Urinary Leukotriene E4 and Prostaglandin D2 Metabolites Increase in Adult and Childhood Severe Asthma Characterized by Type 2 Inflammation. A Clinical Observational Study

Johan Kolmert, Cristina Gómez, David Balgoma, Marcus Sjödin, Johan Bood, Jon R Konradsen, Magnus Ericsson, John-Olof Thörngren, Anna James, Maria Mikus, Ana R Sousa, John H Riley, Stewart Bates, Per S Bakke, Ioannis Pandis, Massimo Caruso, Pascal Chanez, Stephen J Fowler, Thomas Geiser, Peter Howarth, Ildikó Horváth, Norbert Krug, Paolo Montuschi, Marek Sanak, Annelie Behndig, Dominick E Shaw, Richard G Knowles, Cécile T J Holweg, Åsa M Wheelock, Barbro Dahlén, Björn Nordlund, Kjell Alving, Gunilla Hedlin, Kian Fan Chung, Ian M Adcock, Peter J Sterk, Ratko Djukanovic, Sven-Erik Dahlén, Craig E Wheelock, U-BIOPRED Study Group, on behalf of the U-BIOPRED Study Group, H Ahmed, C Auffray, A T Bansal, E H Bel, J Bigler, B Billing, F Baribaud, H Bisgaard, M J Boedigheimer, K Bønnelykke, J Brandsma, P Brinkman, E Bucchioni, D Burg, A Bush, A Chaiboonchoe, C H Compton, J Corfield, D Cunoosamy, A D'Amico, B De Meulder, V J Erpenbeck, D Erzen, K Fichtner, N Fitch, L J Fleming, E Formaggio, U Frey, M Gahlemann, V Goss, Y Guo, S Hashimoto, J Haughney, P W Hekking, T Higenbottam, J M Hohlfeld, A J Knox, N Lazarinis, D Lefaudeux, M J Loza, R Lutter, A Manta, S Masefield, J G Matthews, A Mazein, A Meiser, R J M Middelveld, M Miralpeix, N Mores, C S Murray, J Musial, D Myles, L Pahus, S Pavlidis, A Postle, P Powel, G Praticò, M PuigValls, N Rao, A Roberts, G Roberts, A Rowe, T Sandström, J P R Schofield, W Seibold, A Selby, R Sigmund, F Singer, P J Skipp, M Smicker, K Sun, B Thornton, M Uddin, W M van Aalderen, M van Geest, J Vestbo, N H Vissing, A H Wagener, S S Wagers, Z Weiszhart, S J Wilson, J Östling, H Ahmed, C Auffray, A T Bansal, E H Bel, J Bigler, B Billing, F Baribaud, H Bisgaard, M J Boedigheimer, K Bønnelykke, J Brandsma, P Brinkman, E Bucchioni, D Burg, A Bush, A Chaiboonchoe, C H Compton, J Corfield, D Cunoosamy, A D'Amico, B De Meulder, V J Erpenbeck, D Erzen, K Fichtner, N Fitch, L J Fleming, E Formaggio, U Frey, M Gahlemann, V Goss, Y Guo, S Hashimoto, J Haughney, P W Hekking, T Higenbottam, J M Hohlfeld, A J Knox, N Lazarinis, D Lefaudeux, M J Loza, R Lutter, A Manta, S Masefield, J G Matthews, A Mazein, A Meiser, R J M Middelveld, M Miralpeix, N Mores, C S Murray, J Musial, D Myles, L Pahus, S Pavlidis, A Postle, P Powel, G Praticò, M PuigValls, N Rao, A Roberts, G Roberts, A Rowe, T Sandström, J P R Schofield, W Seibold, A Selby, R Sigmund, F Singer, P J Skipp, M Smicker, K Sun, B Thornton, M Uddin, W M van Aalderen, M van Geest, J Vestbo, N H Vissing, A H Wagener, S S Wagers, Z Weiszhart, S J Wilson, J Östling, Johan Kolmert, Cristina Gómez, David Balgoma, Marcus Sjödin, Johan Bood, Jon R Konradsen, Magnus Ericsson, John-Olof Thörngren, Anna James, Maria Mikus, Ana R Sousa, John H Riley, Stewart Bates, Per S Bakke, Ioannis