trials. Scottish Cystic Fibrosis Group. Eur Respir J. 1999;13(1):107-113. https://doi.org/10.1183/09031936.99.13105399
https://doi.org/10.1183/09031936.99.1310...

136 Fiel SB, Fuchs HJ, Johnson C, Gonda I, Clark AR. Comparison of three jet nebulizer aerosol delivery systems used to administer recombinant human DNase I to patients with cystic fibrosis. The Pulmozyme rhDNase Study Group. Chest. 1995;108(1):153-156. https://doi.org/10.1378/chest.108.1.153
https://doi.org/10.1378/chest.108.1.153...

137 Frederiksen B, Pressler T, Hansen A, Koch C, Høiby N. Effect of aerosolized rhDNase (Pulmozyme) on pulmonary colonization in patients with cystic fibrosis. Acta Paediatr. 2006;95(9):1070-1074. https://doi.org/10.1080/08035250600752466
https://doi.org/10.1080/0803525060075246...

138 Furuya ME, Lezana-Fernández JL, Vargas MH, Hernández-Sierra JF, Ramírez-Figueroa JL. Efficacy of human recombinant DNase in pediatric patients with cystic fibrosis. Arch Med Res. 2001;32(1):30-34. https://doi.org/10.1016/S0188-4409(00)00258-7
https://doi.org/10.1016/S0188-4409(00)00...

139 Heijerman HG, van Rossem RN, Bakker W. Effect of rhDNase on lung function and quality of life in adult cystic fibrosis patients. Neth J Med. 1995;46(6):293-297. https://doi.org/10.1016/0300-2977(95)00022-F
https://doi.org/10.1016/0300-2977(95)000...

140 Hodson M. Multicenter study of rhDNase in cystic fibrosis with severe pulmonary involvement [Article in French]. Arch Pediatr. 1995;2(7):679-681. https://doi.org/10.1016/0929-693X(96)81226-7
https://doi.org/10.1016/0929-693X(96)812...

141 McCoy K, Hamilton S, Johnson C. Effects of 12-week administration of dornase alfa in patients with advanced cystic fibrosis lung disease. Pulmozyme Study Group. Chest. 1996;110(4):889-895. https://doi.org/10.1378/chest.110.4.889
https://doi.org/10.1378/chest.110.4.889...

142 Minasian C, Wallis C, Metcalfe C, Bush A. Comparison of inhaled mannitol, daily rhDNase and a combination of both in children with cystic fibrosis: a randomised trial. Thorax. 2010;65(1):51-56. https://doi.org/10.1136/thx.2009.116970
https://doi.org/10.1136/thx.2009.116970...

143 Quan JM, Tiddens HA, Sy JP, McKenzie SG, Montgomery MD, Robinson PJ, et al. A two-year randomized, placebo-controlled trial of dornase alfa in young patients with cystic fibrosis with mild lung function abnormalities. J Pediatr. 2001;139(6):813-820. https://doi.org/10.1067/mpd.2001.118570
https://doi.org/10.1067/mpd.2001.118570...

144 Robinson TE, Goris ML, Zhu HJ, Chen X, Bhise P, Sheikh F, et al. Dornase alfa reduces air trapping in children with mild cystic fibrosis lung disease: a quantitative analysis. Chest. 2005;128(4):2327-2335. https://doi.org/10.1378/chest.128.4.2327
https://doi.org/10.1378/chest.128.4.2327...

145 Robinson M, Hemming AL, Moriarty C, Eberl S, Bye PT. Effect of a short course of rhDNase on cough and mucociliary clearance in patients with cystic fibrosis. Pediatr Pulmonol. 2000;30(1):16-24. https://doi.org/10.1002/1099-0496(200007)30:1<16::AID-PPUL4>3.0.CO;2-H
https://doi.org/10.1002/1099-0496(200007...

146 Shah PL, Scott SF, Knight RA, Marriott C, Ranasinha C, Hodson ME. In vivo effects of recombinant human DNase I on sputum in patients with cystic fibrosis. Thorax. 1996;51(2):119-125. https://doi.org/10.1136/thx.51.2.119
https://doi.org/10.1136/thx.51.2.119...

147 Suri R, Metcalfe C, Lees B, Grieve R, Flather M, Normand C, et al. Comparison of hypertonic saline and alternate-day or daily recombinant human deoxyribonuclease in children with cystic fibrosis: a randomised trial. Lancet. 2001;358(9290):1316-1321. https://doi.org/10.1016/S0140-6736(01)06412-1
https://doi.org/10.1016/S0140-6736(01)06...

148 Bonestroo HJ, Slieker MG, Arets HG. No positive effect of rhdnase on the pulmonary colonization in children with cystic fibrosis. Monaldi Arch Chest Dis. 2010;73(1):12-17. https://doi.org/10.4081/monaldi.2010.308
https://doi.org/10.4081/monaldi.2010.308...

149 Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr. 2000;159(3):176-181. https://doi.org/10.1007/s004310050046
https://doi.org/10.1007/s004310050046...

150 Davies J, Trindade MT, Wallis C, Rosenthal M, Crawford O, Bush A. Retrospective review of the effects of rhDNase in children with cystic fibrosis. Pediatr Pulmonol. 1997;23(4):243-248. https://doi.org/10.1002/(SICI)1099-0496(199704)23:4<243::AID-PPUL1>3.0.CO;2-N
https://doi.org/10.1002/(SICI)1099-0496(...

151 Derelle J, Bertolo-Houriez E, Marchal F, Weber M, Virion JM, Vidailhet M. Respiratory evolution of patient with mucoviscidosis treated with mucolytic agents plus dornase alfa [Article in French]. Arch Pediatr. 1998;5(4):371-377. https://doi.org/10.1016/S0929-693X(98)80022-5
https://doi.org/10.1016/S0929-693X(98)80...

152 Harms HK, Matouk E, Tournier G, von der Hardt H, Weller PH, Romano L, et al. Multicenter, open-label study of recombinant human DNase in cystic fibrosis patients with moderate lung disease. DNase International Study Group. Pediatr Pulmonol. 1998;26(3):155-161. https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
https://doi.org/10.1002/(SICI)1099-0496(...

153 Hodson ME, McKenzie S, Harms HK, Koch C, Mastella G, Navarro J, et al. Dornase alfa in the treatment of cystic fibrosis in Europe: a report from the Epidemiologic Registry of Cystic Fibrosis. Pediatr Pulmonol. 2003;36(5):427-432. https://doi.org/10.1002/ppul.10348
https://doi.org/10.1002/ppul.10348...

154 Johnson CA, Butler SM, Konstan MW, Breen TJ, Morgan WJ. Estimating effectiveness in an observational study: a case study of dornase alfa in cystic fibrosis. The Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. J Pediatr. 1999;134(6):734-739. https://doi.org/10.1016/S0022-3476(99)70290-8
https://doi.org/10.1016/S0022-3476(99)70...

155 Konstan MW, Wagener JS, Pasta DJ, Millar SJ, Jacobs JR, Yegin A, et al. Clinical use of dornase alpha is associated with a slower rate of FEV1 decline in cystic fibrosis. Pediatr Pulmonol. 2011;46(6):545-553. https://doi.org/10.1002/ppul.21388
https://doi.org/10.1002/ppul.21388...

156 Milla CE. Long-term effects of aerosolised rhDNase on pulmonary disease progression in patients with cystic fibrosis. Thorax. 1998;53(12):1014-1017. https://doi.org/10.1136/thx.53.12.1014
https://doi.org/10.1136/thx.53.12.1014...

157 Rozov T, de Oliveira VZ, Santana MA, Adde FV, Mendes RH, Paschoal IA, et al. Dornase alfa improves the health-related quality of life among Brazilian patients with cystic fibrosis--a one-year prospective study. Pediatr Pulmonol. 2010;45(9):874-882. https://doi.org/10.1002/ppul.21267
https://doi.org/10.1002/ppul.21267...

158 Rozov T, Silva FA, Santana MA, Adde FV, Mendes RH; Brazilian Cystic Fibrosis Multicenter Study Group. A first-year dornase alfa treatment impact on clinical parameters of patients with cystic fibrosis: the Brazilian cystic fibrosis multicenter study. Rev Paul Pediatr. 2013;31(4):420-430. https://doi.org/10.1590/S0103-05822013000400002
https://doi.org/10.1590/S0103-0582201300...

159 Shah PL, Conway S, Scott SF, Rainisio M, Wildman M, Stableforth D, et al. A case-controlled study with dornase alfa to evaluate impact on disease progression over a 4-year period. Respiration. 2001;68(2):160-164. https://doi.org/10.1159/000050486
https://doi.org/10.1159/000050486...

160 Shah PL, Scott SF, Geddes DM, Hodson ME. Two years experience with recombinant human DNase I in the treatment of pulmonary disease in cystic fibrosis. Respir Med. 1995;89(7):499-502. https://doi.org/10.1016/0954-6111(95)90126-4
https://doi.org/10.1016/0954-6111(95)901...
-161161 Wizla-Derambure N, Michaud L, Sardet A, Deschildre A, Loeuille GA, Tassin E, et al. Effect of rhDNase on the respiratory function and nutritional status of children and adolescents with mucoviscidosis [Article in French]. Arch Pediatr. 1998;5(4):378-383. https://doi.org/10.1016/S0929-693X(98)80023-7
https://doi.org/10.1016/S0929-693X(98)80...
18 of which were RCTs that compared the use of dornase alfa with placebo,1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,131131 Amin R, Subbarao P, Lou W, Jabar A, Balkovec S, Jensen R, et al. The effect of dornase alfa on ventilation inhomogeneity in patients with cystic fibrosis. Eur Respir J. 2011;37(4):806-812. https://doi.org/10.1183/09031936.00072510
https://doi.org/10.1183/09031936.0007251...

132 Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, et al. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011;38(6):1328-1335. https://doi.org/10.1183/09031936.00006211
https://doi.org/10.1183/09031936.0000621...

133 Barker M, Franke E, Böhle M, Pfannenstiel C, Heimann G. Effect of DNase on exercise capacity in cystic fibrosis. Pediatr Pulmonol. 2004;38(1):70-74. https://doi.org/10.1002/ppul.20048
https://doi.org/10.1002/ppul.20048...

134 Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, Assael BM, Merkus PJ, Tiddens HA. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011 Dec;38(6):1328-35. https://doi.org/10.1183/09031936.00006211
https://doi.org/10.1183/09031936.0000621...
-135135 Böllert FG, Paton JY, Marshall TG, Calvert J, Greening AP, Innes JA. Recombinant DNase in cystic fibrosis: a protocol for targeted introduction through n-of-1 trials. Scottish Cystic Fibrosis Group. Eur Respir J. 1999;13(1):107-113. https://doi.org/10.1183/09031936.99.13105399
https://doi.org/10.1183/09031936.99.1310...
,140140 Hodson M. Multicenter study of rhDNase in cystic fibrosis with severe pulmonary involvement [Article in French]. Arch Pediatr. 1995;2(7):679-681. https://doi.org/10.1016/0929-693X(96)81226-7
https://doi.org/10.1016/0929-693X(96)812...

141 McCoy K, Hamilton S, Johnson C. Effects of 12-week administration of dornase alfa in patients with advanced cystic fibrosis lung disease. Pulmozyme Study Group. Chest. 1996;110(4):889-895. https://doi.org/10.1378/chest.110.4.889
https://doi.org/10.1378/chest.110.4.889...
-142142 Minasian C, Wallis C, Metcalfe C, Bush A. Comparison of inhaled mannitol, daily rhDNase and a combination of both in children with cystic fibrosis: a randomised trial. Thorax. 2010;65(1):51-56. https://doi.org/10.1136/thx.2009.116970
https://doi.org/10.1136/thx.2009.116970...
,144144 Robinson TE, Goris ML, Zhu HJ, Chen X, Bhise P, Sheikh F, et al. Dornase alfa reduces air trapping in children with mild cystic fibrosis lung disease: a quantitative analysis. Chest. 2005;128(4):2327-2335. https://doi.org/10.1378/chest.128.4.2327
https://doi.org/10.1378/chest.128.4.2327...
,145145 Robinson M, Hemming AL, Moriarty C, Eberl S, Bye PT. Effect of a short course of rhDNase on cough and mucociliary clearance in patients with cystic fibrosis. Pediatr Pulmonol. 2000;30(1):16-24. https://doi.org/10.1002/1099-0496(200007)30:1<16::AID-PPUL4>3.0.CO;2-H
https://doi.org/10.1002/1099-0496(200007...
,149149 Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr. 2000;159(3):176-181. https://doi.org/10.1007/s004310050046
https://doi.org/10.1007/s004310050046...
,151151 Derelle J, Bertolo-Houriez E, Marchal F, Weber M, Virion JM, Vidailhet M. Respiratory evolution of patient with mucoviscidosis treated with mucolytic agents plus dornase alfa [Article in French]. Arch Pediatr. 1998;5(4):371-377. https://doi.org/10.1016/S0929-693X(98)80022-5
https://doi.org/10.1016/S0929-693X(98)80...

