Ticagrelor Reduces Thromboinflammatory Markers in Patients With Pneumonia

Travis R Sexton, Guoying Zhang, Tracy E Macaulay, Leigh A Callahan, Richard Charnigo, Olga A Vsevolozhskaya, Zhenyu Li, Susan Smyth, Travis R Sexton, Guoying Zhang, Tracy E Macaulay, Leigh A Callahan, Richard Charnigo, Olga A Vsevolozhskaya, Zhenyu Li, Susan Smyth

Abstract

Despite treatment advances for sepsis and pneumonia, significant improvements in outcome have not been realized. Antiplatelet therapy may improve outcome in pneumonia and sepsis. In this study, the authors show that ticagrelor reduced leukocytes with attached platelets as well as the inflammatory biomarker interleukin (IL)-6. Pneumonia patients receiving ticagrelor required less supplemental oxygen and lung function tests trended toward improvement. Disruption of the P2Y12 receptor in a murine model protected against inflammatory response, lung permeability, and mortality. Results indicate a mechanistic link between platelets, leukocytes, and lung injury in settings of pneumonia and sepsis, and suggest possible therapeutic approaches to reduce complications.(Targeting Platelet-Leukocyte Aggregates in Pneumonia With Ticagrelor [XANTHIPPE]; NCT01883869).

Keywords: ADP, adenosine diphosphate; CAP, community-acquired pneumonia; CI, confidence interval; COPD, chronic obstructive pulmonary disease; ELISA, enzyme-linked immunosorbent assay; FEV-1, forced expiratory volume in 1 s; HAP, hospital-acquired pneumonia; IL, interleukin; IQR, interquartile range; Kfc, capillary filtration coefficient; LPS, lipopolysaccharide; LTA, light transmission aggregometry; MPO, myeloperoxidase; MVV, maximum ventilation velocity; NE, neutrophil elastase; NET, neutrophil extracellular trap; OR, odds ratio; PRP, platelet-rich plasma; TNF, tumor necrosis factor; TRAP, thrombin receptor activating peptide; WT, wild-type; dsDNA, doubled-stranded DNA; inflammation; leukocytes; platelets; pneumonia; sepsis.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Ticagrelor Reduces Ex Vivo ADP-Induced Platelet Aggregation in Pneumonia Patients Maximum platelet aggregation was measured by light transmission aggregometry in PRP in response to 5 μmol/l ADP (A) and 15 μmol/l TRAP (C). Area under the curve values in the Multiplate assay are shown with ADP (B) or TRAP (D) as an agonist. Patients randomized to placebo are represented by open circles and patients randomized to ticagrelor are represented by filled circles. A significant difference between groups on the change from baseline to 24 h is indicated by an asterisk. ADP = adenosine diphosphate; AU = arbitrary units; BL = baseline; LTA = light transmission aggregometry; TRAP = thrombin receptor activating peptide
Figure 2
Figure 2
Ticagrelor Reduces Platelet–Leukocyte Aggregates The percentage of leukocytes with attached platelets for each subject is plotted at baseline (BL) and at 24 h in patients given placebo (A, open circles) and ticagrelor (A, filled circles). A significant difference between groups on the change from baseline to 24 h is indicated by an asterisk. Delta change of platelet–leukocyte aggregates at 24 h (compared with baseline) is represented for patients randomized to placebo (B, open circles and top dashed line) and ticagrelor (B, filled circles and bottom dashed line).
Figure 3
Figure 3
Ticagrelor and NETosis Biomarkers in Pneumonia Patients The delta change at 24 and 48 h after dosing for the MPO-NE NETosis assay (A) and circulating dsDNA (B) were measured in placebo patients (open circles) and ticagrelor patients (filled circles) that had samples through 48 h after dosing. Significant changes are indicated by an asterisk. dsDNA = doubled-stranded DNA; MPO = myeloperoxidase; NE = neutrophil elastase.
Figure 4
Figure 4
Ticagrelor and Lung Function Within 24 h in Pneumonia Patients Lung function was tested in subjects willing and able to undergo spirometry testing. Data were collected at baseline and at 24 h. Changes at 24 h from baseline (midline in all 3 graphs) are shown for tidal volume (A), FEV-1 (B), and MVV (C). Units reported are liters for tidal volume and FEV-1 (A, B) and liters per minute for MVV (C). Data from patients randomized to placebo are shown in the open boxes, whereas patients randomized to ticagrelor are represented in the shaded boxes. Each box represents the interquartile range (25% to 75%), whereas the whiskers represent the full range of the data. The vertical line in each interquartile range box represents the median value (note that the median for the placebo group in C overlaps with the midline). FEV-1 = forced expiratory volume in 1 s; MVV = maximum ventilation velocity.
Figure 5
Figure 5
Pneumonia Patients Taking Ticagrelor Became Less Reliant on Supplemental Oxygen Supplemental oxygen requirements were recorded from patient records. Data were collected at the time closest to when the patient received the study medication and 24 and 48 h following study dosage. Supplemental oxygen was recorded as none (room air), nasal cannula, and high-flow (HF) devices and mechanical ventilation. Patients randomized to ticagrelor demonstrated a progression to room air and away from supplemental oxygen (odds ratio [OR]: 1.08; 95% confidence interval [CI]: 1.01 to 1.15). Patients randomized to placebo did not change over 48 h after receiving the study dose (OR: 0.99; 95% CI: 0.95 to 1.04).
Figure 6
Figure 6
Ticagrelor Reduces Inflammation, Increases Lung Function, and Protects Against Mortality in a Murine Sepsis Model The concentrations of TNF-α (A) and IL-10 (B) at 0 h up to 8 h post-LPS injection are graphed for control mice and mice treated with ticagrelor or clopidogrel (A and B, reported as mean with bar indicating SE). Significant difference over the time course compared with the control is indicated by an asterisk in A and B. Survival curves of wild-type (WT) mice and P2y12−/− mice injected with LPS are plotted in C, whereas D shows survival curves of WT and P2y12−/− mice treated with clopidogrel or ticagrelor (T10) before LPS injection. Significance between mutant and WT (A) and P2Y12 antagonist treated and untreated (D) is indicated by an asterisk. Capillary filtration coefficients (KFC) are graphed for control mice and mice treated with ticagrelor or clopidogrel (E, reported as mean with bar indicating SE). Significant differences between treatment group and control are indicated by an asterisk. IL = interleukin; LPS = lipopolysaccharide; TNF = tumor necrosis factor.

