331
Views
0
CrossRef citations to date
0
Altmetric
Review

Emerging pharmacotherapies in cystic fibrosis

, , , , &
Pages 843-855 | Received 22 Feb 2018, Accepted 13 Aug 2018, Published online: 19 Sep 2018

References

  • Burns JL, Emerson J, Stapp JR, et al. Microbiology of sputum from patients at cystic fibrosis centers in the United States. Clin Infect Dis. 1998;27:158–163.
  • Wood RE, Boat TF, Doershuk CF. Cystic fibrosis. Am Rev Respir Dis. 1976;113:833–878.
  • De Boeck K, Zolin A, Cuppens H, et al. The relative frequency of CFTR mutation classes in European patients with cystic fibrosis. J Cyst Fibros. 2014;13:403–409.
  • Zielenski J, Markiewicz D, Chen HS, et al. Identification of six mutations (R31L, 441delA, 681delC, 1461ins4, W1089R, E1104X) in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Hum Mutat. 1995;5:43–47.
  • Matsui H, Grubb BR, Tarran R, et al. Evidence for periciliary liquid layer depletion, not abnormal ion composition, in the pathogenesis of cystic fibrosis airways disease. Cell. 1998;95:1005–1015.
  • Boucher RC. Evidence for airway surface dehydration as the initiating event in CF airway disease. J Intern Med. 2007;261:5–16.
  • Boucher RC, Stutts MJ, Knowles MR, et al. Na+ transport in cystic fibrosis respiratory epithelia. Abnormal basal rate and response to adenylate cyclase activation. J Clin Invest. 1986;78:1245–1252.
  • Stutts MJ, Canessa CM, Olsen JC, et al. CFTR as a cAMP-dependent regulator of sodium channels. Science. 1995;269:847–850.
  • Ramsey BW, Davies J, McElvaney NG, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. 2011;365:1663–1672.
  • Van Goor F, Hadida S, Grootenhuis PD, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci U S A. 2009;106:18825–18830.
  • Accurso FJ, Rowe SM, Clancy JP, et al. Effect of VX-770 in persons with cystic fibrosis and the G551D-CFTR mutation. N Engl J Med. 2010;363:1991–2003.
  • Davies JC, Wainwright CE, Canny GJ, 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;187:1219–1225.
  • Rosenfeld M, Wainwright CE, Higgins M, 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. Lancet Respir Med. 2018;6:545–553.
  • Barry PJ, Plant BJ, Nair A, et al. Effects of ivacaftor in patients with cystic fibrosis who carry the G551D mutation and have severe lung disease. Chest. 2014;146:152–158.
  • Rowe SM, Heltshe SL, Gonska T, 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;190:175–184.
  • Sawicki GS, McKone EF, Pasta DJ, et al. Sustained Benefit from ivacaftor demonstrated by combining clinical trial and cystic fibrosis patient registry data. Am J Respir Crit Care Med. 2015;192:836–842.
  • FDA expands approved use of kalydeco to treat additional mutations of cystic fibrosis [Internet]. U.S. Food & Drug Administration; [ cited 2017 May 17]. Available from: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm559212.htm
  • FDA approves KALYDECO® (ivacaftor) for more than 900 people ages 2 and older with cystic fibrosis who have certain residual function mutations [Internet]. Vertex pharmaceuticals; [ cited 2017 May 17]. Available from: http://investors.vrtx.com/releasedetail.cfm?releaseid=1026864
  • Yu H, Burton B, Huang CJ, et al. Ivacaftor potentiation of multiple CFTR channels with gating mutations. J Cyst Fibros. 2012;11:237–245.
  • FDA approves KALYDECO® (ivacaftor) for more than 600 people ages 2 and older with cystic fibrosis who have certain residual function mutations [Internet]. Vertex pharmaceuticals; [ cited 2017 Aug 1]. Available from: http://investors.vrtx.com/releasedetail.cfm?ReleaseID=1035299
  • Bebok Z, Collawn JF, Wakefield J, et al. Failure of cAMP agonists to activate rescued deltaF508 CFTR in CFBE41o- airway epithelial monolayers. J Physiol. 2005;569:601–615.