Pandis, Massimo Caruso, Pascal Chanez, Stephen J Fowler, Thomas Geiser, Peter Howarth, Ildikó Horváth, Norbert Krug, Paolo Montuschi, Marek Sanak, Annelie Behndig, Dominick E Shaw, Richard G Knowles, Cécile T J Holweg, Åsa M Wheelock, Barbro Dahlén, Björn Nordlund, Kjell Alving, Gunilla Hedlin, Kian Fan Chung, Ian M Adcock, Peter J Sterk, Ratko Djukanovic, Sven-Erik Dahlén, Craig E Wheelock, U-BIOPRED Study Group, on behalf of the U-BIOPRED Study Group, H Ahmed, C Auffray, A T Bansal, E H Bel, J Bigler, B Billing, F Baribaud, H Bisgaard, M J Boedigheimer, K Bønnelykke, J Brandsma, P Brinkman, E Bucchioni, D Burg, A Bush, A Chaiboonchoe, C H Compton, J Corfield, D Cunoosamy, A D'Amico, B De Meulder, V J Erpenbeck, D Erzen, K Fichtner, N Fitch, L J Fleming, E Formaggio, U Frey, M Gahlemann, V Goss, Y Guo, S Hashimoto, J Haughney, P W Hekking, T Higenbottam, J M Hohlfeld, A J Knox, N Lazarinis, D Lefaudeux, M J Loza, R Lutter, A Manta, S Masefield, J G Matthews, A Mazein, A Meiser, R J M Middelveld, M Miralpeix, N Mores, C S Murray, J Musial, D Myles, L Pahus, S Pavlidis, A Postle, P Powel, G Praticò, M PuigValls, N Rao, A Roberts, G Roberts, A Rowe, T Sandström, J P R Schofield, W Seibold, A Selby, R Sigmund, F Singer, P J Skipp, M Smicker, K Sun, B Thornton, M Uddin, W M van Aalderen, M van Geest, J Vestbo, N H Vissing, A H Wagener, S S Wagers, Z Weiszhart, S J Wilson, J Östling, H Ahmed, C Auffray, A T Bansal, E H Bel, J Bigler, B Billing, F Baribaud, H Bisgaard, M J Boedigheimer, K Bønnelykke, J Brandsma, P Brinkman, E Bucchioni, D Burg, A Bush, A Chaiboonchoe, C H Compton, J Corfield, D Cunoosamy, A D'Amico, B De Meulder, V J Erpenbeck, D Erzen, K Fichtner, N Fitch, L J Fleming, E Formaggio, U Frey, M Gahlemann, V Goss, Y Guo, S Hashimoto, J Haughney, P W Hekking, T Higenbottam, J M Hohlfeld, A J Knox, N Lazarinis, D Lefaudeux, M J Loza, R Lutter, A Manta, S Masefield, J G Matthews, A Mazein, A Meiser, R J M Middelveld, M Miralpeix, N Mores, C S Murray, J Musial, D Myles, L Pahus, S Pavlidis, A Postle, P Powel, G Praticò, M PuigValls, N Rao, A Roberts, G Roberts, A Rowe, T Sandström, J P R Schofield, W Seibold, A Selby, R Sigmund, F Singer, P J Skipp, M Smicker, K Sun, B Thornton, M Uddin, W M van Aalderen, M van Geest, J Vestbo, N H Vissing, A H Wagener, S S Wagers, Z Weiszhart, S J Wilson, J Östling

Abstract

Rationale: New approaches are needed to guide personalized treatment of asthma.Objectives: To test if urinary eicosanoid metabolites can direct asthma phenotyping.Methods: Urinary metabolites of prostaglandins (PGs), cysteinyl leukotrienes (CysLTs), and isoprostanes were quantified in the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) study including 86 adults with mild-to-moderate asthma (MMA), 411 with severe asthma (SA), and 100 healthy control participants. Validation was performed internally in 302 participants with SA followed up after 12-18 months and externally