152 Harms HK, Matouk E, Tournier G, von der Hardt H, Weller PH, Romano L, et al. Multicenter, open-label study of recombinant human DNase in cystic fibrosis patients with moderate lung disease. DNase International Study Group. Pediatr Pulmonol. 1998;26(3):155-161. https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
https://doi.org/10.1002/(SICI)1099-0496(...

153 Hodson ME, McKenzie S, Harms HK, Koch C, Mastella G, Navarro J, et al. Dornase alfa in the treatment of cystic fibrosis in Europe: a report from the Epidemiologic Registry of Cystic Fibrosis. Pediatr Pulmonol. 2003;36(5):427-432. https://doi.org/10.1002/ppul.10348
https://doi.org/10.1002/ppul.10348...
-154154 Johnson CA, Butler SM, Konstan MW, Breen TJ, Morgan WJ. Estimating effectiveness in an observational study: a case study of dornase alfa in cystic fibrosis. The Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. J Pediatr. 1999;134(6):734-739. https://doi.org/10.1016/S0022-3476(99)70290-8
https://doi.org/10.1016/S0022-3476(99)70...
,159159 Shah PL, Conway S, Scott SF, Rainisio M, Wildman M, Stableforth D, et al. A case-controlled study with dornase alfa to evaluate impact on disease progression over a 4-year period. Respiration. 2001;68(2):160-164. https://doi.org/10.1159/000050486
https://doi.org/10.1159/000050486...
,160160 Shah PL, Scott SF, Geddes DM, Hodson ME. Two years experience with recombinant human DNase I in the treatment of pulmonary disease in cystic fibrosis. Respir Med. 1995;89(7):499-502. https://doi.org/10.1016/0954-6111(95)90126-4
https://doi.org/10.1016/0954-6111(95)901...
and 14 were observational studies,136136 Fiel SB, Fuchs HJ, Johnson C, Gonda I, Clark AR. Comparison of three jet nebulizer aerosol delivery systems used to administer recombinant human DNase I to patients with cystic fibrosis. The Pulmozyme rhDNase Study Group. Chest. 1995;108(1):153-156. https://doi.org/10.1378/chest.108.1.153
https://doi.org/10.1378/chest.108.1.153...

137 Frederiksen B, Pressler T, Hansen A, Koch C, Høiby N. Effect of aerosolized rhDNase (Pulmozyme) on pulmonary colonization in patients with cystic fibrosis. Acta Paediatr. 2006;95(9):1070-1074. https://doi.org/10.1080/08035250600752466
https://doi.org/10.1080/0803525060075246...

138 Furuya ME, Lezana-Fernández JL, Vargas MH, Hernández-Sierra JF, Ramírez-Figueroa JL. Efficacy of human recombinant DNase in pediatric patients with cystic fibrosis. Arch Med Res. 2001;32(1):30-34. https://doi.org/10.1016/S0188-4409(00)00258-7
https://doi.org/10.1016/S0188-4409(00)00...
-139139 Heijerman HG, van Rossem RN, Bakker W. Effect of rhDNase on lung function and quality of life in adult cystic fibrosis patients. Neth J Med. 1995;46(6):293-297. https://doi.org/10.1016/0300-2977(95)00022-F
https://doi.org/10.1016/0300-2977(95)000...
,143143 Quan JM, Tiddens HA, Sy JP, McKenzie SG, Montgomery MD, Robinson PJ, et al. A two-year randomized, placebo-controlled trial of dornase alfa in young patients with cystic fibrosis with mild lung function abnormalities. J Pediatr. 2001;139(6):813-820. https://doi.org/10.1067/mpd.2001.118570
https://doi.org/10.1067/mpd.2001.118570...
,146146 Shah PL, Scott SF, Knight RA, Marriott C, Ranasinha C, Hodson ME. In vivo effects of recombinant human DNase I on sputum in patients with cystic fibrosis. Thorax. 1996;51(2):119-125. https://doi.org/10.1136/thx.51.2.119
https://doi.org/10.1136/thx.51.2.119...

147 Suri R, Metcalfe C, Lees B, Grieve R, Flather M, Normand C, et al. Comparison of hypertonic saline and alternate-day or daily recombinant human deoxyribonuclease in children with cystic fibrosis: a randomised trial. Lancet. 2001;358(9290):1316-1321. https://doi.org/10.1016/S0140-6736(01)06412-1
https://doi.org/10.1016/S0140-6736(01)06...
-148148 Bonestroo HJ, Slieker MG, Arets HG. No positive effect of rhdnase on the pulmonary colonization in children with cystic fibrosis. Monaldi Arch Chest Dis. 2010;73(1):12-17. https://doi.org/10.4081/monaldi.2010.308
https://doi.org/10.4081/monaldi.2010.308...
,150150 Davies J, Trindade MT, Wallis C, Rosenthal M, Crawford O, Bush A. Retrospective review of the effects of rhDNase in children with cystic fibrosis. Pediatr Pulmonol. 1997;23(4):243-248. https://doi.org/10.1002/(SICI)1099-0496(199704)23:4<243::AID-PPUL1>3.0.CO;2-N
https://doi.org/10.1002/(SICI)1099-0496(...
,155155 Konstan MW, Wagener JS, Pasta DJ, Millar SJ, Jacobs JR, Yegin A, et al. Clinical use of dornase alpha is associated with a slower rate of FEV1 decline in cystic fibrosis. Pediatr Pulmonol. 2011;46(6):545-553. https://doi.org/10.1002/ppul.21388
https://doi.org/10.1002/ppul.21388...

156 Milla CE. Long-term effects of aerosolised rhDNase on pulmonary disease progression in patients with cystic fibrosis. Thorax. 1998;53(12):1014-1017. https://doi.org/10.1136/thx.53.12.1014
https://doi.org/10.1136/thx.53.12.1014...

157 Rozov T, de Oliveira VZ, Santana MA, Adde FV, Mendes RH, Paschoal IA, et al. Dornase alfa improves the health-related quality of life among Brazilian patients with cystic fibrosis--a one-year prospective study. Pediatr Pulmonol. 2010;45(9):874-882. https://doi.org/10.1002/ppul.21267
https://doi.org/10.1002/ppul.21267...
-158158 Rozov T, Silva FA, Santana MA, Adde FV, Mendes RH; Brazilian Cystic Fibrosis Multicenter Study Group. A first-year dornase alfa treatment impact on clinical parameters of patients with cystic fibrosis: the Brazilian cystic fibrosis multicenter study. Rev Paul Pediatr. 2013;31(4):420-430. https://doi.org/10.1590/S0103-05822013000400002
https://doi.org/10.1590/S0103-0582201300...
,161161 Wizla-Derambure N, Michaud L, Sardet A, Deschildre A, Loeuille GA, Tassin E, et al. Effect of rhDNase on the respiratory function and nutritional status of children and adolescents with mucoviscidosis [Article in French]. Arch Pediatr. 1998;5(4):378-383. https://doi.org/10.1016/S0929-693X(98)80023-7
https://doi.org/10.1016/S0929-693X(98)80...
as illustrated in Figure S7 and Chart S8.

For each of the articles, the quality of evidence for this question is summarized in Tables S7A and S7B. A detailed description of the findings can be found in the Supplementary Material (Question S7).

Recommendation

For CF patients, we suggest the use of inhaled dornase alfa (conditional recommendation, very low quality of evidence).

Comments

The differences in mortality rates between the intervention and placebo groups were not significant, because few patients died. Two RCTs evaluated adverse effects1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,149149 Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr. 2000;159(3):176-181. https://doi.org/10.1007/s004310050046
https://doi.org/10.1007/s004310050046...
and found no significant differences in terms of the frequency of adverse events. One RCT and two observational studies131131 Amin R, Subbarao P, Lou W, Jabar A, Balkovec S, Jensen R, et al. The effect of dornase alfa on ventilation inhomogeneity in patients with cystic fibrosis. Eur Respir J. 2011;37(4):806-812. https://doi.org/10.1183/09031936.00072510
https://doi.org/10.1183/09031936.0007251...
,155155 Konstan MW, Wagener JS, Pasta DJ, Millar SJ, Jacobs JR, Yegin A, et al. Clinical use of dornase alpha is associated with a slower rate of FEV1 decline in cystic fibrosis. Pediatr Pulmonol. 2011;46(6):545-553. https://doi.org/10.1002/ppul.21388
https://doi.org/10.1002/ppul.21388...
,156156 Milla CE. Long-term effects of aerosolised rhDNase on pulmonary disease progression in patients with cystic fibrosis. Thorax. 1998;53(12):1014-1017. https://doi.org/10.1136/thx.53.12.1014
https://doi.org/10.1136/thx.53.12.1014...
assessing quality of life demonstrated improvements in the intervention groups.

Nine RCTs,1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,131131 Amin R, Subbarao P, Lou W, Jabar A, Balkovec S, Jensen R, et al. The effect of dornase alfa on ventilation inhomogeneity in patients with cystic fibrosis. Eur Respir J. 2011;37(4):806-812. https://doi.org/10.1183/09031936.00072510
https://doi.org/10.1183/09031936.0007251...
,141141 McCoy K, Hamilton S, Johnson C. Effects of 12-week administration of dornase alfa in patients with advanced cystic fibrosis lung disease. Pulmozyme Study Group. Chest. 1996;110(4):889-895. https://doi.org/10.1378/chest.110.4.889
https://doi.org/10.1378/chest.110.4.889...
,142142 Minasian C, Wallis C, Metcalfe C, Bush A. Comparison of inhaled mannitol, daily rhDNase and a combination of both in children with cystic fibrosis: a randomised trial. Thorax. 2010;65(1):51-56. https://doi.org/10.1136/thx.2009.116970
https://doi.org/10.1136/thx.2009.116970...
,144144 Robinson TE, Goris ML, Zhu HJ, Chen X, Bhise P, Sheikh F, et al. Dornase alfa reduces air trapping in children with mild cystic fibrosis lung disease: a quantitative analysis. Chest. 2005;128(4):2327-2335. https://doi.org/10.1378/chest.128.4.2327
https://doi.org/10.1378/chest.128.4.2327...
,149149 Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr. 2000;159(3):176-181. https://doi.org/10.1007/s004310050046
https://doi.org/10.1007/s004310050046...
,152152 Harms HK, Matouk E, Tournier G, von der Hardt H, Weller PH, Romano L, et al. Multicenter, open-label study of recombinant human DNase in cystic fibrosis patients with moderate lung disease. DNase International Study Group. Pediatr Pulmonol. 1998;26(3):155-161. https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
https://doi.org/10.1002/(SICI)1099-0496(...
,154154 Johnson CA, Butler SM, Konstan MW, Breen TJ, Morgan WJ. Estimating effectiveness in an observational study: a case study of dornase alfa in cystic fibrosis. The Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. J Pediatr. 1999;134(6):734-739. https://doi.org/10.1016/S0022-3476(99)70290-8
https://doi.org/10.1016/S0022-3476(99)70...
,160160 Shah PL, Scott SF, Geddes DM, Hodson ME. Two years experience with recombinant human DNase I in the treatment of pulmonary disease in cystic fibrosis. Respir Med. 1995;89(7):499-502. https://doi.org/10.1016/0954-6111(95)90126-4
https://doi.org/10.1016/0954-6111(95)901...
evaluated collectively, showed that FEV1 values were 5% higher among patients receiving dornase alfa than among those receiving a placebo. Although that is a modest improvement, FEV1 is a proxy for mortality in population studies. In addition, 2 RCTs1717 Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
https://doi.org/10.1056/NEJM199409083311...
,152152 Harms HK, Matouk E, Tournier G, von der Hardt H, Weller PH, Romano L, et al. Multicenter, open-label study of recombinant human DNase in cystic fibrosis patients with moderate lung disease. DNase International Study Group. Pediatr Pulmonol. 1998;26(3):155-161. https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
https://doi.org/10.1002/(SICI)1099-0496(...
demonstrated that the rate of exacerbations was 7% lower in the intervention groups than in the placebo groups. This outcome is important, because a reduction in exacerbations is associated with favorable lung function outcomes and, indirectly, with lower mortality.

The search of the literature for these guidelines did not include studies involving children under 6 years of age. Therefore, it is not possible to extrapolate this recommendation to any younger age group.