  • Wang W, Li G, Clancy JP, et al. Activating cystic fibrosis transmembrane conductance regulator channels with pore blocker analogs. J Biol Chem. 2005;280:23622–23630.
  • Flume PA, Liou TG, Borowitz DS, et al. Ivacaftor in subjects with cystic fibrosis who are homozygous for the F508del-CFTR mutation. Chest. 2012;142:718–724.
  • Van Goor F, Hadida S, Grootenhuis PD, et al. Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. Proc Natl Acad Sci U S A. 2011;108:18843–18848.
  • Van Goor F, Hadida S, Grootenhuis PD, et al. Abstract S9.4: VX-809, a CFTR corrector, increases the cell surface density of functional F508del-CFTR in pre-clinical models of cystic fibrosis Pediatr. Pulmonol. 2009;44:154–155.
  • Clancy JP, Rowe SM, Accurso FJ, et al. Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for the F508del-CFTR mutation. Thorax. 2012;67:12–18.
  • Boyle MP, Bell SC, Konstan MW, et al. A CFTR corrector (lumacaftor) and a CFTR potentiator (ivacaftor) for treatment of patients with cystic fibrosis who have a phe508del CFTR mutation: a phase 2 randomised controlled trial. Lancet Respir Med. 2014;2:527–538.
  • Wainwright CE, Elborn JS, Ramsey BW, et al. Lumacaftor-ivacaftor in patients with cystic fibrosis homozygous for phe508del CFTR. N Engl J Med. 2015;373:220–231.
  • Veit G, Rg A, Perdomo D, et al. Some gating potentiators, including VX-770, diminish DeltaF508-CFTR functional expression. Sci Transl Med. 2014;6:246ra97.
  • Clancy JP. CFTR potentiators: not an open and shut case. Sci Transl Med. 2014;6:246fs27.
  • Ratjen F, Hug C, Marigowda G, 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. Lancet Respir Med. 2017;5:557–567.
  • Konstan MW, McKone EF, Moss RB, 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;5:107–118.
  • Jm P, Cooke J, Ja L-H, et al. Abstract WS01.4: VX-661 in combination with ivacaftor in patients with cystic fibrosis and the F508del-CFTR mutation. J Cyst Fibros. 2015;14:S1.
  • Taylor-Cousar JL, Munck A, McKone EF, et al. Tezacaftor-ivacaftor in patients with cystic fibrosis homozygous for phe508del. N Engl J Med. 2017;377:2013–2023.
  • Grasemann H. CFTR modulator therapy for cystic fibrosis. N Engl J Med. 2017;377:2085–2088.
  • Vertex provides update on ongoing phase 3 program for VX-661 in combination with ivacaftor for the treatment of cystic fibrosis [Internet]. Vertex pharmaceuticals; [ cited 2016 Aug 15]. Available from: http://investors.vrtx.com/releasedetail.cfm?ReleaseID=984388
  • Rowe SM, Daines C, Ringshausen FC, et al. Tezacaftor-ivacaftor in residual-function heterozygotes with cystic fibrosis. N Engl J Med. 2017;377:2024–2035.
  • Timmins GS. Deuterated drugs: where are we now? Expert Opin Ther Pat. 2014;24:1067–1075.
  • Harbeson SL, Morgan AJ, Liu JF, et al. Altering metabolic profiles of drugs by precision deuteration 2: discovery of a deuterated analog of ivacaftor with differentiated pharmacokinetics for clinical development. J Pharmacol Exp Ther. 2017;362:359–367.
  • Uttamsineh V, Pilja L, Grotbeck B, et al. WS13.6 CTP-656 tablet confirmed superiority of pharmacokinetic profile relative to kalydeco® in phase I clinical studies. J Cyst Fibros. 2016 Jun;15:S22.