in 95 adolescents with asthma.Measurement and Main Results: Metabolite concentrations in healthy control participants were unrelated to age, body mass index, and sex, except for the PGE2 pathway. Eicosanoid concentrations were generally greater in participants with MMA relative to healthy control participants, with further elevations in participants with SA. However, PGE2 metabolite concentrations were either the same or lower in male nonsmokers with asthma than in healthy control participants. Metabolite concentrations were unchanged in those with asthma who adhered to oral corticosteroid treatment as documented by urinary prednisolone detection, whereas those with SA treated with omalizumab had lower concentrations of LTE4 and the PGD2 metabolite 2,3-dinor-11β-PGF2α. High concentrations of LTE4 and PGD2 metabolites were associated with lower lung function and increased amounts of exhaled nitric oxide and eosinophil markers in blood, sputum, and urine in U-BIOPRED participants and in adolescents with asthma. These type 2 (T2) asthma associations were reproduced in the follow-up visit of the U-BIOPRED study and were found to be as sensitive to detect T2 inflammation as the established biomarkers.Conclusions: Monitoring of urinary eicosanoids can identify T2 asthma and introduces a new noninvasive approach for molecular phenotyping of adult and adolescent asthma.Clinical trial registered with www.clinicaltrials.gov (NCT01976767).

Keywords: U-BIOPRED; mass spectrometry; severe asthma; type 2 inflammation; urinary eicosanoid metabolites.

Figures

Figure 1.
Figure 1.
Schematic overview of arachidonic acid–derived lipid mediators (eicosanoids) following both enzymatic and nonenzymatic metabolism. Blue text indicates the known or proposed biologic effect of the indicated pathway. Gray boxes highlight eicosanoids quantified in urine from participants in the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) study by UPLC-MS/MS. 5-LOX = 5-lipoxygenase; COX = cyclooxygenase; cPLA2 = cytosolic phospholipase A2; FLAP = five lipoxygenase-activating protein; iPF2α = isoprostane-F2α; LT = leukotriene; LTC4S = LTC4-synthase; PG = prostaglandin; PGDS = PGD-synthase; PGES = PGE-synthases; PGFS = PGF-synthase; PGIS = PGI-synthase; tetranorPGDM = tetranor PGD2 metabolite; tetranorPGEM = tetranor PGE2 metabolite; TX = thromboxane; TXAS = TXA-synthase; UPLC–MS/MS = ultraperformance liquid chromatography–tandem mass spectrometry.
Figure 2.
Figure 2.
Median (interquartile range) urinary concentration of individual eicosanoids in the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) adult baseline healthy participant (healthy control) group (n = 100). TetranorPGEM concentrations are stratified by sex. CysLT = cysteinyl LT; iPF2α = isoprostane-F2α; LT = leukotriene; PG = prostaglandin; tetranorPGDM = tetranor PGD2 metabolite; tetranorPGEM = tetranor PGE2 metabolite; TX = thromboxane.
Figure 3.
Figure 3.