Question 8. Should we recommend antimicrobial eradication treatment in CF patients with airway colonization by B. cepacia complex strains?

The B. cepacia complex comprises 22 species,162162 LiPuma JJ, Spilker T, Gill LH, Campbell PW 3rd, Liu L, Mahenthiralingam E. Disproportionate distribution of Burkholderia cepacia complex species and transmissibility markers in cystic fibrosis. Am J Respir Crit Care Med. 2001;164(1):92-96. https://doi.org/10.1164/ajrccm.164.1.2011153
https://doi.org/10.1164/ajrccm.164.1.201...
the most common of which in CF are B. multivorans and B. cenocepacia. The clinical picture is quite variable, ranging from chronic, oligosymptomatic infection to severe cases, with necrotizing pneumonia, respiratory failure, and sepsis (cepacia syndrome).163163 Zlosnik JE, Zhou G, Brant R, Henry DA, Hird TJ, Mahenthiralingam E, et al. Burkholderia species infections in patients with cystic fibrosis in British Columbia, Canada. 30 years' experience. Ann Am Thorac Soc. 2015;12(1):70-78. https://doi.org/10.1513/AnnalsATS.201408-395OC
https://doi.org/10.1513/AnnalsATS.201408...
The B. cepacia complex has a peculiar bacterial resistance profile, which makes the choice of antibiotic treatment difficult, and a combination of antimicrobial drugs is commonly suggested, preferably guided by antimicrobial susceptibility testing.164164 Horsley A, Webb K, Bright-Thomas R, Govan J, Jones A. Can early Burkholderia cepacia complex infection in cystic fibrosis be eradicated with antibiotic therapy?. Front Cell Infect Microbiol. 2011;1:18. https://doi.org/10.3389/fcimb.2011.00018
https://doi.org/10.3389/fcimb.2011.00018...

Evidence

Using the methodology described, we selected 3 studies,2323 Garcia BA, Carden JL, Goodwin DL, Smith TA, Gaggar A, Leon K, et al. Implementation of a successful eradication protocol for Burkholderia Cepacia complex in cystic fibrosis patients. BMC Pulm Med. 2018;18(1):35. https://doi.org/10.1186/s12890-018-0594-8
https://doi.org/10.1186/s12890-018-0594-...
,165165 Tullis DE, Burns JL, Retsch-Bogart GZ, Bresnik M, Henig NR, Lewis SA, et al. Inhaled aztreonam for chronic Burkholderia infection in cystic fibrosis: a placebo-controlled trial. J Cyst Fibros. 2014;13(3):296-305. https://doi.org/10.1016/j.jcf.2013.08.011
https://doi.org/10.1016/j.jcf.2013.08.01...
,166166 Uluer AZ, Waltz DA, Kalish LA, Adams S, Gerard C, Ericson DA. Inhaled amiloride and tobramycin solutions fail to eradicate Burkholderia dolosa in patients with cystic fibrosis. J Cyst Fibros. 2013;12(1):54-59. https://doi.org/10.1016/j.jcf.2012.06.006
https://doi.org/10.1016/j.jcf.2012.06.00...
as illustrated in Figure S8 and Chart S9. Only 1 study was characterized as an RCT.165165 Tullis DE, Burns JL, Retsch-Bogart GZ, Bresnik M, Henig NR, Lewis SA, et al. Inhaled aztreonam for chronic Burkholderia infection in cystic fibrosis: a placebo-controlled trial. J Cyst Fibros. 2014;13(3):296-305. https://doi.org/10.1016/j.jcf.2013.08.011
https://doi.org/10.1016/j.jcf.2013.08.01...

For each of the articles, the quality of evidence for this question is summarized in Tables S8A and S8B. A detailed description of the findings can be found in the Supplementary Material (Question S8).

Recommendation

For CF patients, we do not have enough evidence to recommend or not recommend the use of eradication therapy for B. cepacia complex.

Comments

None of the selected studies showed any benefits of eradication therapy for B. cepacia complex when evaluating the many critical outcomes considered important in the methodology of these guidelines. Only a few studies on the subject were found in the literature. Most of those studies had low methodological quality, and there was considerable heterogeneity among them. The eradication regimens described were not standardized. The higher rates of adverse events found with the use of inhaled aztreonam cannot be extrapolated to other therapeutic regimens, especially because that antibiotic is not commonly recommended for this type of infection. Therefore, our guideline committee decided that it is not possible to make a recommendation either for or against this therapy, given the scarcity of published information. Further studies are needed in order to evaluate this issue in greater detail.

FINAL CONSIDERATIONS

A summary of the recommendations for the pharmacological treatment of respiratory disease in CF is presented in Chart 5.

Chart 5
Summary of recommendations for pulmonary pharmacological treatment in cystic fibrosis.

It is up to the prescribers to determine which treatments are appropriate for a given patient or group of patients, and that it is up to them to help patients and their families make consistent and well-founded decisions, consistent with the strength of the recommendations and the quality of existing evidence. Cost analyses and pharmacoeconomic aspects were not considered in these recommendations.

Although there is still no drug with curative capacity in CF, the present guidelines suggest several interventions with potential benefits for the treatment of the disease. Some drugs, such as dornase alfa, and strategies to control chronic infection by P. aeruginosa have been approved for more than a decade and are considered standard therapy in the management of CF. However, those interventions work to control the consequences of the disease, such as increased mucus viscosity and recurrent infectious exacerbations. More recently, a new class of drugs, CFTR modulators, has initiated a new phase in the treatment of CF by acting on the underlying cause of the disease.99 Guimbellot J, Sharma J, Rowe SM. Toward inclusive therapy with CFTR modulators: Progress and challenges. Pediatr Pulmonol. 2017;52(S48):S4-S14. https://doi.org/10.1002/ppul.23773
https://doi.org/10.1002/ppul.23773...
Substantial gains in lung function and reduction in exacerbation rates were found when ivacaftor was used in patients with class III mutations1111 Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Drevinek P, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. https://doi.org/10.1056/NEJMoa1105185
https://doi.org/10.1056/NEJMoa1105185...
and when tezacaftor+ivacaftor was used in patients with residual function mutations.1414 Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
https://doi.org/10.1056/NEJMoa1709847...
However, despite the statistically significant benefits, such results were not achieved in CF patients homozygous for the F508del mutation who were treated with lumacaftor+ivacaftor1212 Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
https://doi.org/10.1056/NEJMoa1409547...
or tezacaftor+ivacaftor.1313 Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
https://doi.org/10.1056/NEJMoa1709846...
These data indicate the need for an adequate assessment of the benefits of each treatment according to the population evaluated. Treatment with CFTR modulators can be considered a targeted therapy, and CFTR modulators are chosen according to the action of different drugs on specific groups of CFTR mutations. A new triple combination of CFTR modulators (elexacaftor+tezacaftor+ivacaftor) appears to be an effective and safe option for patients with at least one F508del allele.1515 Heijerman HGM, McKone EF, Downey DG, Van Braeckel E, Rowe SM, Tullis E, et al. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial [published correction appears in Lancet. 2020 May 30;395(10238):1694]. Lancet. 2019;394(10212):1940-1948. https://doi.org/10.1016/S0140-6736(19)32597-8
https://doi.org/10.1016/S0140-6736(19)32...
,1616 Middleton PG, Mall MA, Drevínek P, Lands LC, McKone EF, Polineni D, et al. Elexacaftor-Tezacaftor-Ivacaftor for Cystic Fibrosis with a Single Phe508del Allele. N Engl J Med. 2019;381(19):1809-1819. https://doi.org/10.1056/NEJMoa1908639
https://doi.org/10.1056/NEJMoa1908639...

In view of the many advances in the management of CF in recent years, it is recommended that even proven effective treatments, such as dornase alfa and chronic P. aeruginosa suppression therapy, be reassessed in the future, given the greater access to CFTR modulators in eligible patients.

Regarding the other interventions evaluated, the expert panel was not able to issue an evidence-based recommendation because of the low quality of the evidence obtained for the eradication of P. aeruginosa, MRSA, and B. cepacia complex. More studies on the topic are needed in order to make a formal recommendation. However, clinical physicians must evaluate the particularities of each patient and recognize the possible benefits of these therapies in selected cases.

It should be borne in mind that these guidelines sought to answer questions regarding only eight of the main pharmacological interventions for the treatment of the respiratory consequences of CF. Other therapies, such as nebulization with hypertonic saline and long-term use of macrolides, were not evaluated in these guidelines. That does not mean that such therapies do not present clinical benefits or that they cannot be used according to their own eligibility criteria. Nonpharmacological care, including vaccination, physical activity, respiratory physiotherapy, and pulmonary rehabilitation, is also essential in the management of CF. Finally, because CF is a multisystemic disease, the patient must be cared for in a multidisciplinary way, involving gastrointestinal, endocrinological, and otolaryngological aspects that were not within the scope of these guidelines.77 Athanazio RA, Silva Filho LVRF, Vergara AA, Ribeiro AF, Riedi CA, Procianoy EDFA, et al. Brazilian guidelines for the diagnosis and treatment of cystic fibrosis. J Bras Pneumol. 2017;43(3):219-245. https://doi.org/10.1590/s1806-37562017000000065
https://doi.org/10.1590/s1806-3756201700...

It is important to clarify that the very low quality of evidence for some recommendations does not mean that they should not be considered or implemented. In these guidelines, it was decided that RCT and observational studies should be included, with the aim of better evaluating the effect of some therapies approved many years ago for CF. This strategy allows the evaluation of a large sample of patients with high external validity. However, it increases the uncertainty of the results because of the biases inherent to the different study designs. It is important to emphasize that the consistent finding of results in RCT and observational studies favors the clinical applicability of these interventions for a greater proportion of patients with CF. In addition, our recommendations are in line with those of other international guidelines.88 Castellani C, Duff AJA, Bell SC, Heijerman HGM, Munck A, Ratjen F, et al. ECFS best practice guidelines: the 2018 revision. J Cyst Fibros. 2018;17(2):153-178. https://doi.org/10.1016/j.jcf.2018.02.006
https://doi.org/10.1016/j.jcf.2018.02.00...

We believe that these guidelines constitute an important tool to be incorporated into the approach to patients with CF, mainly aiming to favor its management, as well as helping define public policies related to the disease.

ACKNOWLEDGMENTS

We would like to thank Dr. Anna Menezes and Dr. Fernando Wehrmeister for the methodological and statistical support during the development of this project.