  • Van der Plas SE, Kelgtermans H, De Munck T, et al. Discovery of N-(3-carbamoyl-5,5,7,7-tetramethyl-5,7-dihydro-4H-thieno[2,3-c]pyran-2-yl)-lH-pyr azole-5-carboxamide (GLPG1837), a novel potentiator which can open class iii mutant cystic fibrosis transmembrane conductance regulator (CFTR) channels to a high extent. J Med Chem. 2018;61(4):1425-1435.
  • SAPHIRA 1 topline results [Internet]. Galapagos; [ cited 2016 Dec 21]
  • Andrews M, Van Der Plas S, Gees M, et al. Abstract 20: characterization of a novel potentiator series for treating cystic fbirosis. Pediatr Pulmonol. 2016;51:S201.
  • Van De Steen O, Namour F, Kanters D, et al. Abstract 252: safety, tolerability and pharmacokinetics of a novel CFTR corrector molecule GLPG2222 in healthy volunteers. Pediatr Pulmonol. 2016;51:S287.
  • Kazani S, Alcantara J, Debonnett L, et al. Qbw251 is a safe and efficacious cftr potentiator for patients with cystic fibrosis. Am J Respir Crit Care Med. 2016;193:A7789.
  • Vertex announces positive phase 1 & phase 2 data from three different triple combination regimens in people with cystic fibrosis who have one F508del mutation and one minimal function mutation (F508del/min) [Internet]. Vertex pharmaceuticals; [ cited 2017 Jul 18]. Available from: http://investors.vrtx.com/releasedetail.cfm?ReleaseID=1033559
  • VX-659: Phase 2 Study (VX16-659-101) Preliminary Results. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-initiates-first-phase-3-study-vx-659-tezacaftor-and
  • Vertex Initiates Phase 3 Studies of VX-445, Tezacaftor and Ivacaftor as a Triple Combination Regimen for People with Cystic Fibrosis. Available from: https://investors.vrtx.com/news-releases/news-release-details/vertex-initiates-phase-3-studies-vx-445-tezacaftor-and-ivacaftor
  • Pradhan K, Sydykov A, Tian X, et al. Soluble guanylate cyclase stimulator riociguat and phosphodiesterase 5 inhibitor sildenafil ameliorate pulmonary hypertension due to left heart disease in mice. Int J Cardiol. 2016;216:85–91.
  • Ghofrani HA, Humbert M, Langleben D, et al. Riociguat: mode of action and clinical development in pulmonary hypertension. Chest. 2017;151:468–480.
  • Golin-Bisello F, Bradbury N, Ameen N. STa and cGMP stimulate CFTR translocation to the surface of villus enterocytes in rat jejunum and is regulated by protein kinase G. Am J Physiol Cell Physiol. 2005;289:C708–16.
  • Poschet JF, Fazio JA, Timmins GS, et al. Endosomal hyperacidification in cystic fibrosis is due to defective nitric oxide-cylic GMP signalling cascade. EMBO Rep. 2006;7:553–559.
  • Poschet J, Perkett E, Deretic V. Hyperacidification in cystic fibrosis: links with lung disease and new prospects for treatment. Trends Mol Med. 2002;8:512–519.
  • Poschet JF, Timmins GS, Taylor-Cousar JL, et al. Pharmacological modulation of cGMP levels by phosphodiesterase 5 inhibitors as a therapeutic strategy for treatment of respiratory pathology in cystic fibrosis. Am J Physiol Lung Cell Mol Physiol. 2007;293:L712–9.
  • Quon BS, Rowe SM. New and emerging targeted therapies for cystic fibrosis. BMJ. 2016;352:i859.
  • Km K, Xue X, Gunn G, et al. Therapeutics based on stop codon readthrough. Annu Rev Genomics Hum Genet. 2014;15:371–394.