Distribution of urinary eicosanoid concentrations in HC (n = 100), MMA (n = 86), SAn (n = 302), and SAs/ex (n = 109) for (A and B) PGD2 metabolites, (C) PGF2α, (D) LTE4, (E and F) thromboxane metabolites, (GI) isoprostanes, (J) PGE2, and (K and L) and PGE2 metabolites. Data are plotted on a log2 scale. Boxes highlight the interquartile range with the group median; bars display the total distribution range (minimum to maximum). Significant group differences are indicated by P values determined by the Mann-Whitney U test. HC = healthy control participants; iPF2α = isoprostane-F2α; LT = leukotriene; MMA = participants with mild-to-moderate asthma; PG = prostaglandin; SAn = nonsmokers with severe asthma; SAs/ex = smokers or ex-smokers with severe asthma; tetranorPGDM = tetranor PGD2 metabolite; tetranorPGEM = tetranor PGE2 metabolite; TX = thromboxane.
Figure 4.
Figure 4.
Effect of oral corticosteroids (OCS) on observed urinary eicosanoid concentrations. (A) Stratification of adult U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) participants with severe asthma according to reported daily use of OCS and detection of prednisolone or prednisolone metabolites in urine (OCS, n = 90; no OCS, n = 167) (data from Table 4). (B) Median (interquartile range [IQR]) of urinary LTE4, tetranorPGDM, and 2,3-dinor-11β-PGF2α concentrations across participants with detectable urinary prednisolone in the adult U-BIOPRED study. Of the 90 individuals in which prednisolone or its metabolites were detected, parent prednisolone was only detected in 68 individuals. Quantified urinary prednisolone is stratified as follows: <500 (n = 17), 500–2,000 (n = 27), and >2,000 ng/ml (n = 24). (C) Median (IQR) of urinary concentration of CysLT and PGD2 metabolites across the three inhaled corticosteroid budesonide dose groups in adolescent children from the Swedish Search study. Budesonide eq. is stratified as follows: <500 (n = 38), 500–<1,000 (n = 46), and ≥1,000 μg (n = 11). CysLT = cysteinyl LT; eq. = equivalents; iPF2α = isoprostane-F2α; LT = leukotriene; PG = prostaglandin; tetranorPGDM = tetranor PGD2 metabolite; tetranorPGEM = tetranor PGE2 metabolite; TX = thromboxane.
Figure 5.
Figure 5.
Participants with SA from the adult U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) study with a history of omalizumab treatment (n = 52) were compared in a case-control design (1:2) to a group (n = 104) matched for similarities in serum IgE. The violin-plot horizontal lines correspond to group median and interquartile-range values for the two PGD2 metabolites and LTE4. Group comparisons were evaluated by the Mann-Whitney U test (data from Table E3). LT = leukotriene; PG = prostaglandin; SA = severe asthma; tetranorPGDM = tetranor PGD2 metabolite.
Figure 6.
Figure 6.
Selection of U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) adult participants with mild-to-moderate asthma, nonsmokers with severe asthma, and smokers/ex-smokers with severe asthma at baseline using the 25th and 75th concentration percentile of urinary LTE4 and calculated composite (log2-transformed and z-scored) variables for PGD2 metabolites (c-PGD2). A complete list of significant associations is provided in Table E6. At the 12- to 18-month follow-up visit (longitudinal), 302 participants with severe asthma were used for internal validation of the type 2 associations. Data are presented as the median and interquartile range and were evaluated by using the Mann-Whitney U test. c-PGD2 = combined PGD2 metabolites; FeNO = fractional exhaled nitric oxide; LTE4 = leukotriene E4; PGD2 = prostaglandin D2; ppb = parts per billion.
Figure 7.
Figure 7.
Validation of adult type 2 associations in adolescent participants from the Swedish Search study. Participants with severe or controlled persistent asthma were stratified using the 25th and 75th concentration percentile of urinary metabolites of PGD2 and CysLTs. For each percentile, the corresponding variable group median (interquartile range) differences were evaluated by the Mann-Whitney U test. *Total urinary PGD2 metabolites were determined by enzyme immunoassay, which has a 9:1 binding ratio toward 2,3-dinor-11β-PGF2α:11β-PGF2α as determined by a cross-reactivity test and ultraperformance liquid chromatography–tandem mass spectrometry (9). CysLT = cysteinyl leukotrienes; EDN = eosinophil-derived neurotoxin; FeNO = fractional exhaled nitric oxide; PGD2 = prostaglandin D2; PGF2α = prostaglandin F2α; ppb = parts per billion.