REFERENCES

  • 1
    Ratjen F, Bell SC, Rowe SM, Goss CH, Quittner AL, Bush A. Cystic fibrosis. Nat Rev Dis Primers. 2015;1:15010. https://doi.org/10.1038/nrdp.2015.10
    » https://doi.org/10.1038/nrdp.2015.10
  • 2
    Bell SC, Mall MA, Gutierrez H, Macek M, Madge S, Davies JC, et al. The future of cystic fibrosis care: a global perspective [published correction appears in Lancet Respir Med. 2019 Dec;7(12):e40]. Lancet Respir Med. 2020;8(1):65-124. https://doi.org/10.1016/S2213-2600(19)30337-6
    » https://doi.org/10.1016/S2213-2600(19)30337-6
  • 3
    Elborn JS. CFTR Modulators: Deciding What Is Best for Individuals in an Era of Precision Medicine. Ann Am Thorac Soc. 2018;15(3):298-300. https://doi.org/10.1513/AnnalsATS.201712-951ED
    » https://doi.org/10.1513/AnnalsATS.201712-951ED
  • 4
    Habib AR, Kajbafzadeh M, Desai S, Yang CL, Skolnik K, Quon BS. A Systematic Review of the Clinical Efficacy and Safety of CFTR Modulators in Cystic Fibrosis. Sci Rep. 2019;9(1):7234. https://doi.org/10.1038/s41598-019-43652-2
    » https://doi.org/10.1038/s41598-019-43652-2
  • 5
    Flume PA, Robinson KA, O'Sullivan BP, Finder JD, Vender RL, Willey-Courand DB, et al. Cystic fibrosis pulmonary guidelines: airway clearance therapies. Respir Care. 2009;54(4):522-537.
  • 6
    Mogayzel PJ Jr, Naureckas ET, Robinson KA, Mueller G, Hadjiliadis D, Hoag JB, et al. Cystic fibrosis pulmonary guidelines. Chronic medications for maintenance of lung health. Am J Respir Crit Care Med. 2013;187(7):680-689. https://doi.org/10.1164/rccm.201207-1160OE
    » https://doi.org/10.1164/rccm.201207-1160OE
  • 7
    Athanazio RA, Silva Filho LVRF, Vergara AA, Ribeiro AF, Riedi CA, Procianoy EDFA, et al. Brazilian guidelines for the diagnosis and treatment of cystic fibrosis. J Bras Pneumol. 2017;43(3):219-245. https://doi.org/10.1590/s1806-37562017000000065
    » https://doi.org/10.1590/s1806-37562017000000065
  • 8
    Castellani C, Duff AJA, Bell SC, Heijerman HGM, Munck A, Ratjen F, et al. ECFS best practice guidelines: the 2018 revision. J Cyst Fibros. 2018;17(2):153-178. https://doi.org/10.1016/j.jcf.2018.02.006
    » https://doi.org/10.1016/j.jcf.2018.02.006
  • 9
    Guimbellot J, Sharma J, Rowe SM. Toward inclusive therapy with CFTR modulators: Progress and challenges. Pediatr Pulmonol. 2017;52(S48):S4-S14. https://doi.org/10.1002/ppul.23773
    » https://doi.org/10.1002/ppul.23773
  • 10
    Chaudary N. Triplet CFTR modulators: future prospects for treatment of cystic fibrosis. Ther Clin Risk Manag. 2018;14:2375-2383. https://doi.org/10.2147/TCRM.S147164
    » https://doi.org/10.2147/TCRM.S147164
  • 11
    Ramsey BW, Davies J, McElvaney NG, Tullis E, Bell SC, Drevinek P, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365(18):1663-1672. https://doi.org/10.1056/NEJMoa1105185
    » https://doi.org/10.1056/NEJMoa1105185
  • 12
    Wainwright CE, Elborn JS, Ramsey BW, Marigowda G, Huang X, Cipolli M, et al. Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del CFTR. N Engl J Med. 2015;373(3):220-231. https://doi.org/10.1056/NEJMoa1409547
    » https://doi.org/10.1056/NEJMoa1409547
  • 13
    Taylor-Cousar JL, Munck A, McKone EF, van der Ent CK, Moeller A, Simard C, et al. Tezacaftor-Ivacaftor in Patients with Cystic Fibrosis Homozygous for Phe508del. N Engl J Med. 2017;377(21):2013-2023. https://doi.org/10.1056/NEJMoa1709846
    » https://doi.org/10.1056/NEJMoa1709846
  • 14
    Rowe SM, Daines C, Ringshausen FC, Kerem E, Wilson J, Tullis E, et al. Tezacaftor-Ivacaftor in Residual-Function Heterozygotes with Cystic Fibrosis. N Engl J Med. 2017;377(21):2024-2035. https://doi.org/10.1056/NEJMoa1709847
    » https://doi.org/10.1056/NEJMoa1709847
  • 15
    Heijerman HGM, McKone EF, Downey DG, Van Braeckel E, Rowe SM, Tullis E, et al. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial [published correction appears in Lancet. 2020 May 30;395(10238):1694]. Lancet. 2019;394(10212):1940-1948. https://doi.org/10.1016/S0140-6736(19)32597-8
    » https://doi.org/10.1016/S0140-6736(19)32597-8
  • 16
    Middleton PG, Mall MA, Drevínek P, Lands LC, McKone EF, Polineni D, et al. Elexacaftor-Tezacaftor-Ivacaftor for Cystic Fibrosis with a Single Phe508del Allele. N Engl J Med. 2019;381(19):1809-1819. https://doi.org/10.1056/NEJMoa1908639
    » https://doi.org/10.1056/NEJMoa1908639
  • 17
    Fuchs HJ, Borowitz DS, Christiansen DH, Morris EM, Nash ML, Ramsey BW, et al. Effect of aerosolized recombinant human DNase on exacerbations of respiratory symptoms and on pulmonary function in patients with cystic fibrosis. The Pulmozyme Study Group. N Engl J Med. 1994;331(10):637-642. https://doi.org/10.1056/NEJM199409083311003
    » https://doi.org/10.1056/NEJM199409083311003
  • 18
    Goss CH, MacNeill SJ, Quinton HB, Marshall BC, Elbert A, Knapp EA, et al. Children and young adults with CF in the USA have better lung function compared with the UK. Thorax. 2015;70(3):229-236. https://doi.org/10.1136/thoraxjnl-2014-205718
    » https://doi.org/10.1136/thoraxjnl-2014-205718
  • 19
    Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
    » https://doi.org/10.1002/14651858.CD001127.pub5
  • 20
    Grupo Brasileiro de Estudos de Fibrose Cística (GBEFC) [homepage on the Internet]. Curitiba: GBEFC; c2023 [cited 2023 Feb 4]. Registro Brasileiro de Fibrose Cística 2019. [Adobe Acrobat document, 61p.]. Available from: http://portalgbefc.org.br/ckfinder/userfiles/files/REBRAFC_2019.pdf
    » http://portalgbefc.org.br/ckfinder/userfiles/files/REBRAFC_2019.pdf
  • 21
    Mogayzel PJ Jr, Naureckas ET, Robinson KA, Brady C, Guill M, Lahiri T, et al. Cystic Fibrosis Foundation pulmonary guideline. pharmacologic approaches to prevention and eradication of initial Pseudomonas aeruginosa infection. Ann Am Thorac Soc. 2014;11(10):1640-1650. https://doi.org/10.1513/AnnalsATS.201404-166OC
    » https://doi.org/10.1513/AnnalsATS.201404-166OC
  • 22
    Lo DK, Muhlebach MS, Smyth AR. Interventions for the eradication of meticillin-resistant Staphylococcus aureus (MRSA) in people with cystic fibrosis. Cochrane Database Syst Rev. 2018;7(7):CD009650. https://doi.org/10.1002/14651858.CD009650.pub4
    » https://doi.org/10.1002/14651858.CD009650.pub4
  • 23
    Garcia BA, Carden JL, Goodwin DL, Smith TA, Gaggar A, Leon K, et al. Implementation of a successful eradication protocol for Burkholderia Cepacia complex in cystic fibrosis patients. BMC Pulm Med. 2018;18(1):35. https://doi.org/10.1186/s12890-018-0594-8
    » https://doi.org/10.1186/s12890-018-0594-8
  • 24
    Sawicki GS, Tiddens H. Managing treatment complexity in cystic fibrosis: challenges and opportunities. Pediatr Pulmonol. 2012;47(6):523-533. https://doi.org/10.1002/ppul.22546
    » https://doi.org/10.1002/ppul.22546
  • 25
    Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336(7650):924-926. https://doi.org/10.1136/bmj.39489.470347.AD
    » https://doi.org/10.1136/bmj.39489.470347.AD
  • 26
    Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck-Ytter Y, Schünemann HJ; et al. What is "quality of evidence" and why is it important to clinicians?. BMJ. 2008;336(7651):995-998. https://doi.org/10.1136/bmj.39490.551019.BE
    » https://doi.org/10.1136/bmj.39490.551019.BE
  • 27
    Guyatt GH, Oxman AD, Kunz R, Falck-Ytter Y, Vist GE, Liberati A, et al. Going from evidence to recommendations [published correction appears in BMJ. 2008 Jun 21;336(7658): doi:10.1136/bmj.a402]. BMJ. 2008;336(7652):1049-1051. https://doi.org/10.1136/bmj.a402
  • 28
    Abou Alaiwa MH, Launspach JL, Grogan B, Carter S, Zabner J, Stoltz DA, et al. Ivacaftor-induced sweat chloride reductions correlate with increases in airway surface liquid pH in cystic fibrosis. JCI Insight. 2018;3(15):e121468. https://doi.org/10.1172/jci.insight.121468
    » https://doi.org/10.1172/jci.insight.121468
  • 29
    Accurso FJ, Rowe SM, Clancy JP, Boyle MP, Dunitz JM, Durie PR, et al. Effect of VX-770 in persons with cystic fibrosis and the G551D-CFTR mutation. N Engl J Med. 2010;363(21):1991-2003. https://doi.org/10.1056/NEJMoa0909825
    » https://doi.org/10.1056/NEJMoa0909825
  • 30
    Barry PJ, Plant BJ, Nair A, Bicknell S, Simmonds NJ, Bell NJ, et al. Effects of ivacaftor in patients with cystic fibrosis who carry the G551D mutation and have severe lung disease. Chest. 2014;146(1):152-158. https://doi.org/10.1378/chest.13-2397
    » https://doi.org/10.1378/chest.13-2397
  • 31
    Bessonova L, Volkova N, Higgins M, Bengtsson L, Tian S, Simard C, et al. Data from the US and UK cystic fibrosis registries support disease modification by CFTR modulation with ivacaftor. Thorax. 2018;73(8):731-740. https://doi.org/10.1136/thoraxjnl-2017-210394
    » https://doi.org/10.1136/thoraxjnl-2017-210394
  • 32
    Borowitz D, Lubarsky B, Wilschanski M, Munck A, Gelfond D, Bodewes F, et al. Nutritional Status Improved in Cystic Fibrosis Patients with the G551D Mutation After Treatment with Ivacaftor. Dig Dis Sci. 2016;61(1):198-207. https://doi.org/10.1007/s10620-015-3834-2
    » https://doi.org/10.1007/s10620-015-3834-2
  • 33
    Davies JC, Wainwright CE, Canny GJ, Chilvers MA, Howenstine MS, Munck A, et al. Efficacy and safety of ivacaftor in patients aged 6 to 11 years with cystic fibrosis with a G551D mutation. Am J Respir Crit Care Med. 2013;187(11):1219-1225. https://doi.org/10.1164/rccm.201301-0153OC
    » https://doi.org/10.1164/rccm.201301-0153OC
  • 34
    De Boeck K, Munck A, Walker S, Faro A, Hiatt P, Gilmartin G, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis and a non-G551D gating mutation. J Cyst Fibros. 2014;13(6):674-680. https://doi.org/10.1016/j.jcf.2014.09.005
    » https://doi.org/10.1016/j.jcf.2014.09.005
  • 35
    Donaldson SH, Laube BL, Corcoran TE, Bhambhvani P, Zeman K, Ceppe A, et al. Effect of ivacaftor on mucociliary clearance and clinical outcomes in cystic fibrosis patients with G551D-CFTR. JCI Insight. 2018;3(24):e122695. https://doi.org/10.1172/jci.insight.122695
    » https://doi.org/10.1172/jci.insight.122695
  • 36
    Edgeworth D, Keating D, Ellis M, Button B, Williams E, Clark D, et al. Improvement in exercise duration, lung function and well-being in G551D-cystic fibrosis patients: a double-blind, placebo-controlled, randomized, cross-over study with ivacaftor treatment. Clin Sci (Lond). 2017;131(15):2037-2045. https://doi.org/10.1042/CS20170995
    » https://doi.org/10.1042/CS20170995
  • 37
    Gomez-Pastrana D, Nwokoro C, McLean M, Brown S, Christiansen N, Pao CS. Real-world effectiveness of ivacaftor in children with cystic fibrosis and the G551D mutation [Article in Spanish]. An Pediatr (Engl Ed). 2019;90(3):148-156. https://doi.org/10.1016/j.anpedi.2018.05.022
    » https://doi.org/10.1016/j.anpedi.2018.05.022