  • Scheunemann AE, Graham WD, Vendeix FA, et al. Binding of aminoglycoside antibiotics to helix 69 of 23S rRNA. Nucleic Acids Res. 2010;38:3094–3105.
  • Salas-Marco J, Bedwell DM. Discrimination between defects in elongation fidelity and termination efficiency provides mechanistic insights into translational readthrough. J Mol Biol. 2005;348:801–815.
  • Kramer EB, Vallabhaneni H, Mayer LM, et al. A comprehensive analysis of translational missense errors in the yeast Saccharomyces cerevisiae. RNA. 2010;16:1797–1808.
  • Nudelman I, Rebibo-Sabbah A, Cherniavsky M, et al. Development of novel aminoglycoside (NB54) with reduced toxicity and enhanced suppression of disease-causing premature stop mutations. J Med Chem. 2009;52:2836–2845.
  • Rowe SM, Sloane P, Tang LP, et al. Suppression of CFTR premature termination codons and rescue of CFTR protein and function by the synthetic aminoglycoside NB54. J Mol Med (Berl). 2011;89:1149–1161.
  • Xue X, Mutyam V, Tang L, et al. Synthetic aminoglycosides efficiently suppress cystic fibrosis transmembrane conductance regulator nonsense mutations and are enhanced by ivacaftor. Am J Respir Cell Mol Biol. 2014;50:805–816.
  • Du M, Liu X, Welch EM, et al. PTC124 is an orally bioavailable compound that promotes suppression of the human CFTR-G542X nonsense allele in a CF mouse model. Proc Natl Acad Sci U S A. 2008;105:2064–2069.
  • Welch EM, Barton ER, Zhuo J, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007;447:87–91.
  • Shoseyov D, Cohen-Cymberknoh M, Wilschanski M. Ataluren for the treatment of cystic fibrosis. Expert Rev Respir Med. 2016;10(4):387-391.
  • Sermet-Gaudelus I, Boeck KD, Casimir GJ, et al. Ataluren (PTC124) induces cystic fibrosis transmembrane conductance regulator protein expression and activity in children with nonsense mutation cystic fibrosis. Am J Respir Crit Care Med. 2010;182:1262–1272.
  • Siddiqui N, Sonenberg N. Proposing a mechanism of action for Ataluren. Proc Natl Acad Sci U S A. 2016;113:12353–12355.
  • Auld DS, Thorne N, Maguire WF, et al. Mechanism of PTC124 activity in cell-based luciferase assays of nonsense codon suppression. Proc Natl Acad Sci U S A. 2009;106:3585–3590.
  • McElroy SP, Nomura T, Torrie LS, et al. A lack of premature termination codon read-through efficacy of PTC124 (Ataluren) in a diverse array of reporter assays. PLoS Biol. 2013;11:e1001593.
  • Kerem E, Konstan MW, De Boeck K, et al. Ataluren for the treatment of nonsense-mutation cystic fibrosis: a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Respir Med. 2014;2:539–547.
  • Crystal RG, McElvaney NG, Rosenfeld MA, et al. Administration of an adenovirus containing the human CFTR cDNA to the respiratory tract of individuals with cystic fibrosis. Nat Genet. 1994;8:42–51.
  • McElvaney NG, Rg C. IL-6 release and airway administration of human CFR cDNA adenovirus vector. Nat Med. 1995;1:182–184.
  • Crystal RG, Jaffe A, Brody S, et al. A phase 1 study, in cystic fibrosis patients, of the safety, toxicity, and biological efficacy of a single administration of a replication deficient, recombinant adenovirus carrying the cDNA of the normal cystic fibrosis transmembrane conductance regulator gene in the lung. Hum Gene Ther. 1995;6:643–666.
  • Hay JG, McElvaney NG, Herena J, et al. Modification of nasal epithelial potential differences of individuals with cystic fibrosis consequent to local administration of a normal CFTR cDNA adenovirus gene transfer vector. Hum Gene Ther. 1995;6:1487–1496.