Figure 8.
Figure 8.
Relationship between proposed type 2 (T2) markers (A) at baseline and (B) at the 12- to 18-month longitudinal follow-up visit in the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) study. The heat map displays the percentage overlap between high amounts of four common T2 markers and the two urinary eicosanoid markers leukotriene E4 (LTE4) and combined prostaglandin D2 metabolites (c-PGD2). Established cutoffs (21) for the T2 markers were used (blood eosinophils ≥ 300 counts/μl, FeNO ≥ 30 ppb, periostin ≥ 55 ng/ml, and IgE ≥ 150 IU/ml). Cutoff values for the urinary metabolites were calculated from the median + 1 SD in the healthy control group (Table 3). Each cell represents the percentage of participants satisfying both cutoffs for a given comparison. For example, in the first row in A, 62% of the n = 195 participants with blood eosinophils ≥ 300 counts/μl also have urinary LTE4 ≥ 6.4 ng/mmol creatinine. The total number of participants positive for each row criterion is displayed in the far-right column. Serum IgE data were only available at baseline. c-PGD2 is a z-scored composite variable consisting of tetranorPGDM and 2,3-dinor-11β-PGF2α (see Methods). FeNO = fractional exhaled nitric oxide; ppb = parts per billion; tetranorPGDM = tetranor PGD2 metabolite.
Figure 9.
Figure 9.
Multivariate correlation analysis between the latent variable consisting of the markers employed to calculate the Refractory Asthma Stratification Program (RASP) type 2 severity score (B-eos, FeNO, and serum periostin) (21) and the latent variable representing concentrations of the three urinary eicosanoid metabolites: LTE4, tetranorPGDM, and 2,3-dinor-11β-PGF2α. The correlation for participants with a high RASP score with urinary eicosanoid concentrations was significant across all U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Diseases Outcomes) asthma groups at both (A) baseline (n = 112) and (B) the longitudinal follow-up (n = 52). In contrast, the correlation for baseline participants with a (C) intermediate (n = 200) or (D) low (n = 75) RASP score displayed no relevant correlation with the urinary eicosanoid concentrations, as evidenced by the flat slopes (y = 0.04x). PGD2-metabolites include tetranorPGDM and 2,3-dinor-11β-PGF2α. B-eos = blood eosinophils; FeNO = fractional exhaled nitric oxide; LTE4 = leukotriene E4; MMA = participants with mild-to-moderate asthma; PGD2 = prostaglandin D2; SAn = nonsmokers with severe asthma; SAs/ex = smokers or ex-smokers with severe asthma; t = scores vector for x-block, calculated from the variables defined in brackets; tetranorPGDM = tetranor PGD2 metabolite; u = scores vector for y-block, calculated from the variables defined in brackets.

References

    1. Papi A, Brightling C, Pedersen SE, Reddel HK. Asthma. Lancet. 2018;391:783–800.
    1. Diamant Z, Vijverberg S, Alving K, Bakirtas A, Bjermer L, Custovic A, et al. Toward clinically applicable biomarkers for asthma: an EAACI position paper. Allergy. 2019;74:1835–1851.
    1. Shaw DE, Sousa AR, Fowler SJ, Fleming LJ, Roberts G, Corfield J, et al. U-BIOPRED Study Group. Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort. Eur Respir J. 2015;46:1308–1321.
    1. Dennis EA, Norris PC. Eicosanoid storm in infection and inflammation. Nat Rev Immunol. 2015;15:511–523.
    1. Dahlén SE, Hedqvist P, Hammarström S, Samuelsson B. Leukotrienes are potent constrictors of human bronchi. Nature. 1980;288:484–486.