  • 38
    Guerra L, D'Oria S, Favia M, Castellani S, Santostasi T, Polizzi AM, et al. CFTR-dependent chloride efflux in cystic fibrosis mononuclear cells is increased by ivacaftor therapy. Pediatr Pulmonol. 2017;52(7):900-908. https://doi.org/10.1002/ppul.23712
    » https://doi.org/10.1002/ppul.23712
  • 39
    Guimbellot J, Solomon GM, Baines A, Heltshe SL, VanDalfsen J, Joseloff E, et al. Effectiveness of ivacaftor in cystic fibrosis patients with non-G551D gating mutations. J Cyst Fibros. 2019;18(1):102-109. https://doi.org/10.1016/j.jcf.2018.04.004
    » https://doi.org/10.1016/j.jcf.2018.04.004
  • 40
    Harris JK, Wagner BD, Zemanick ET, Robertson CE, Stevens MJ, Heltshe SL, et al. Changes in Airway Microbiome and Inflammation with Ivacaftor Treatment in Patients with Cystic Fibrosis and the G551D Mutation. Ann Am Thorac Soc. 2020;17(2):212-220. https://doi.org/10.1513/AnnalsATS.201907-493OC
    » https://doi.org/10.1513/AnnalsATS.201907-493OC
  • 41
    Hebestreit H, Sauer-Heilborn A, Fischer R, Käding M, Mainz JG. Effects of ivacaftor on severely ill patients with cystic fibrosis carrying a G551D mutation. J Cyst Fibros. 2013;12(6):599-603. https://doi.org/10.1016/j.jcf.2013.05.006
    » https://doi.org/10.1016/j.jcf.2013.05.006
  • 42
    Hubert D, Dehillotte C, Munck A, David V, Baek J, Mely L, et al. Retrospective observational study of French patients with cystic fibrosis and a Gly551Asp-CFTR mutation after 1 and 2years of treatment with ivacaftor in a real-world setting. J Cyst Fibros. 2018;17(1):89-95. https://doi.org/10.1016/j.jcf.2017.07.001
    » https://doi.org/10.1016/j.jcf.2017.07.001
  • 43
    McKone EF, Borowitz D, Drevinek P, Griese M, Konstan MW, Wainwright C, et al. Long-term safety and efficacy of ivacaftor in patients with cystic fibrosis who have the Gly551Asp-CFTR mutation: a phase 3, open-label extension study (PERSIST). Lancet Respir Med. 2014;2(11):902-910. https://doi.org/10.1016/S2213-2600(14)70218-8
    » https://doi.org/10.1016/S2213-2600(14)70218-8
  • 44
    Moss RB, Flume PA, Elborn JS, Cooke J, Rowe SM, McColley SA, et al. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med. 2015;3(7):524-533. https://doi.org/10.1016/S2213-2600(15)00201-5
    » https://doi.org/10.1016/S2213-2600(15)00201-5
  • 45
    Quittner A, Suthoff E, Rendas-Baum R, Bayliss MS, Sermet-Gaudelus I, Castiglione B, et al. Effect of ivacaftor treatment in patients with cystic fibrosis and the G551D-CFTR mutation: patient-reported outcomes in the STRIVE randomized, controlled trial. Health Qual Life Outcomes. 2015;13:93. https://doi.org/10.1186/s12955-015-0293-6
    » https://doi.org/10.1186/s12955-015-0293-6
  • 46
    Ronan NJ, Einarsson GG, Twomey M, Mooney D, Mullane D, NiChroinin M, et al. CORK Study in Cystic Fibrosis: Sustained Improvements in Ultra-Low-Dose Chest CT Scores After CFTR Modulation With Ivacaftor. Chest. 2018;153(2):395-403. https://doi.org/10.1016/j.chest.2017.10.005
    » https://doi.org/10.1016/j.chest.2017.10.005
  • 47
    Rosenfeld M, Wainwright CE, Higgins M, Wang LT, McKee C, Campbell D, et al. Ivacaftor treatment of cystic fibrosis in children aged 12 to <24 months and with a CFTR gating mutation (ARRIVAL): a phase 3 single-arm study [published correction appears in Lancet Respir Med. 2018 Jul;6(7):e35] [published correction appears in Lancet Respir Med. 2019 Apr;7(4):e15]. Lancet Respir Med. 2018;6(7):545-553. https://doi.org/10.1016/S2213-2600(18)30202-9
    » https://doi.org/10.1016/S2213-2600(18)30202-9
  • 48
    Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, et al. Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med. 2014;190(2):175-184. https://doi.org/10.1164/rccm.201404-0703OC
    » https://doi.org/10.1164/rccm.201404-0703OC
  • 49
    Salvatore D, Carnovale V, Iacotucci P, Braggion C, Castellani C, Cimino G, et al. Effectivenesss of ivacaftor in severe cystic fibrosis patients and non-G551D gating mutations. Pediatr Pulmonol. 2019;54(9):1398-1403. https://doi.org/10.1002/ppul.24424
    » https://doi.org/10.1002/ppul.24424
  • 50
    Sheikh SI, Long FR, McCoy KS, Johnson T, Ryan-Wenger NA, Hayes D Jr. Computed tomography correlates with improvement with ivacaftor in cystic fibrosis patients with G551D mutation. J Cyst Fibros. 2015;14(1):84-89. https://doi.org/10.1016/j.jcf.2014.06.011
    » https://doi.org/10.1016/j.jcf.2014.06.011
  • 51
    Sheikh SI, Long FR, McCoy KS, Johnson T, Ryan-Wenger NA, Hayes D Jr. Ivacaftor improves appearance of sinus disease on computerised tomography in cystic fibrosis patients with G551D mutation. Clin Otolaryngol. 2015;40(1):16-21. https://doi.org/10.1111/coa.12310
    » https://doi.org/10.1111/coa.12310
  • 52
    Stallings VA, Sainath N, Oberle M, Bertolaso C, Schall JI. Energy Balance and Mechanisms of Weight Gain with Ivacaftor Treatment of Cystic Fibrosis Gating Mutations. J Pediatr. 2018;201:229-237.e4. https://doi.org/10.1016/j.jpeds.2018.05.018
    » https://doi.org/10.1016/j.jpeds.2018.05.018
  • 53
    Taylor-Cousar J, Niknian M, Gilmartin G, Pilewski JM; VX11-770-901 investigators. Effect of ivacaftor in patients with advanced cystic fibrosis and a G551D-CFTR mutation: Safety and efficacy in an expanded access program in the United States. J Cyst Fibros. 2016;15(1):116-122. https://doi.org/10.1016/j.jcf.2015.01.008
    » https://doi.org/10.1016/j.jcf.2015.01.008
  • 54
    van de Peppel IP, Doktorova M, Berkers G, de Jonge HR, Houwen RHJ, Verkade HJ, et al. IVACAFTOR restores FGF19 regulated bile acid homeostasis in cystic fibrosis patients with an S1251N or a G551D gating mutation. J Cyst Fibros. 2019;18(2):286-293. https://doi.org/10.1016/j.jcf.2018.09.001
    » https://doi.org/10.1016/j.jcf.2018.09.001
  • 55
    Volkova N, Moy K, Evans J, Campbell D, Tian S, Simard C, et al. Disease progression in patients with cystic fibrosis treated with ivacaftor: Data from national US and UK registries. J Cyst Fibros. 2020;19(1):68-79. https://doi.org/10.1016/j.jcf.2019.05.015
    » https://doi.org/10.1016/j.jcf.2019.05.015
  • 56
    Konstan MW, McKone EF, Moss RB, Marigowda G, Tian S, Waltz D, et al. Assessment of safety and efficacy of long-term treatment with combination lumacaftor and ivacaftor therapy in patients with cystic fibrosis homozygous for the F508del-CFTR mutation (PROGRESS): a phase 3, extension study. Lancet Respir Med. 2017;5(2):107-118. https://doi.org/10.1016/S2213-2600(16)30427-1
    » https://doi.org/10.1016/S2213-2600(16)30427-1
  • 57
    Ratjen F, Hug C, Marigowda G, Tian S, Huang X, Stanojevic S, et al. Efficacy and safety of lumacaftor and ivacaftor in patients aged 6-11 years with cystic fibrosis homozygous for F508del-CFTR: a randomised, placebo-controlled phase 3 trial [published correction appears in Lancet Respir Med. 2017 Aug;5(8):e28]. Lancet Respir Med. 2017;5(7):557-567. https://doi.org/10.1016/S2213-2600(17)30215-1
    » https://doi.org/10.1016/S2213-2600(17)30215-1
  • 58
    Burgel PR, Munck A, Durieu I, Chiron R, Mely L, Prevotat A, et al. Real-Life Safety and Real-Life Safety and Effectiveness of Lumacaftor-Ivacaftor in Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2020;201(2):188-197. https://doi.org/10.1164/rccm.201906-1227OC
    » https://doi.org/10.1164/rccm.201906-1227OC
  • 59
    Diab-Cáceres L, Girón-Moreno RM, Pastor-Sanz MT, Quintana-Gallego E, Delgado-Pecellín I, Blanco-Aparicio M, et al. Compassionate Use of Lumacaftor/Ivacaftor in Cystic Fibrosis: Spanish Experience. Arch Bronconeumol (Engl Ed). 2018;54(12):614-618. https://doi.org/10.1016/j.arbr.2018.05.020
    » https://doi.org/10.1016/j.arbr.2018.05.020
  • 60
    Graeber SY, Dopfer C, Naehrlich L, Gyulumyan L, Scheuermann H, Hirtz S, et al. Effects of Lumacaftor-Ivacaftor Therapy on Cystic Fibrosis Transmembrane Conductance Regulator Function in Phe508del Homozygous Patients with Cystic Fibrosis. Am J Respir Crit Care Med. 2018;197(11):1433-1442. https://doi.org/10.1164/rccm.201710-1983OC
    » https://doi.org/10.1164/rccm.201710-1983OC
  • 61
    Hubert D, Chiron R, Camara B, Grenet D, Prévotat A, Bassinet L, et al. Real-life initiation of lumacaftor/ivacaftor combination in adults with cystic fibrosis homozygous for the Phe508del CFTR mutation and severe lung disease. J Cyst Fibros. 2017;16(3):388-391. https://doi.org/10.1016/j.jcf.2017.03.003
    » https://doi.org/10.1016/j.jcf.2017.03.003
  • 62
    Jennings MT, Dezube R, Paranjape S, West NE, Hong G, Braun A, et al. An Observational Study of Outcomes and Tolerances in Patients with Cystic Fibrosis Initiated on Lumacaftor/Ivacaftor. Ann Am Thorac Soc. 2017;14(11):1662-1666. https://doi.org/10.1513/AnnalsATS.201701-058OC
    » https://doi.org/10.1513/AnnalsATS.201701-058OC
  • 63
    Masson A, Schneider-Futschik EK, Baatallah N, Nguyen-Khoa T, Girodon E, Hatton A, et al. Predictive factors for lumacaftor/ivacaftor clinical response. J Cyst Fibros. 2019;18(3):368-374. https://doi.org/10.1016/j.jcf.2018.12.011
    » https://doi.org/10.1016/j.jcf.2018.12.011
  • 64
    McNamara JJ, McColley SA, Marigowda G, Liu F, Tian S, Owen CA, et al. Safety, pharmacokinetics, and pharmacodynamics of lumacaftor and ivacaftor combination therapy in children aged 2-5 years with cystic fibrosis homozygous for F508del-CFTR: an open-label phase 3 study. Lancet Respir Med. 2019;7(4):325-335. https://doi.org/10.1016/S2213-2600(18)30460-0
    » https://doi.org/10.1016/S2213-2600(18)30460-0
  • 65
    Milla CE, Ratjen F, Marigowda G, Liu F, Waltz D, Rosenfeld M; et al. Lumacaftor/Ivacaftor in Patients Aged 6-11 Years with Cystic Fibrosis and Homozygous for F508del-CFTR. Am J Respir Crit Care Med. 2017;195(7):912-920. https://doi.org/10.1164/rccm.201608-1754OC
    » https://doi.org/10.1164/rccm.201608-1754OC
  • 66
    Murer C, Huber LC, Kurowski T, Hirt A, Robinson CA, Bürgi U, et al. First experience in Switzerland in Phe508del homozygous cystic fibrosis patients with end-stage pulmonary disease enrolled in a lumacaftor-ivacaftor therapy trial - preliminary results. Swiss Med Wkly. 2018;148:w14593. https://doi.org/10.4414/smw.2018.14593
    » https://doi.org/10.4414/smw.2018.14593
  • 67
    Taylor-Cousar JL, Jain M, Barto TL, Haddad T, Atkinson J, Tian S, et al. Lumacaftor/ivacaftor in patients with cystic fibrosis and advanced lung disease homozygous for F508del-CFTR. J Cyst Fibros. 2018;17(2):228-235. https://doi.org/10.1016/j.jcf.2017.09.012
    » https://doi.org/10.1016/j.jcf.2017.09.012
  • 68
    Tesell MA, Alper CJ, Bacon R, Greenwood BC, Lenz K, Jeffrey PL, et al. Effect of Lumacaftor/Ivacaftor on Pulmonary Exacerbation Rates in Members with Cystic Fibrosis in a Medicaid Population. J Manag Care Spec Pharm. 2019;25(9):1021-1025. https://doi.org/10.18553/jmcp.2019.25.9.1021