  • Alton EW, Middleton PG, Caplen NJ, et al. Non-invasive liposome-mediated gene delivery can correct the ion transport defect in cystic fibrosis mutant mice. Nat Genet. 1993;5:135–142.
  • Caplen NJ, Alton EW, Middleton PG, et al. Liposome-mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Nat Med. 1995;1:39–46.
  • Yoshimura K, Rosenfeld MA, Nakamura H, et al. Expression of the human cystic fibrosis transmembrane conductance regulator gene in the mouse lung after in vivo intratracheal plasmid-mediated gene transfer. Nucleic Acids Res. 1992;20:3233–3240.
  • Rosenfeld MA, Yoshimura K, Trapnell BC, et al. In vivo transfer of the human cystic fibrosis transmembrane conductance regulator gene to the airway epithelium. Cell. 1992;68:143–155.
  • Zabner J, Couture LA, Gregory RJ, et al. Adenovirus-mediated gene transfer transiently corrects the chloride transport defect in nasal epithelia of patients with cystic fibrosis. Cell. 1993;75:207–216.
  • Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci U S A. 1996;93:1156–1160.
  • Boch J, Scholze H, Schornack S, et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science. 2009;326:1509–1512.
  • Boch J. TALEs of genome targeting. Nat Biotechnol. 2011;29:135–136.
  • Urnov FD, Rebar EJ, Holmes MC, et al. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11:636–646.
  • Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339:823–826.
  • Jinek M, East A, Cheng A, et al. RNA-programmed genome editing in human cells. Elife. 2013;2:e00471.
  • Esvelt KM, Mali P, Braff JL, et al. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat Methods. 2013;10:1116–1121.
  • Schwank G, Koo BK, Sasselli V, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13:653–658.
  • Mandegar MA, Huebsch N, Frolov EB, et al. CRISPR interference efficiently induces specific and reversible gene silencing in human iPSCs. Cell Stem Cell. 2016;18:541–553.
  • Amabile A, Migliara A, Capasso P, et al. Inheritable silencing of endogenous genes by hit-and-run targeted epigenetic editing. Cell. 2016;167:219–32e14.
  • Birrer P, McElvaney NG, Rudeberg A, et al. Protease-antiprotease imbalance in the lungs of children with cystic fibrosis. Am J Respir Crit Care Med. 1994;150:207–213.
  • McElvaney NG. Alpha-1 antitrypsin therapy in cystic fibrosis and the lung disease associated with Alpha-1 antitrypsin deficiency. Ann Am Thorac Soc. 2016;13(Suppl 2):S191–6.
  • Griese M, Kappler M, Gaggar A, et al. Inhibition of airway proteases in cystic fibrosis lung disease. Eur Respir J. 2008;32:783–795.
  • Carrell RW, Jeppsson JO, Laurell CB, et al. Structure and variation of human alpha 1-antitrypsin. Nature. 1982;298:329–334.
  • Thompson RC, Ohlsson K. Isolation, properties, and complete amino acid sequence of human secretory leukocyte protease inhibitor, a potent inhibitor of leukocyte elastase. Proc Natl Acad Sci U S A. 1986;83:6692–6696.
  • Bingle L, Tetley TD. Secretory leukoprotease inhibitor: partnering alpha 1-proteinase inhibitor to combat pulmonary inflammation. Thorax. 1996;51:1273–1274.
  • Wiedow O, Schroder JM, Gregory H, et al. Elafin: an elastase-specific inhibitor of human skin. Purification, characterization, and complete amino acid sequence. J Biol Chem. 1990;265:14791–14795.
  • Wewers MD, Herzyk DJ, Gadek JE. Alveolar fluid neutrophil elastase activity in the adult respiratory distress syndrome is complexed to alpha-2-macroglobulin. J Clin Invest. 1988;82:1260–1267.
  • Stockley RA. The role of proteinases in the pathogenesis of chronic bronchitis. Am J Respir Crit Care Med. 1994;150:S109–13.
