TRAnexamic acid in hemorrhagic CESarean section (TRACES) randomized placebo controlled dose-ranging pharmacobiological ancillary trial: study protocol for a randomized controlled trial

Anne-Sophie Ducloy-Bouthors, Emmanuelle Jeanpierre, Imen Saidi, Anne-Sophie Baptiste, Elodie Simon, Damien Lannoy, Alain Duhamel, Delphine Allorge, Sophie Susen, Benjamin Hennart, Anne-Sophie Ducloy-Bouthors, Emmanuelle Jeanpierre, Imen Saidi, Anne-Sophie Baptiste, Elodie Simon, Damien Lannoy, Alain Duhamel, Delphine Allorge, Sophie Susen, Benjamin Hennart

Abstract

Background: Evidence increases that a high or a standard dose of tranexamic acid (TA) reduces postpartum bleeding. The TRACES pharmacobiological substudy aims to establish a therapeutic strategy in hemorrhagic (H) Cesarean section (CS) with respect to the intensity of fibrinolysis by using innovative assays.

Method/design: The TRACES trial is a multicenter, randomized, double-blind, placebo-controlled, TA dose-ranging study that measures simultaneously plasmatic and uterine and urine TA concentrations and the plasmin peak inhibition tested by a simultaneous thrombin plasmin generation assay described by Van Geffen (novel hemostasis assay [NHA]). Patients undergoing H CS (>800 mL) will receive blindly TA 0.5 g or 1 g or placebo. A non-hemorrhagic (NH) group will be recruited to establish plasmin generation profile. Venous blood will be sampled before, at the end, and then at 30, 60, 120, and 360 min after injection. Uterine bleeding will be sampled after injection. Urine will be sampled 2 h and 6 h after injection. The number of patients entered into the study will be 114 H + 48 NH out of the 390 patients of the TRACES clinical trial.

Discussion: To explore the two innovative assays, a preliminary pilot study was conducted. Blood samples were performed repeatedly in patients undergoing either a H (>800 mL) or NH (<800 mL) CS and in non-pregnant women (NP). H patients received TA (0-2 g). Dose-dependent TA plasmatic concentrations were determined by LC-MS/MS quantification. Plasmin generation and its inhibition were tested in vitro and in vivo using the simultaneous thrombin-plasmin generation assay (STPGA). The pilot study included 15 patients in the H group, ten patients in the NH group, and seven patients in the NP group. TA plasmatic concentration showed a dose-dependent variation. STPGA inter-assay variation coefficients were < 20% for all plasmin parameters. Inter-individual dispersion of plasmin generation capacity was higher in H and NH groups than in NP group. Profile evolution over time was different between groups. This preliminary technical validation study allows TRACES pharmacobiological trial to be conducted.

Trial registration: ClinicalTrials.gov, NCT02797119. Registered on 13 June 2016.

Keywords: Cesarean section; D-dimers; Fibrinolysis; Pharmacokinetics; Plasmin; Postpartum hemorrhage; Tranexamic acid.

Conflict of interest statement

Ethics approval and consent to participate

The TRACES trial obtained approval from the competent national authorities (ANSM 201500249926) and the Ethics Committee (CPP 15/50 020216) before beginning the study, in accordance with article L1121-4 of the Public Healthcare Code. According to the ethical approval, informed consent to participate will be obtained from all participants in the study. Informed consent to participate to the pilot study has been obtained from all participants. This trial has been declared on the clinical trials registration on 13 June 2016 under the number CT 02797119. Registration will be performed in accordance with decree dated 14 November 2006 about gathering data in the national register of individuals participating in biomedical research.