    » https://doi.org/10.18553/jmcp.2019.25.9.1021
  • 69
    Wark PAB, Cookson K, Thiruchelvam T, Brannan J, Dorahy DJ. Lumacaftor/ Ivacaftor improves exercise tolerance in patients with Cystic Fibrosis and severe airflow obstruction. BMC Pulm Med. 2019;19(1):106. https://doi.org/10.1186/s12890-019-0866-y
    » https://doi.org/10.1186/s12890-019-0866-y
  • 70
    Kosorok MR, Zeng L, West SE, Rock MJ, Splaingard ML, Laxova A, et al. Acceleration of lung disease in children with cystic fibrosis after Pseudomonas aeruginosa acquisition. Pediatr Pulmonol. 2001;32(4):277-287. https://doi.org/10.1002/ppul.2009.abs
    » https://doi.org/10.1002/ppul.2009.abs
  • 71
    Blanchard AC, Horton E, Stanojevic S, Taylor L, Waters V, Ratjen F. Effectiveness of a stepwise Pseudomonas aeruginosa eradication protocol in children with cystic fibrosis. J Cyst Fibros. 2017;16(3):395-400. https://doi.org/10.1016/j.jcf.2017.01.007
    » https://doi.org/10.1016/j.jcf.2017.01.007
  • 72
    Claude F, Rochat I, Hafen GM. No benefit of longer eradication therapy of Pseudomonas aeruginosa primoinfections in pediatric cystic fibrosis. BMC Res Notes. 2019;12(1):115. https://doi.org/10.1186/s13104-019-4157-8
    » https://doi.org/10.1186/s13104-019-4157-8
  • 73
    Davies JC, Sermet-Gaudelus I, Naehrlich L, Harris RS, Campbell D, Ahluwalia N, et al. A phase 3, double-blind, parallel-group study to evaluate the efficacy and safety of tezacaftor in combination with ivacaftor in participants 6 through 11 years of age with cystic fibrosis homozygous for F508del or heterozygous for the F508del-CFTR mutation and a residual function mutation. J Cyst Fibros. 2021;20(1):68-77. https://doi.org/10.1016/j.jcf.2020.07.023
    » https://doi.org/10.1016/j.jcf.2020.07.023
  • 74
    Donaldson SH, Pilewski JM, Griese M, Cooke J, Viswanathan L, Tullis E, et al. Tezacaftor/Ivacaftor in Subjects with Cystic Fibrosis and F508del/F508del-CFTR or F508del/G551D-CFTR. Am J Respir Crit Care Med. 2018;197(2):214-224. https://doi.org/10.1164/rccm.201704-0717OC
    » https://doi.org/10.1164/rccm.201704-0717OC
  • 75
    Walker S, Flume P, McNamara J, Solomon M, Chilvers M, Chmiel J, et al. A phase 3 study of tezacaftor in combination with ivacaftor in children aged 6 through 11 years with cystic fibrosis. J Cyst Fibros. 2019;18(5):708-713. https://doi.org/10.1016/j.jcf.2019.06.009
    » https://doi.org/10.1016/j.jcf.2019.06.009
  • 76
    Emerson J, Rosenfeld M, McNamara S, Ramsey B, Gibson RL. Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis. Pediatr Pulmonol. 2002;34(2):91-100. https://doi.org/10.1002/ppul.10127
    » https://doi.org/10.1002/ppul.10127
  • 77
    Cohen-Cymberknoh M, Gilead N, Gartner S, Rovira S, Blau H, Mussaffi H, et al. Eradication failure of newly acquired Pseudomonas aeruginosa isolates in cystic fibrosis. J Cyst Fibros. 2016;15(6):776-782. https://doi.org/10.1016/j.jcf.2016.04.006
    » https://doi.org/10.1016/j.jcf.2016.04.006
  • 78
    Emiralioglu N, Yalcin E, Meral A, Sener B, Dogru D, Ozcelik U, et al. The success of the different eradication therapy regimens for Pseudomonas aeruginosa in cystic fibrosis. J Clin Pharm Ther. 2016;41(4):419-423. https://doi.org/10.1111/jcpt.12407
    » https://doi.org/10.1111/jcpt.12407
  • 79
    Frederiksen B, Koch C, Høiby N. Antibiotic treatment of initial colonization with Pseudomonas aeruginosa postpones chronic infection and prevents deterioration of pulmonary function in cystic fibrosis. Pediatr Pulmonol. 1997;23(5):330-335. https://doi.org/10.1002/(SICI)1099-0496(199705)23:5<330::AID-PPUL4>3.0.CO;2-O
    » https://doi.org/10.1002/(SICI)1099-0496(199705)23:5<330::AID-PPUL4>3.0.CO;2-O
  • 80
    Giugno H, Castaños C, Lubatti A, Pinheiro JL, Hernández C, González Pena H. Early antibiotic treatment for eradication of initial infection by Pseudomonas aeruginosa in patients with cystic fibrosis. Arch Argent Pediatr. 2010;108(2):141-147. https://doi.org/10.1590/S0325-00752010000200009
    » https://doi.org/10.1590/S0325-00752010000200009
  • 81
    Hewer SCL, Smyth AR, Brown M, Jones AP, Hickey H, Kenna D, et al. Intravenous versus oral antibiotics for eradication of Pseudomonas aeruginosa in cystic fibrosis (TORPEDO-CF): a randomised controlled trial. Lancet Respir Med. 2020;8(10):975-986. https://doi.org/10.1016/S2213-2600(20)30331-3
    » https://doi.org/10.1016/S2213-2600(20)30331-3
  • 82
    Kenny SL, Shaw TD, Downey DG, Moore JE, Rendall JC, Elborn JS. Eradication of Pseudomonas aeruginosa in adults with cystic fibrosis. BMJ Open Respir Res. 2014;1(1):e000021. https://doi.org/10.1136/bmjresp-2014-000021
    » https://doi.org/10.1136/bmjresp-2014-000021
  • 83
    Mayer-Hamblett N, Kloster M, Rosenfeld M, Gibson RL, Retsch-Bogart GZ, Emerson J, et al. Impact of Sustained Eradication of New Pseudomonas aeruginosa Infection on Long-term Outcomes in Cystic Fibrosis. Clin Infect Dis. 2015;61(5):707-715. https://doi.org/10.1093/cid/civ377
    » https://doi.org/10.1093/cid/civ377
  • 84
    Proesmans M, Vermeulen F, Boulanger L, Verhaegen J, De Boeck K. Comparison of two treatment regimens for eradication of Pseudomonas aeruginosa infection in children with cystic fibrosis. J Cyst Fibros. 2013;12(1):29-34. https://doi.org/10.1016/j.jcf.2012.06.001
    » https://doi.org/10.1016/j.jcf.2012.06.001
  • 85
    Ratjen F, Munck A, Kho P, Angyalosi G; ELITE Study Group. Treatment of early Pseudomonas aeruginosa infection in patients with cystic fibrosis: the ELITE trial. Thorax. 2010;65(4):286-291. https://doi.org/10.1136/thx.2009.121657
    » https://doi.org/10.1136/thx.2009.121657
  • 86
    Ratjen F, Moeller A, McKinney ML, Asherova I, Alon N, Maykut R, et al. Eradication of early P. aeruginosa infection in children <7 years of age with cystic fibrosis: The early study. J Cyst Fibros. 2019;18(1):78-85. https://doi.org/10.1016/j.jcf.2018.04.002
    » https://doi.org/10.1016/j.jcf.2018.04.002
  • 87
    Taccetti G, Campana S, Festini F, Mascherini M, Döring G. Early eradication therapy against Pseudomonas aeruginosa in cystic fibrosis patients. Eur Respir J. 2005;26(3):458-461. https://doi.org/10.1183/09031936.05.00009605
    » https://doi.org/10.1183/09031936.05.00009605
  • 88
    Taccetti G, Bianchini E, Cariani L, Buzzetti R, Costantini D, Trevisan F, et al. Early antibiotic treatment for Pseudomonas aeruginosa eradication in patients with cystic fibrosis: a randomised multicentre study comparing two different protocols. Thorax. 2012;67(10):853-859. https://doi.org/10.1136/thoraxjnl-2011-200832
    » https://doi.org/10.1136/thoraxjnl-2011-200832
  • 89
    Treggiari MM, Retsch-Bogart G, Mayer-Hamblett N, Khan U, Kulich M, Kronmal R, et al. Comparative efficacy and safety of 4 randomized regimens to treat early Pseudomonas aeruginosa infection in children with cystic fibrosis. Arch Pediatr Adolesc Med. 2011;165(9):847-856. https://doi.org/10.1001/archpediatrics.2011.136
    » https://doi.org/10.1001/archpediatrics.2011.136
  • 90
    Valerius NH, Koch C, Høiby N. Prevention of chronic Pseudomonas aeruginosa colonisation in cystic fibrosis by early treatment. Lancet. 1991;338(8769):725-726. https://doi.org/10.1016/0140-6736(91)91446-2
    » https://doi.org/10.1016/0140-6736(91)91446-2
  • 91
    Wiesemann HG, Steinkamp G, Ratjen F, Bauernfeind A, Przyklenk B, Döring G, et al. Placebo-controlled, double-blind, randomized study of aerosolized tobramycin for early treatment of Pseudomonas aeruginosa colonization in cystic fibrosis. Pediatr Pulmonol. 1998;25(2):88-92. https://doi.org/10.1002/(SICI)1099-0496(199802)25:2<88::AID-PPUL3>3.0.CO;2-J
    » https://doi.org/10.1002/(SICI)
  • 92
    Lee TW, Brownlee KG, Conway SP, Denton M, Littlewood JM. Evaluation of a new definition for chronic Pseudomonas aeruginosa infection in cystic fibrosis patients. J Cyst Fibros. 2003;2(1):29-34. https://doi.org/10.1016/S1569-1993(02)00141-8
    » https://doi.org/10.1016/S1569-1993(02)00141-8
  • 93
    Lund-Palau H, Turnbull AR, Bush A, Bardin E, Cameron L, Soren O, et al. Pseudomonas aeruginosa infection in cystic fibrosis: pathophysiological mechanisms and therapeutic approaches. Expert Rev Respir Med. 2016;10(6):685-697. https://doi.org/10.1080/17476348.2016.1177460
    » https://doi.org/10.1080/17476348.2016.1177460
  • 94
    Flume PA, VanDevanter DR, Morgan EE, Dudley MN, Loutit JS, Bell SC, et al. A phase 3, multi-center, multinational, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of levofloxacin inhalation solution (APT-1026) in stable cystic fibrosis patients. J Cyst Fibros. 2016;15(4):495-502. https://doi.org/10.1016/j.jcf.2015.12.004
    » https://doi.org/10.1016/j.jcf.2015.12.004
  • 95
    Assael BM, Pressler T, Bilton D, Fayon M, Fischer R, Chiron R, et al. Inhaled aztreonam lysine vs. inhaled tobramycin in cystic fibrosis: a comparative efficacy trial. J Cyst Fibros. 2013;12(2):130-140. https://doi.org/10.1016/j.jcf.2012.07.006
    » https://doi.org/10.1016/j.jcf.2012.07.006
  • 96
    Chuchalin A, Csiszér E, Gyurkovics K, Bartnicka MT, Sands D, Kapranov N, et al. A formulation of aerosolized tobramycin (Bramitob) in the treatment of patients with cystic fibrosis and Pseudomonas aeruginosa infection: a double-blind, placebo-controlled, multicenter study. Paediatr Drugs. 2007;9 Suppl 1:21-31. https://doi.org/10.2165/00148581-200709001-00004
    » https://doi.org/10.2165/00148581-200709001-00004
  • 97
    Geller DE, Flume PA, Staab D, Fischer R, Loutit JS, Conrad DJ; et al. Levofloxacin inhalation solution (MP-376) in patients with cystic fibrosis with Pseudomonas aeruginosa. Am J Respir Crit Care Med. 2011;183(11):1510-1516. https://doi.org/10.1164/rccm.201008-1293OC
    » https://doi.org/10.1164/rccm.201008-1293OC
  • 98
    Hodson ME, Penketh AR, Batten JC. Aerosol carbenicillin and gentamicin treatment of Pseudomonas aeruginosa infection in patients with cystic fibrosis. Lancet. 1981;2(8256):1137-1139. https://doi.org/10.1016/S0140-6736(81)90588-2
    » https://doi.org/10.1016/S0140-6736(81)90588-2
  • 99
    Hodson ME, Gallagher CG, Govan JR. A randomised clinical trial of nebulised tobramycin or colistin in cystic fibrosis. Eur Respir J. 2002;20(3):658-664. https://doi.org/10.1183/09031936.02.00248102
    » https://doi.org/10.1183/09031936.02.00248102
  • 100
    Jensen T, Pedersen SS, Garne S, Heilmann C, Høiby N, Koch C. Colistin inhalation therapy in cystic fibrosis patients with chronic Pseudomonas aeruginosa lung infection. J Antimicrob Chemother. 1987;19(6):831-838. https://doi.org/10.1093/jac/19.6.831
    » https://doi.org/10.1093/jac/19.6.831
  • 101