References

    1. Restrepo M.I., Faverio P., Anzueto A. Long-term prognosis in community-acquired pneumonia. Curr Opin Infect Dis. 2013;26:151–158.
    1. Cangemi R., Casciaro M., Rossi E. Platelet activation is associated with myocardial infarction in patients with pneumonia. J Am Coll Cardiol. 2014;64:1917–1925.
    1. Chen L.F., Chen H.P., Huang Y.S., Huang K.Y., Chou P., Lee C.C. Pneumococcal pneumonia and the risk of stroke: a population-based follow-up study. PLoS One. 2012;7:e51452.
    1. Chiang C.H., Huang C.C., Chan W.L. Association between Mycoplasma pneumonia and increased risk of ischemic stroke: a nationwide study. Stroke. 2011;42:2940–2943.
    1. Mandal P., Chalmers J.D., Choudhury G., Akram A.R., Hill A.T. Vascular complications are associated with poor outcome in community-acquired pneumonia. QJM. 2011;104:489–495.
    1. Ribeiro D.D., Lijfering W.M., Van Hylckama Vlieg A., Rosendaal F.R., Cannegieter S.C. Pneumonia and risk of venous thrombosis: results from the MEGA study. J Thromb Haemost. 2012;10:1179–1182.
    1. Ruane L., Buckley T., Hoo S.Y.S. Triggering of acute myocardial infarction by respiratory infection. Intern Med J. 2017;47:522–529.
    1. Smeeth L., Cook C., Thomas S., Hall A.J., Hubbard R., Vallance P. Risk of deep vein thrombosis and pulmonary embolism after acute infection in a community setting. Lancet. 2006;367:1075–1079.
    1. Smeeth L., Thomas S.L., Hall A.J., Hubbard R., Farrington P., Vallance P. Risk of myocardial infarction and stroke after acute infection or vaccination. N Engl J Med. 2004;351:2611–2618.
    1. Caudrillier A., Kessenbrock K., Gilliss B.M. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J Clin Invest. 2012;122:2661–2671.
    1. Grommes J., Alard J.E., Drechsler M. Disruption of platelet-derived chemokine heteromers prevents neutrophil extravasation in acute lung injury. Am J Respir Crit Care Med. 2012;185:628–636.
    1. Weyrich A.S., Zimmerman G.A. Platelets in lung biology. Annu Rev Physiol. 2013;75:569–591.
    1. Gross A.K., Dunn S.P., Feola D.J. Clopidogrel treatment and the incidence and severity of community acquired pneumonia in a cohort study and meta-analysis of antiplatelet therapy in pneumonia and critical illness. J Thromb Thrombolysis. 2013;35:147–154.
    1. Winning J., Reichel J., Eisenhut Y. Anti-platelet drugs and outcome in severe infection: clinical impact and underlying mechanisms. Platelets. 2009;20:50–57.
    1. Winning J., Neumann J., Kohl M. Antiplatelet drugs and outcome in mixed admissions to an intensive care unit. Crit Care Med. 2010;38:32–37.
    1. Storey R.F., James S.K., Siegbahn A. Lower mortality following pulmonary adverse events and sepsis with ticagrelor compared to clopidogrel in the PLATO study. Platelets. 2014;25:517–525.
    1. Andell P., James S.K., Cannon C.P. Ticagrelor versus clopidogrel in patients with acute coronary syndromes and chronic obstructive pulmonary disease: an analysis from the Platelet Inhibition and Patient Outcomes (PLATO) trial. J Am Heart Assoc. 2015;4:e002490.
    1. Rondina M.T., Carlisle M., Fraughton T. Platelet-monocyte aggregate formation and mortality risk in older patients with severe sepsis and septic shock. J Gerontol A Biol Sci Med Sci. 2015;70:225–231.