Consent for publication

Authors and sponsors have given their consent and defined the publication rules.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Inhibition of the natural hyperfibrinolysis by a high dose of TA administered at the early stage of PPH. D-Dimers and PAP complex evolution at the onset of PPH and 30, 120, and 360 min later in the untreated H group compared to the TA-treated group
Fig. 2
Fig. 2
TA plasma concentration measurement method: determination of the optimal timing. A timed urinary sample is collected in a graduated urinary bag between inclusion (T0) and T120, then T120 and T360, and end of the observation period of 6 h to measure the cumulative dose of TA excreted in urine
Fig. 3
Fig. 3
Parameters studied in the simultaneous thrombin–plasmin generation assay. Thrombin parameters: (1) lag time (min); (2) thrombin peak time (min); (3) thrombin peak (nM); and (4) area under the curve (AUC) (nM.min). Plasmin parameters: (5) plasmin peak (nM); (6) fibrin lysis time (FLT) (min); and (7) AUC during FLT (plasmin potential) (nM.min)
Fig. 4
Fig. 4
a In vitro STPGA with different TA concentrations: thrombin generation. In vitro dose effect relationship between TA and thrombin generation. Thrombin generation in the STPGA performed with a pooled plasma sample and different TA concentrations in the range of 0–20 mg/L. b In vitro STPGA with different TA concentrations: plasmin generation. In vitro dose effect relationship between TA and plasmin generation. Plasmin generation in the STPGA performed with a pooled plasma sample and different TA concentrations in the range of 0–20 mg/L
Fig. 5
Fig. 5
a In vivo study: TA dose impact on thrombin generation profiles at T30. In vivo thrombin generation variations at T30 depending on TA dose administered: four patients who received 0, 0.5, 1, and 2 g (IV bolus), respectively. b In vivo study: TA dose impact on plasmin generation profiles at T30. In vivo plasmin generation variations at T30 depending on TA dose administered: four patients who received 0, 0.5, 1, and 2 g (IV bolus), respectively.
Fig. 6
Fig. 6
Time repeated profiles: evolution of thrombin (a) and plasmin (b) profiles over time points: T0, T30, T60, T120, and T360, in patients in the NH group, the H group, and TA group with increasing doses of TA: TA1/2: 0.5 g, TA1: 1 g, and TA2: 2 g. A-NH evolution of thrombin generation profile over time for a NH patient, B-NH evolution of plasmin generation profile over time for a NH patient, A-0 evolution of thrombin generation profile over time for a H patient receiving 0 g TA, B-0 evolution of plasmin generation profile over time for a H patient receiving 0 g TA, A-TA1/2 evolution of thrombin generation profile over time for a H patient receiving 0.5 g TA, B-TA1/2 evolution of plasmin generation profile over time for a H patient receiving 0.5 g TA, A-1 evolution of thrombin generation profile over time for a H patient receiving 1 g TA, B-1 evolution of plasmin generation profile over time for a H patient receiving 1 g TA, A-2 evolution of thrombin generation profile over time for a H patient receiving 2 g TA, B-2 evolution of plasmin generation profile over time for a H patient receiving 2 g TA
Fig. 7
Fig. 7
Thrombin (a) and plasmin (b) AUCs measured in STPGA at T0 in NH and H + TA groups compared to the NP group. AUC area under the curve, NP non-pregnant group, NH non-hemorrhagic group, H hemorrhagic untreated group, TA hemorrhagic treated group. *Thrombin AUC in NP vs NH group: p = 0.02; **Plasmin AUC in NP vs NH group: p = 0.002
Fig. 8
Fig. 8
Plasmin generation profile at T0 (baseline) and corresponding TA peak plasma concentration at T1 (following injection) in TA-treated patients. Plasmin generation profiles at T0 (baseline, before injection) and corresponding TA peak plasma concentrations at T1 (just after injection, values in mg/L bracketed in little sidebars) of six patients treated by TA: two patients by dose (0.5, 1, and 2 g)
Fig. 9
Fig. 9
Kinetics of both TA plasma concentration and plasmin AUC in TA-treated patients: evolution of TA plasma concentration and plasmin AUC over time points: T0, T15, T30, T60, T120, T180, and T360, in six patients treated by TA (0.5, 1, or 2 g). a Kinetics of TA plasma concentration and plasmin AUC: group TA0.5 - patient 20. b Kinetics of TA plasma concentration and plasmin AUC: group TA0.5 - patient 19. c Kinetics of TA plasma concentration and plasmin AUC: group TA1 - patient 24. d Kinetics of TA plasma concentration and plasmin AUC: group TA1 - patient 25. e Kinetics of TA plasma concentration and plasmin AUC: group TA2 - patient 12. f Kinetics of TA plasma concentration and plasmin AUC: group TA2 - patient 17