    Konstan MW, Geller DE, Minic P, Brockhaus F, Zhang J, Angyalosi G. Tobramycin inhalation powder for P. aeruginosa infection in cystic fibrosis: the EVOLVE trial. Pediatr Pulmonol. 2011;46(3):230-238. https://doi.org/10.1002/ppul.21356
    » https://doi.org/10.1002/ppul.21356
  • 102
    Konstan MW, Flume PA, Kappler M, Chiron R, Higgins M, Brockhaus F, et al. Safety, efficacy and convenience of tobramycin inhalation powder in cystic fibrosis patients: The EAGER trial. J Cyst Fibros. 2011;10(1):54-61. https://doi.org/10.1016/j.jcf.2010.10.003
    » https://doi.org/10.1016/j.jcf.2010.10.003
  • 103
    Kun P, Landau LI, Phelan PD. Nebulized gentamicin in children and adolescents with cystic fibrosis. Aust Paediatr J. 1984;20(1):43-45. https://doi.org/10.1111/j.1440-1754.1984.tb00035.x
    » https://doi.org/10.1111/j.1440-1754.1984.tb00035.x
  • 104
    Lenoir G, Antypkin YG, Miano A, Moretti P, Zanda M, Varoli G, et al. Efficacy, safety, and local pharmacokinetics of highly concentrated nebulized tobramycin in patients with cystic fibrosis colonized with Pseudomonas aeruginosa. Paediatr Drugs. 2007;9 Suppl 1:11-20. https://doi.org/10.2165/00148581-200709001-00003
    » https://doi.org/10.2165/00148581-200709001-00003
  • 105
    MacLusky IB, Gold R, Corey M, Levison H. Long-term effects of inhaled tobramycin in patients with cystic fibrosis colonized with Pseudomonas aeruginosa. Pediatr Pulmonol. 1989;7(1):42-48. https://doi.org/10.1002/ppul.1950070110
    » https://doi.org/10.1002/ppul.1950070110
  • 106
    McCoy KS, Quittner AL, Oermann CM, Gibson RL, Retsch-Bogart GZ, Montgomery AB. Inhaled aztreonam lysine for chronic airway Pseudomonas aeruginosa in cystic fibrosis. Am J Respir Crit Care Med. 2008;178(9):921-928. https://doi.org/10.1164/rccm.200712-1804OC
    » https://doi.org/10.1164/rccm.200712-1804OC
  • 107
    Moss RB. Administration of aerosolized antibiotics in cystic fibrosis patients. Chest. 2001;120(3 Suppl):107S-113S. https://doi.org/10.1378/chest.120.3_suppl.107S
    » https://doi.org/10.1378/chest.120.3_suppl.107S
  • 108
    Murphy TD, Anbar RD, Lester LA, Nasr SZ, Nickerson B, VanDevanter DR, et al. Treatment with tobramycin solution for inhalation reduces hospitalizations in young CF subjects with mild lung disease. Pediatr Pulmonol. 2004;38(4):314-320. https://doi.org/10.1002/ppul.20097
    » https://doi.org/10.1002/ppul.20097
  • 109
    Nasr SZ, Sakmar E, Christodoulou E, Eckhardt BP, Streetman DS, Strouse PJ. The use of high resolution computerized tomography (HRCT) of the chest in evaluating the effect of tobramycin solution for inhalation in cystic fibrosis lung disease. Pediatr Pulmonol. 2010;45(5):440-449. https://doi.org/10.1002/ppul.21188
    » https://doi.org/10.1002/ppul.21188
  • 110
    Nikolaizik WH, Vietzke D, Ratjen F. A pilot study to compare tobramycin 80 mg injectable preparation with 300 mg solution for inhalation in cystic fibrosis patients. Can Respir J. 2008;15(5):259-262. https://doi.org/10.1155/2008/202464
    » https://doi.org/10.1155/2008/202464
  • 111
    Quintana-Gallego E, Lopez-Campos JL, Calero C, Dapena FJ. Nebulized colistin versus tobramycin in the treatment of chronic Pseudomonas colonization in cystic fibrosis patients [Article in Spanish]. Med Clin (Barc). 2014;142(2):59-63. https://doi.org/10.1016/j.medcli.2012.11.026
    » https://doi.org/10.1016/j.medcli.2012.11.026
  • 112
    Ramsey BW, Pepe MS, Quan JM, Otto KL, Montgomery AB, Williams-Warren J, et al. Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group. N Engl J Med. 1999;340(1):23-30. https://doi.org/10.1056/NEJM199901073400104
    » https://doi.org/10.1056/NEJM199901073400104
  • 113
    Retsch-Bogart GZ, Quittner AL, Gibson RL, Oermann CM, McCoy KS, Montgomery AB, et al. Efficacy and safety of inhaled aztreonam lysine for airway pseudomonas in cystic fibrosis. Chest. 2009;135(5):1223-1232. https://doi.org/10.1378/chest.08-1421
    » https://doi.org/10.1378/chest.08-1421
  • 114
    Schuster A, Haliburn C, Döring G, Goldman MH; Freedom Study Group. Safety, efficacy and convenience of colistimethate sodium dry powder for inhalation (Colobreathe DPI) in patients with cystic fibrosis: a randomised study. Thorax. 2013;68(4):344-350. https://doi.org/10.1136/thoraxjnl-2012-202059
    » https://doi.org/10.1136/thoraxjnl-2012-202059
  • 115
    Stead RJ, Hodson ME, Batten JC. Inhaled ceftazidime compared with gentamicin and carbenicillin in older patients with cystic fibrosis infected with Pseudomonas aeruginosa. Br J Dis Chest. 1987;81(3):272-279. https://doi.org/10.1016/0007-0971(87)90161-6
    » https://doi.org/10.1016/0007-0971(87)90161-6
  • 116
    Stuart Elborn J, Geller DE, Conrad D, Aaron SD, Smyth AR, Fischer R, et al. A phase 3, open-label, randomized trial to evaluate the safety and efficacy of levofloxacin inhalation solution (APT-1026) versus tobramycin inhalation solution in stable cystic fibrosis patients. J Cyst Fibros. 2015;14(4):507-514. https://doi.org/10.1016/j.jcf.2014.12.013
    » https://doi.org/10.1016/j.jcf.2014.12.013
  • 117
    Toukan Y, Gur M, Hakim FH, Geffen Y, Bar-Yoseph R, Nir V, et al. Intermittent inhaled tobramycin and systemic cytokines response in CF patients with Pseudomonas aeruginosa. Clin Invest Med. 2018;41(3):E136-E143. https://doi.org/10.25011/cim.v41i3.30792
    » https://doi.org/10.25011/cim.v41i3.30792
  • 118
    Westerman EM, Le Brun PP, Touw DJ, Frijlink HW, Heijerman HG. Effect of nebulized colistin sulphate and colistin sulphomethate on lung function in patients with cystic fibrosis: a pilot study. J Cyst Fibros. 2004;3(1):23-28. https://doi.org/10.1016/j.jcf.2003.12.005
    » https://doi.org/10.1016/j.jcf.2003.12.005
  • 119
    Dasenbrook EC, Checkley W, Merlo CA, Konstan MW, Lechtzin N, Boyle MP. Association between respiratory tract methicillin-resistant Staphylococcus aureus and survival in cystic fibrosis. JAMA. 2010;303(23):2386-2392. https://doi.org/10.1001/jama.2010.791
    » https://doi.org/10.1001/jama.2010.791
  • 120
    Hall H, Gadhok R, Alshafi K, Bilton D, Simmonds NJ. Eradication of respiratory tract MRSA at a large adult cystic fibrosis centre. Respir Med. 2015;109(3):357-363. https://doi.org/10.1016/j.rmed.2015.01.013
    » https://doi.org/10.1016/j.rmed.2015.01.013
  • 121
    Vallières E, Rendall JC, Moore JE, McCaughan J, Hoeritzauer AI, Tunney MM, et al. MRSA eradication of newly acquired lower respiratory tract infection in cystic fibrosis. ERJ Open Res. 2016;2(1):00064-2015. https://doi.org/10.1183/23120541.00064-2015
    » https://doi.org/10.1183/23120541.00064-2015
  • 122
    Vanderhelst E, De Wachter E, Willekens J, Schuermans D, Vincken W, Malfroot A, et al. Increase in ventilated air spaces after eradication of chronic methicillin-resistant Staphylococcus aureus infection in cystic fibrosis patients. Acta Clin Belg. 2015;70(1):30-33. https://doi.org/10.1179/2295333714Y.0000000079
    » https://doi.org/10.1179/2295333714Y.0000000079
  • 123
    Dolce D, Neri S, Grisotto L, Campana S, Ravenni N, Miselli F, et al. Methicillin-resistant Staphylococcus aureus eradication in cystic fibrosis patients: A randomized multicenter study. PLoS One. 2019;14(3):e0213497. https://doi.org/10.1371/journal.pone.0213497
    » https://doi.org/10.1371/journal.pone.0213497
  • 124
    Garske LA, Kidd TJ, Gan R, Bunting JP, Franks CA, Coulter C, et al. Rifampicin and sodium fusidate reduces the frequency of methicillin-resistant Staphylococcus aureus (MRSA) isolation in adults with cystic fibrosis and chronic MRSA infection. J Hosp Infect. 2004;56(3):208-214. https://doi.org/10.1016/j.jhin.2003.12.003
    » https://doi.org/10.1016/j.jhin.2003.12.003
  • 125
    Kappler M, Nagel F, Feilcke M, Kröner C, Pawlita I, Naehrig S, et al. Eradication of methicillin resistant Staphylococcus aureus detected for the first time in cystic fibrosis: A single center observational study. Pediatr Pulmonol. 2016;51(10):1010-1019. https://doi.org/10.1002/ppul.23519
    » https://doi.org/10.1002/ppul.23519
  • 126
    Muhlebach MS, Beckett V, Popowitch E, Miller MB, Baines A, Mayer-Hamblett N, et al. Microbiological efficacy of early MRSA treatment in cystic fibrosis in a randomised controlled trial. Thorax. 2017;72(4):318-326. https://doi.org/10.1136/thoraxjnl-2016-208949
    » https://doi.org/10.1136/thoraxjnl-2016-208949
  • 127
    Solís A, Brown D, Hughes J, Van Saene HK, Heaf DP. Methicillin-resistant Staphylococcus aureus in children with cystic fibrosis: An eradication protocol. Pediatr Pulmonol. 2003;36(3):189-195. https://doi.org/10.1002/ppul.10231
    » https://doi.org/10.1002/ppul.10231
  • 128
    Yang C, Montgomery M. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2021;3(3):CD001127. https://doi.org/10.1002/14651858.CD001127.pub5
    » https://doi.org/10.1002/14651858.CD001127.pub5
  • 129
    Aitken ML, Burke W, McDonald G, Shak S, Montgomery AB, Smith A. Recombinant human DNase inhalation in normal subjects and patients with cystic fibrosis. A phase 1 study. JAMA. 1992;267(14):1947-1951. https://doi.org/10.1001/jama.1992.03480140073036
    » https://doi.org/10.1001/jama.1992.03480140073036
  • 130
    Hubbard RC, McElvaney NG, Birrer P, Shak S, Robinson WW, Jolley C, et al. A preliminary study of aerosolized recombinant human deoxyribonuclease I in the treatment of cystic fibrosis. N Engl J Med. 1992;326(12):812-815. https://doi.org/10.1056/NEJM199203193261207
    » https://doi.org/10.1056/NEJM199203193261207
  • 131
    Amin R, Subbarao P, Lou W, Jabar A, Balkovec S, Jensen R, et al. The effect of dornase alfa on ventilation inhomogeneity in patients with cystic fibrosis. Eur Respir J. 2011;37(4):806-812. https://doi.org/10.1183/09031936.00072510
    » https://doi.org/10.1183/09031936.00072510
  • 132
    Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, et al. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011;38(6):1328-1335. https://doi.org/10.1183/09031936.00006211
    » https://doi.org/10.1183/09031936.00006211
  • 133
    Barker M, Franke E, Böhle M, Pfannenstiel C, Heimann G. Effect of DNase on exercise capacity in cystic fibrosis. Pediatr Pulmonol. 2004;38(1):70-74. https://doi.org/10.1002/ppul.20048
    » https://doi.org/10.1002/ppul.20048
  • 134
    Bakker EM, Volpi S, Salonini E, van der Wiel-Kooij EC, Sintnicolaas CJ, Hop WC, Assael BM, Merkus PJ, Tiddens HA. Improved treatment response to dornase alfa in cystic fibrosis patients using controlled inhalation. Eur Respir J. 2011 Dec;38(6):1328-35. https://doi.org/10.1183/09031936.00006211
    » https://doi.org/10.1183/09031936.00006211
  • 135
    Böllert FG, Paton JY, Marshall TG, Calvert J, Greening AP, Innes JA. Recombinant DNase in cystic fibrosis: a protocol for targeted introduction through n-of-1 trials. Scottish Cystic Fibrosis Group. Eur Respir J. 1999;13(1):107-113. https://doi.org/10.1183/09031936.99.13105399