    1. Peters-Golden M, Henderson WR., Jr Leukotrienes. N Engl J Med. 2007;357:1841–1854.
    1. Dahlén S-E, Malmström K, Nizankowska E, Dahlén B, Kuna P, Kowalski M, et al. Improvement of aspirin-intolerant asthma by montelukast, a leukotriene antagonist: a randomized, double-blind, placebo-controlled trial. Am J Respir Crit Care Med. 2002;165:9–14.
    1. Israel E, Rubin P, Kemp JP, Grossman J, Pierson W, Siegel SC, et al. The effect of inhibition of 5-lipoxygenase by zileuton in mild-to-moderate asthma. Ann Intern Med. 1993;119:1059–1066.
    1. Bood JR, Sundblad B-M, Delin I, Sjödin M, Larsson K, Anderson SD, et al. Urinary excretion of lipid mediators in response to repeated eucapnic voluntary hyperpnea in asthmatic subjects. J Appl Physiol (1985) 2015;119:272–279.
    1. Kolmert J, Fauland A, Fuchs D, Säfholm J, Gómez C, Adner M, et al. Lipid mediator quantification in isolated human and guinea pig airways: an expanded approach for respiratory research. Anal Chem. 2018;90:10239–10248.
    1. Bauer J, Ripperger A, Frantz S, Ergün S, Schwedhelm E, Benndorf RA. Pathophysiology of isoprostanes in the cardiovascular system: implications of isoprostane-mediated thromboxane A2 receptor activation. Br J Pharmacol. 2014;171:3115–3131.
    1. Milne GL, Yin H, Hardy KD, Davies SS, Roberts LJ., II Isoprostane generation and function. Chem Rev. 2011;111:5973–5996.
    1. Kumlin M, Dahlén B, Björck T, Zetterström O, Granström E, Dahlén SE. Urinary excretion of leukotriene E4 and 11-dehydro-thromboxane B2 in response to bronchial provocations with allergen, aspirin, leukotriene D4, and histamine in asthmatics. Am Rev Respir Dis. 1992;146:96–103.
    1. O’Sullivan S, Dahlén B, Dahlén SE, Kumlin M. Increased urinary excretion of the prostaglandin D2 metabolite 9 alpha, 11 beta-prostaglandin F2 after aspirin challenge supports mast cell activation in aspirin-induced airway obstruction. J Allergy Clin Immunol. 1996;98:421–432.
    1. Gómez C, Gonzalez-Riano C, Barbas C, Kolmert J, Hyung Ryu M, Carlsten C, et al. Quantitative metabolic profiling of urinary eicosanoids for clinical phenotyping. J Lipid Res. 2019;60:1164–1173.
    1. Sjödin M, Kolmert J, Balgoma D, Delin I, Wheelock CE, Dahlén SE. Urinary LTE4 is a new strong predictor of TH2-driven asthma: Initial data from the Pan-European U-BIOPRED IMI project [abstract] ERS. 2014
    1. Wheelock C, Kolmert J, Lefaudeux D, Sjödin M, Balgoma D, Sousa A, et al. Non-invasive sub-phenotyping of asthma in the U-BIOPRED study by analysis of urinary lipid mediator excretion patterns [abstract] Am J Respir Crit Care Med. 2016;193:A4632.
    1. Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43:343–373. [Published erratum appears in Eur Respir J 43:1216.]
    1. Konradsen JR, Skantz E, Nordlund B, Lidegran M, James A, Ono J, et al. Predicting asthma morbidity in children using proposed markers of Th2-type inflammation. Pediatr Allergy Immunol. 2015;26:772–779.
    1. Balgoma D, Larsson J, Rokach J, Lawson JA, Daham K, Dahlén B, et al. Quantification of lipid mediator metabolites in human urine from asthma patients by electrospray ionization mass spectrometry: controlling matrix effects. Anal Chem. 2013;85:7866–7874.