    1. Czaikoski P.G., Mota J.M., Nascimento D.C. Neutrophil extracellular traps induce organ damage during experimental and clinical sepsis. PLoS One. 2016;11:e0148142.
    1. Lowe K.L., Finney B.A., Deppermann C. Podoplanin and CLEC-2 drive cerebrovascular patterning and integrity during development. Blood. 2015;125:3769–3777.
    1. Sexton T.R., Wallace E.L., Macaulay T.E. The effect of rosuvastatin on thromboinflammation in the setting of acute coronary syndrome. J Thromb Thrombolysis. 2015;39:186–195.
    1. Sexton T.R., Wallace E.L., Chen A. Thromboinflammatory response and predictors of outcomes in patients undergoing transcatheter aortic valve replacement. J Thromb Thrombolysis. 2016;41:384–393.
    1. Miller M.R., Hankinson J., Brusasco V. Standardisation of spirometry. Eur Respir J. 2005;26:319–338.
    1. Foster C.J., Prosser D.M., Agans J.M. Molecular identification and characterization of the platelet ADP receptor targeted by thienopyridine antithrombotic drugs. J Clin Invest. 2001;107:1591–1598.
    1. Zarbock A., Ley K. The role of platelets in acute lung injury (ALI) Front Biosci (Landmark Ed) 2009;14:150–158.
    1. Cannon C.P., Harrington R.A., James S. Comparison of ticagrelor with clopidogrel in patients with a planned invasive strategy for acute coronary syndromes (PLATO): a randomised double-blind study. Lancet. 2010;375:283–293.
    1. Seidel M., Winning J., Claus R.A., Bauer M., Losche W. Beneficial effect of clopidogrel in a mouse model of polymicrobial sepsis. J Thromb Haemost. 2009;7:1030–1032.
    1. Liverani E., Rico M.C., Tsygankov A.Y., Kilpatrick L.E., Kunapuli S.P. P2Y12 receptor modulates sepsis-induced inflammation. Arterioscler Thromb Vasc Biol. 2016;36:961–971.
    1. Winning J., Claus R.A., Pletz M.W., Bauer M., Losche W. Adenosine diphosphate receptor antagonist clopidogrel sulfate attenuates LPS-induced systemic inflammation in a rat model. Shock. 2011;36:317. author reply 318.
    1. Alexopoulos D., Galati A., Xanthopoulou I. Ticagrelor versus prasugrel in acute coronary syndrome patients with high on-clopidogrel platelet reactivity following percutaneous coronary intervention: a pharmacodynamic study. J Am Coll Cardiol. 2012;60:193–199.
    1. Li P., Yang Y., Chen T. Ticagrelor overcomes high platelet reactivity in patients with acute myocardial infarction or coronary artery in-stent restenosis: a randomized controlled trial. Sci Rep. 2015;5:13789.
    1. Cattaneo M., Schulz R., Nylander S. Adenosine-mediated effects of ticagrelor: evidence and potential clinical relevance. J Am Coll Cardiol. 2014;63:2503–2509.
    1. Tunjungputri R.N., Van Der Ven A.J., Schonsberg A. Reduced platelet hyperreactivity and platelet-monocyte aggregation in HIV-infected individuals receiving a raltegravir-based regimen. AIDS. 2014;28:2091–2096.
    1. Schoergenhofer C., Schwameis M., Hobl E.L. Potent irreversible P2Y12 inhibition does not reduce LPS-induced coagulation activation in a randomized, double-blind, placebo-controlled trial. Clin Sci (Lond) 2016;130:433–440.
    1. Akinosoglou K., Alexopoulos D. Use of antiplatelet agents in sepsis: a glimpse into the future. Thromb Res. 2014;133:131–138.
    1. Kuebler W.M. Selectins revisited: the emerging role of platelets in inflammatory lung disease. J Clin Invest. 2006;116:3106–3108.

Source: PubMed

3
S'abonner