    » https://doi.org/10.1183/09031936.99.13105399
  • 136
    Fiel SB, Fuchs HJ, Johnson C, Gonda I, Clark AR. Comparison of three jet nebulizer aerosol delivery systems used to administer recombinant human DNase I to patients with cystic fibrosis. The Pulmozyme rhDNase Study Group. Chest. 1995;108(1):153-156. https://doi.org/10.1378/chest.108.1.153
    » https://doi.org/10.1378/chest.108.1.153
  • 137
    Frederiksen B, Pressler T, Hansen A, Koch C, Høiby N. Effect of aerosolized rhDNase (Pulmozyme) on pulmonary colonization in patients with cystic fibrosis. Acta Paediatr. 2006;95(9):1070-1074. https://doi.org/10.1080/08035250600752466
    » https://doi.org/10.1080/08035250600752466
  • 138
    Furuya ME, Lezana-Fernández JL, Vargas MH, Hernández-Sierra JF, Ramírez-Figueroa JL. Efficacy of human recombinant DNase in pediatric patients with cystic fibrosis. Arch Med Res. 2001;32(1):30-34. https://doi.org/10.1016/S0188-4409(00)00258-7
    » https://doi.org/10.1016/S0188-4409(00)00258-7
  • 139
    Heijerman HG, van Rossem RN, Bakker W. Effect of rhDNase on lung function and quality of life in adult cystic fibrosis patients. Neth J Med. 1995;46(6):293-297. https://doi.org/10.1016/0300-2977(95)00022-F
    » https://doi.org/10.1016/0300-2977(95)00022-F
  • 140
    Hodson M. Multicenter study of rhDNase in cystic fibrosis with severe pulmonary involvement [Article in French]. Arch Pediatr. 1995;2(7):679-681. https://doi.org/10.1016/0929-693X(96)81226-7
    » https://doi.org/10.1016/0929-693X(96)81226-7
  • 141
    McCoy K, Hamilton S, Johnson C. Effects of 12-week administration of dornase alfa in patients with advanced cystic fibrosis lung disease. Pulmozyme Study Group. Chest. 1996;110(4):889-895. https://doi.org/10.1378/chest.110.4.889
    » https://doi.org/10.1378/chest.110.4.889
  • 142
    Minasian C, Wallis C, Metcalfe C, Bush A. Comparison of inhaled mannitol, daily rhDNase and a combination of both in children with cystic fibrosis: a randomised trial. Thorax. 2010;65(1):51-56. https://doi.org/10.1136/thx.2009.116970
    » https://doi.org/10.1136/thx.2009.116970
  • 143
    Quan JM, Tiddens HA, Sy JP, McKenzie SG, Montgomery MD, Robinson PJ, et al. A two-year randomized, placebo-controlled trial of dornase alfa in young patients with cystic fibrosis with mild lung function abnormalities. J Pediatr. 2001;139(6):813-820. https://doi.org/10.1067/mpd.2001.118570
    » https://doi.org/10.1067/mpd.2001.118570
  • 144
    Robinson TE, Goris ML, Zhu HJ, Chen X, Bhise P, Sheikh F, et al. Dornase alfa reduces air trapping in children with mild cystic fibrosis lung disease: a quantitative analysis. Chest. 2005;128(4):2327-2335. https://doi.org/10.1378/chest.128.4.2327
    » https://doi.org/10.1378/chest.128.4.2327
  • 145
    Robinson M, Hemming AL, Moriarty C, Eberl S, Bye PT. Effect of a short course of rhDNase on cough and mucociliary clearance in patients with cystic fibrosis. Pediatr Pulmonol. 2000;30(1):16-24. https://doi.org/10.1002/1099-0496(200007)30:1<16::AID-PPUL4>3.0.CO;2-H
    » https://doi.org/10.1002/1099-0496(200007)30:1<16::AID-PPUL4>3.0.CO;2-H
  • 146
    Shah PL, Scott SF, Knight RA, Marriott C, Ranasinha C, Hodson ME. In vivo effects of recombinant human DNase I on sputum in patients with cystic fibrosis. Thorax. 1996;51(2):119-125. https://doi.org/10.1136/thx.51.2.119
    » https://doi.org/10.1136/thx.51.2.119
  • 147
    Suri R, Metcalfe C, Lees B, Grieve R, Flather M, Normand C, et al. Comparison of hypertonic saline and alternate-day or daily recombinant human deoxyribonuclease in children with cystic fibrosis: a randomised trial. Lancet. 2001;358(9290):1316-1321. https://doi.org/10.1016/S0140-6736(01)06412-1
    » https://doi.org/10.1016/S0140-6736(01)06412-1
  • 148
    Bonestroo HJ, Slieker MG, Arets HG. No positive effect of rhdnase on the pulmonary colonization in children with cystic fibrosis. Monaldi Arch Chest Dis. 2010;73(1):12-17. https://doi.org/10.4081/monaldi.2010.308
    » https://doi.org/10.4081/monaldi.2010.308
  • 149
    Cobos N, Danés I, Gartner S, González M, Liñán S, Arnau JM. DNase use in the daily care of cystic fibrosis: who benefits from it and to what extent? Results of a cohort study of 199 patients in 13 centres. DNase National Study Group. Eur J Pediatr. 2000;159(3):176-181. https://doi.org/10.1007/s004310050046
    » https://doi.org/10.1007/s004310050046
  • 150
    Davies J, Trindade MT, Wallis C, Rosenthal M, Crawford O, Bush A. Retrospective review of the effects of rhDNase in children with cystic fibrosis. Pediatr Pulmonol. 1997;23(4):243-248. https://doi.org/10.1002/(SICI)1099-0496(199704)23:4<243::AID-PPUL1>3.0.CO;2-N
    » https://doi.org/10.1002/(SICI)1099-0496(199704)23:4<243::AID-PPUL1>3.0.CO;2-N
  • 151
    Derelle J, Bertolo-Houriez E, Marchal F, Weber M, Virion JM, Vidailhet M. Respiratory evolution of patient with mucoviscidosis treated with mucolytic agents plus dornase alfa [Article in French]. Arch Pediatr. 1998;5(4):371-377. https://doi.org/10.1016/S0929-693X(98)80022-5
    » https://doi.org/10.1016/S0929-693X(98)80022-5
  • 152
    Harms HK, Matouk E, Tournier G, von der Hardt H, Weller PH, Romano L, et al. Multicenter, open-label study of recombinant human DNase in cystic fibrosis patients with moderate lung disease. DNase International Study Group. Pediatr Pulmonol. 1998;26(3):155-161. https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
    » https://doi.org/10.1002/(SICI)1099-0496(199809)26:3<155::AID-PPUL1>3.0.CO;2-K
  • 153
    Hodson ME, McKenzie S, Harms HK, Koch C, Mastella G, Navarro J, et al. Dornase alfa in the treatment of cystic fibrosis in Europe: a report from the Epidemiologic Registry of Cystic Fibrosis. Pediatr Pulmonol. 2003;36(5):427-432. https://doi.org/10.1002/ppul.10348
    » https://doi.org/10.1002/ppul.10348
  • 154
    Johnson CA, Butler SM, Konstan MW, Breen TJ, Morgan WJ. Estimating effectiveness in an observational study: a case study of dornase alfa in cystic fibrosis. The Investigators and Coordinators of the Epidemiologic Study of Cystic Fibrosis. J Pediatr. 1999;134(6):734-739. https://doi.org/10.1016/S0022-3476(99)70290-8
    » https://doi.org/10.1016/S0022-3476(99)70290-8
  • 155
    Konstan MW, Wagener JS, Pasta DJ, Millar SJ, Jacobs JR, Yegin A, et al. Clinical use of dornase alpha is associated with a slower rate of FEV1 decline in cystic fibrosis. Pediatr Pulmonol. 2011;46(6):545-553. https://doi.org/10.1002/ppul.21388
    » https://doi.org/10.1002/ppul.21388
  • 156
    Milla CE. Long-term effects of aerosolised rhDNase on pulmonary disease progression in patients with cystic fibrosis. Thorax. 1998;53(12):1014-1017. https://doi.org/10.1136/thx.53.12.1014
    » https://doi.org/10.1136/thx.53.12.1014
  • 157
    Rozov T, de Oliveira VZ, Santana MA, Adde FV, Mendes RH, Paschoal IA, et al. Dornase alfa improves the health-related quality of life among Brazilian patients with cystic fibrosis--a one-year prospective study. Pediatr Pulmonol. 2010;45(9):874-882. https://doi.org/10.1002/ppul.21267
    » https://doi.org/10.1002/ppul.21267
  • 158
    Rozov T, Silva FA, Santana MA, Adde FV, Mendes RH; Brazilian Cystic Fibrosis Multicenter Study Group. A first-year dornase alfa treatment impact on clinical parameters of patients with cystic fibrosis: the Brazilian cystic fibrosis multicenter study. Rev Paul Pediatr. 2013;31(4):420-430. https://doi.org/10.1590/S0103-05822013000400002
    » https://doi.org/10.1590/S0103-05822013000400002
  • 159
    Shah PL, Conway S, Scott SF, Rainisio M, Wildman M, Stableforth D, et al. A case-controlled study with dornase alfa to evaluate impact on disease progression over a 4-year period. Respiration. 2001;68(2):160-164. https://doi.org/10.1159/000050486
    » https://doi.org/10.1159/000050486
  • 160
    Shah PL, Scott SF, Geddes DM, Hodson ME. Two years experience with recombinant human DNase I in the treatment of pulmonary disease in cystic fibrosis. Respir Med. 1995;89(7):499-502. https://doi.org/10.1016/0954-6111(95)90126-4
    » https://doi.org/10.1016/0954-6111(95)90126-4
  • 161
    Wizla-Derambure N, Michaud L, Sardet A, Deschildre A, Loeuille GA, Tassin E, et al. Effect of rhDNase on the respiratory function and nutritional status of children and adolescents with mucoviscidosis [Article in French]. Arch Pediatr. 1998;5(4):378-383. https://doi.org/10.1016/S0929-693X(98)80023-7
    » https://doi.org/10.1016/S0929-693X(98)80023-7
  • 162
    LiPuma JJ, Spilker T, Gill LH, Campbell PW 3rd, Liu L, Mahenthiralingam E. Disproportionate distribution of Burkholderia cepacia complex species and transmissibility markers in cystic fibrosis. Am J Respir Crit Care Med. 2001;164(1):92-96. https://doi.org/10.1164/ajrccm.164.1.2011153
    » https://doi.org/10.1164/ajrccm.164.1.2011153
  • 163
    Zlosnik JE, Zhou G, Brant R, Henry DA, Hird TJ, Mahenthiralingam E, et al. Burkholderia species infections in patients with cystic fibrosis in British Columbia, Canada. 30 years' experience. Ann Am Thorac Soc. 2015;12(1):70-78. https://doi.org/10.1513/AnnalsATS.201408-395OC
    » https://doi.org/10.1513/AnnalsATS.201408-395OC
  • 164
    Horsley A, Webb K, Bright-Thomas R, Govan J, Jones A. Can early Burkholderia cepacia complex infection in cystic fibrosis be eradicated with antibiotic therapy?. Front Cell Infect Microbiol. 2011;1:18. https://doi.org/10.3389/fcimb.2011.00018
    » https://doi.org/10.3389/fcimb.2011.00018
  • 165
    Tullis DE, Burns JL, Retsch-Bogart GZ, Bresnik M, Henig NR, Lewis SA, et al. Inhaled aztreonam for chronic Burkholderia infection in cystic fibrosis: a placebo-controlled trial. J Cyst Fibros. 2014;13(3):296-305. https://doi.org/10.1016/j.jcf.2013.08.011
    » https://doi.org/10.1016/j.jcf.2013.08.011
  • 166
    Uluer AZ, Waltz DA, Kalish LA, Adams S, Gerard C, Ericson DA. Inhaled amiloride and tobramycin solutions fail to eradicate Burkholderia dolosa in patients with cystic fibrosis. J Cyst Fibros. 2013;12(1):54-59. https://doi.org/10.1016/j.jcf.2012.06.006
    » https://doi.org/10.1016/j.jcf.2012.06.006
  • Financial support:

    This project received partial financial support through unrestricted educational support from Vertex Farmacêutica do Brasil Ltda, São Paulo (SP) Brazil.

Publication Dates

  • Publication in this collection
    15 May 2023
  • Date of issue
    2023

History

  • Received
    19 Mar 2023
  • Accepted
    31 Mar 2023
Sociedade Brasileira de Pneumologia e Tisiologia SCS Quadra 1, Bl. K salas 203/204, 70398-900 - Brasília - DF - Brasil, Fone/Fax: 0800 61 6218 ramal 211, (55 61)3245-1030/6218 ramal 211 - São Paulo - SP - Brazil
E-mail: jbp@sbpt.org.br