    1. Hanratty CE, Matthews JG, Arron JR, Choy DF, Pavord ID, Bradding P, et al. RASP-UK (Refractory Asthma Stratification Programme) Consortium. A randomised pragmatic trial of corticosteroid optimization in severe asthma using a composite biomarker algorithm to adjust corticosteroid dose versus standard care: study protocol for a randomised trial. Trials. 2018;19:5.
    1. Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, et al. Long-term smoking alters abundance of over half of the proteome in bronchoalveolar lavage cell in smokers with normal spirometry, with effects on molecular pathways associated with COPD. Respir Res. 2018;19:40.
    1. Hayashi H, Mitsui C, Nakatani E, Fukutomi Y, Kajiwara K, Watai K, et al. Omalizumab reduces cysteinyl leukotriene and 9α,11β-prostaglandin F2 overproduction in aspirin-exacerbated respiratory disease. J Allergy Clin Immunol. 2016;137:1585–1587, e4.
    1. Grootendorst DC, Dahlén S-E, Van Den Bos JW, Duiverman EJ, Veselic-Charvat M, Vrijlandt EJLE, et al. Benefits of high altitude allergen avoidance in atopic adolescents with moderate to severe asthma, over and above treatment with high dose inhaled steroids. Clin Exp Allergy. 2001;31:400–408.
    1. Daham K, James A, Balgoma D, Kupczyk M, Billing B, Lindeberg A, et al. Effects of selective COX-2 inhibition on allergen-induced bronchoconstriction and airway inflammation in asthma. J Allergy Clin Immunol. 2014;134:306–313.
    1. Taylor GW, Taylor I, Black P, Maltby NH, Turner N, Fuller RW, et al. Urinary leukotriene E4 after antigen challenge and in acute asthma and allergic rhinitis. Lancet. 1989;1:584–588.
    1. Fitzgerald DJ, Roy L, Catella F, FitzGerald GA. Platelet activation in unstable coronary disease. N Engl J Med. 1986;315:983–989.
    1. Gülen T, Möller Westerberg C, Lyberg K, Ekoff M, Kolmert J, Bood J, et al. Assessment of in vivo mast cell reactivity in patients with systemic mastocytosis. Clin Exp Allergy. 2017;47:909–917.
    1. Seyberth HW, Sweetman BJ, Frolich JC, Oates JA. Quantifications of the major urinary metabolite of the E prostaglandins by mass spectrometry: evaluation of the method’s application to clinical studies. Prostaglandins. 1976;11:381–397.
    1. Lupinetti MD, Sheller JR, Catella F, Fitzgerald GA. Thromboxane biosynthesis in allergen-induced bronchospasm: evidence for platelet activation. Am Rev Respir Dis. 1989;140:932–935.
    1. Capra V, Rovati GE, Mangano P, Buccellati C, Murphy RC, Sala A. Transcellular biosynthesis of eicosanoid lipid mediators. Biochim Biophys Acta. 2015;1851:377–382.
    1. Säfholm J, Manson ML, Bood J, Delin I, Orre A-C, Bergman P, et al. Prostaglandin E2 inhibits mast cell-dependent bronchoconstriction in human small airways through the E prostanoid subtype 2 receptor. J Allergy Clin Immunol. 2015;136:1232–9.e1.
    1. Cahill KN, Cui J, Kothari P, Murphy K, Raby BA, Singer J, et al. Unique effect of aspirin therapy on biomarkers in aspirin-exacerbated respiratory disease: a prospective trial. Am J Respir Crit Care Med. 2019;200:704–711.
    1. Maric J, Ravindran A, Mazzurana L, Björklund ÅK, Van Acker A, Rao A, et al. Prostaglandin E2 suppresses human group 2 innate lymphoid cell function. J Allergy Clin Immunol. 2018;141:1761–1773, e6.