  • Voynow JA, Young LR, Wang Y, et al. Neutrophil elastase increases MUC5AC mRNA and protein expression in respiratory epithelial cells. Am J Physiol. 1999;276:L835–43.
  • Amitani R, Wilson R, Rutman A, et al. Effects of human neutrophil elastase and Pseudomonas aeruginosa proteinases on human respiratory epithelium. Am J Respir Cell Mol Biol. 1991;4:26–32.
  • Berger M, Sorensen RU, Tosi MF, et al. Complement receptor expression on neutrophils at an inflammatory site, the Pseudomonas-infected lung in cystic fibrosis. J Clin Invest. 1989;84:1302–1313.
  • Tosi MF, Zakem H, Berger M. Neutrophil elastase cleaves C3bi on opsonized pseudomonas as well as CR1 on neutrophils to create a functionally important opsonin receptor mismatch. J Clin Invest. 1990;86:300–308.
  • Devaney JM, Greene CM, Taggart CC, et al. Neutrophil elastase up-regulates interleukin-8 via toll-like receptor 4. FEBS Lett. 2003;544:129–132.
  • Nakamura H, Yoshimura K, McElvaney NG, et al. Neutrophil elastase in respiratory epithelial lining fluid of individuals with cystic fibrosis induces interleukin-8 gene expression in a human bronchial epithelial cell line. J Clin Invest. 1992;89:1478–1484.
  • McElvaney NG, Nakamura H, Birrer P, et al. Modulation of airway inflammation in cystic fibrosis. In vivo suppression of interleukin-8 levels on the respiratory epithelial surface by aerosolization of recombinant secretory leukoprotease inhibitor. J Clin Invest. 1992;90:1296–1301.
  • Sly PD, Gangell CL, Chen L, et al. Risk factors for bronchiectasis in children with cystic fibrosis. N Engl J Med. 2013;368:1963–1970.
  • McElvaney NG, Hubbard RC, Birrer P, et al. Aerosol alpha 1-antitrypsin treatment for cystic fibrosis. Lancet. 1991;337:392–394.
  • Griese M, Latzin P, Kappler M, et al. alpha1-Antitrypsin inhalation reduces airway inflammation in cystic fibrosis patients. Eur Respir J. 2007;29:240–250.
  • Martin SL, Downey D, Bilton D, et al. Safety and efficacy of recombinant alpha(1)-antitrypsin therapy in cystic fibrosis. Pediatr Pulmonol. 2006;41:177–183.
  • Hartl D, Latzin P, Hordijk P, et al. Cleavage of CXCR1 on neutrophils disables bacterial killing in cystic fibrosis lung disease. Nat Med. 2007;13:1423–1430.
  • Berger MKM, Hilliard JB. Aerosolized prolastin (α1-protease inhibitor) in CF. Pediatr Pulmonol. 1995;20:421.
  • Cantin AM, Berthiaume Y, Cloutier D, et al. Prolastin aerosol therapy and sputum taurine in cystic fibrosis. Clin Invest Med. 2006;29:201–207.
  • McElvaney OJ, Gunaratnam C, Reeves EP, et al. A specialized method of sputum collection and processing for therapeutic interventions in cystic fibrosis. J Cyst Fibros. 2018. [Epub ahead of print].
  • Elborn JS, Perrett J, Forsman-Semb K, et al. Efficacy, safety and effect on biomarkers of AZD9668 in cystic fibrosis. Eur Respir J. 2012;40:969–976.
  • von Nussbaum F, Li VM, Meibom D, et al. Potent and selective human neutrophil elastase inhibitors with novel equatorial ring topology: in vivo efficacy of the polar pyrimidopyridazine BAY-8040 in a pulmonary arterial hypertension rat model. ChemMedChem. 2016;11:199–206.
  • Gaggar A, Chen J, Chmiel JF, et al. Inhaled alpha1-proteinase inhibitor therapy in patients with cystic fibrosis. J Cyst Fibros. 2016;15:227–233.
  • Vasconcelos A, Azoia NG, Carvalho AC, et al. Tailoring elastase inhibition with synthetic peptides. Eur J Pharmacol. 2011;666:53–60.
  • Henriksen PA. The potential of neutrophil elastase inhibitors as anti-inflammatory therapies. Curr Opin Hematol. 2014;21:23–28.
  • Iwata K, Doi A, Ohji G, et al. Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): a systematic review and meta-analysis. Intern Med. 2010;49:2423–2432.
  • McElvaney OJ, O’Reilly N, White M, et al. The effect of the decoy molecule PA401 on CXCL8 levels in bronchoalveolar lavage fluid of patients with cystic fibrosis. Mol Immunol. 2015;63:550–558.
  • Karp CL, Flick LM, Park KW, et al. Defective lipoxin-mediated anti-inflammatory activity in the cystic fibrosis airway. Nat Immunol. 2004;5:388–392.
  • Filep JG, Zouki C, Petasis NA, et al. Anti-inflammatory actions of lipoxin A(4) stable analogs are demonstrable in human whole blood: modulation of leukocyte adhesion molecules and inhibition of neutrophil-endothelial interactions. Blood. 1999;94:4132–4142.
  • Jozsef L, Zouki C, Petasis NA, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci U S A. 2002;99:13266–13271.
  • Nagaoka I, Tamura H, Hirata M. An antimicrobial cathelicidin peptide, human CAP18/LL-37, suppresses neutrophil apoptosis via the activation of formyl-peptide receptor-like 1 and P2X7. J Immunol. 2006;176:3044–3052.
  • Herrera BS, Hasturk H, Kantarci A, et al. Impact of resolvin E1 on murine neutrophil phagocytosis in type 2 diabetes. Infect Immun. 2015;83:792–801.
  • Freire MO, Dalli J, Serhan CN, et al. Neutrophil resolvin E1 receptor expression and function in Type 2 diabetes. J Immunol. 2017;198:718–728.
  • Kurihara T, Jones CN, Yu YM, et al. Resolvin D2 restores neutrophil directionality and improves survival after burns. FASEB J. 2013;27:2270–2281.
  • Schwab JM, Chiang N, Arita M, et al. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature. 2007;447:869–874.
  • Hsiao HM, Thatcher TH, Levy EP, et al. Resolvin D1 attenuates polyinosinic-polycytidylic acid-induced inflammatory signaling in human airway epithelial cells via TAK1. J Immunol. 2014;193:4980–4987.
  • Ringholz FC, Higgins G, Hatton A, et al. Resolvin D1 regulates epithelial ion transport and inflammation in cystic fibrosis airways. J Cyst Fibros. 2018;17(5);607-615.
  • Pertwee RG. Endocannabinoids and their pharmacological actions. Handb Exp Pharmacol. 2015;231:1–37.
  • Gkoumassi E, Dekkers BG, Droge MJ, et al. Virodhamine and CP55,940 modulate cAMP production and IL-8 release in human bronchial epithelial cells. Br J Pharmacol. 2007;151:1041–1048.
  • Motwani MP, Bennett F, Norris PC, et al. Potent anti-inflammatory and pro-resolving effects of anabasum in a human model of self-resolving acute inflammation. Clin Pharmacol Ther. 2017. [Epub ahead of print].
  • Martiniano SL, Toprak D, Ong T, et al. Highlights from the 2017 North American cystic fibrosis conference. Pediatric pulmonology. 2018
  • Gardiner P, Malmgren A, Ersdal E, et al. Enac inhibitor Azd5634 first in human trial reveals promising clinical profile for the treatment of cystic fibrosis. D94 ADVANCES IN CYSTIC FIBROSIS AND NON-CYSTIC FIBROSIS BRONCHIECTASIS. Am Thorac Soc. 2017;A7306–A.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.