    1. Manso G, Baker AJ, Taylor IK, Fuller RW. In vivo and in vitro effects of glucocorticosteroids on arachidonic acid metabolism and monocyte function in nonasthmatic humans. Eur Respir J. 1992;5:712–716.
    1. Gyllfors P, Dahlén S-E, Kumlin M, Larsson K, Dahlén B. Bronchial responsiveness to leukotriene D4 is resistant to inhaled fluticasone propionate. J Allergy Clin Immunol. 2006;118:78–83.
    1. Sebaldt RJ, Sheller JR, Oates JA, Roberts LJ, II, FitzGerald GA. Inhibition of eicosanoid biosynthesis by glucocorticoids in humans. Proc Natl Acad Sci U S A. 1990;87:6974–6978.
    1. Dworski R, Fitzgerald GA, Oates JA, Sheller JR. Effect of oral prednisone on airway inflammatory mediators in atopic asthma. Am J Respir Crit Care Med. 1994;149:953–959.
    1. Vachier I, Kumlin M, Dahlén SE, Bousquet J, Godard P, Chanez P. High levels of urinary leukotriene E4 excretion in steroid treated patients with severe asthma. Respir Med. 2003;97:1225–1229.
    1. Hayashi H, Fukutomi Y, Mitsui C, Kajiwara K, Watai K, Kamide Y, et al. Omalizumab for aspirin hypersensitivity and leukotriene overproduction in aspirin-exacerbated respiratory disease: a randomized controlled trial. Am J Respir Crit Care Med. 2020;201:1488–1498.
    1. Uematsu T, Kanamaru M, Kosuge K, Hara K, Uchiyama N, Takenaga N, et al. Pharmacokinetic and pharmacodynamic analysis of a novel leukotriene biosynthesis inhibitor, MK-0591, in healthy volunteers. Br J Clin Pharmacol. 1995;40:59–66.
    1. Dahlén B, Nizankowska E, Szczeklik A, Zetterström O, Bochenek G, Kumlin M, et al. Benefits from adding the 5-lipoxygenase inhibitor zileuton to conventional therapy in aspirin-intolerant asthmatics. Am J Respir Crit Care Med. 1998;157:1187–1194.
    1. Carmella SG, Heskin AK, Tang MK, Jensen J, Luo X, Le CT, et al. Longitudinal stability in cigarette smokers of urinary eicosanoid biomarkers of oxidative damage and inflammation. PLoS One. 2019;14:e0215853.
    1. Inoue Y, Izuhara K, Ohta S, Ono J, Shimojo N. No increase in the serum periostin level is detected in elementary school-age children with allergic diseases. Allergol Int. 2015;64:289–290.
    1. van ’t Erve TJ, Kadiiska MB, London SJ, Mason RP. Classifying oxidative stress by F2-isoprostane levels across human diseases: a meta-analysis. Redox Biol. 2017;12:582–599.
    1. Serhan CN, Levy BD. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J Clin Invest. 2018;128:2657–2669.
    1. Duvall MG, Bruggemann TR, Levy BD. Bronchoprotective mechanisms for specialized pro-resolving mediators in the resolution of lung inflammation. Mol Aspects Med. 2017;58:44–56.
    1. Gijón MA, Almstrand A-C, Johnson CA, Murphy RC, Zarini S. Identification of metabolites of maresin 1 in human neutrophils [abstract] FASEB J. 2017;31:lb230.
    1. Balas L, Risé P, Gandrath D, Rovati G, Bolego C, Stellari F, et al. Rapid metabolization of protectin D1 by β-oxidation of its polar head chain. J Med Chem. 2019;62:9961–9975.
    1. Kolmert J, Forngren B, Lindberg J, Öhd J, Åberg KM, Nilsson G, et al. A quantitative LC/MS method targeting urinary 1-methyl-4-imidazoleacetic acid for safety monitoring of the global histamine turnover in clinical studies. Anal Bioanal Chem. 2014;406:1751–1762.

Source: PubMed

3
订阅