References

    1. Ker K, Edwards P, Perel P, Shakur H. Effect of tranexamic acid on surgical bleeding: systematic review and cumulative metanalysis. BMJ. 2012;344:e3054. doi: 10.1136/bmj.e3054.
    1. McCormack PL. Tranexamic acid: a review of its use in the treatment of hyperfibrinolysis. Drugs. 2012;72:585–617. doi: 10.2165/11209070-000000000-00000.
    1. Ducloy-Bouthors A-S, Jude B, Duhamel A, Broisin F, Huissoud C, Keita-Meyer H, et al. High-dose tranexamic acid reduces blood loss in postpartum haemorrhage. Crit Care Lond Engl. 2011;15:R117. doi: 10.1186/cc10143.
    1. Ducloy-Bouthors AS, Duhamel A, Kipnis E, Tournoys A, Prado-Dupont A, Elkalioubie A, et al. Postpartum haemorrhage related early increase in D-dimers is inhibited by tranexamic acid: haemostasis parameters of a randomized controlled open labelled trial. Br J Anaesth. 2016;116:641–8. doi: 10.1093/bja/aew021.
    1. WOMAN Trial Collaborators Effect of early tranexamic acid administration on mortality, hysterectomy, and other morbidities in women with post-partum haemorrhage (WOMAN): an international, randomised, double-blind, placebo-controlled trial. Lancet. 2017;389:2105–16. doi: 10.1016/S0140-6736(17)30638-4.
    1. Walzman M, Bonnar J. Effects of tranexamic acid on the coagulation and fibrinolytic systems in pregnancy complicated by placental bleeding. Arch Toxicol Suppl Arch Toxikol Suppl. 1982;5:214–20. doi: 10.1007/978-3-642-68511-8_39.
    1. Fodstad H, Pilbrant A, Schannong M, Strömberg S. Determination of tranexamic acid (AMCA) and fibrin/fibrinogen degradation products in cerebrospinal fluid after aneurysmal subarachnoid haemorrhage. Acta Neurochir (Wien) 1981;58:1–13. doi: 10.1007/BF01401679.
    1. Nilsson IM. Clinical pharmacology of aminocaproic and tranexamic acids. J Clin Pathol Suppl (R Coll Pathol) 1980;14:41–7. doi: 10.1136/jcp.33.Suppl_14.41.
    1. Pilbrant A, Schannong M, Vessman J. Pharmacokinetics and bioavailability of tranexamic acid. Eur J Clin Pharmacol. 1981;20:65–72. doi: 10.1007/BF00554669.
    1. Astedt B. Clinical pharmacology of tranexamic acid. Scand J Gastroenterol Suppl. 1987;137:22–5. doi: 10.3109/00365528709089756.
    1. Horrow JC, Van Riper DF, Strong MD, Grunewald KE, Parmet JL. The dose-response relationship of tranexamic acid. Anesthesiology. 1995;82:383–92. doi: 10.1097/00000542-199502000-00009.
    1. Dowd NP, Karski JM, Cheng DC, Carroll JA, Lin Y, James RL, et al. Pharmacokinetics of tranexamic acid during cardiopulmonary bypass. Anesthesiology. 2002;97:390–9. doi: 10.1097/00000542-200208000-00016.
    1. Koster A, Schirmer U. Re-evaluation of the role of antifibrinolytic therapy with lysine analogs during cardiac surgery in the post aprotinin era. Curr Opin Anaesthesiol. 2011;24:92–7. doi: 10.1097/ACO.0b013e32833ff3eb.
    1. Sigaut S, Tremey B, Ouattara A, Couturier R, Taberlet C, Grassin-Delyle S, et al. Comparison of two doses of tranexamic acid in adults undergoing cardiac surgery with cardiopulmonary bypass. Anesthesiology. 2014;120:590–600. doi: 10.1097/ALN.0b013e3182a443e8.
    1. Goobie SM, Meier PM, Sethna NF, Soriano SG, Zurakowski D, Samant S, et al. Population pharmacokinetics of tranexamic acid in paediatric patients undergoing craniosynostosis surgery. Clin Pharmacokinet. 2013;52:267–76. doi: 10.1007/s40262-013-0033-1.
    1. Grassin-Delyle S, Couturier R, Abe E, Alvarez JC, Devillier P, Urien S. A practical tranexamic acid dosing scheme based on population pharmacokinetics in children undergoing cardiac surgery. Anesthesiology. 2013;118:853–62. doi: 10.1097/ALN.0b013e318283c83a.
    1. Faraoni D, Cacheux C, Van Aelbrouck C, Ickx BE, Barvais L, Levy JH. Effect of two doses of tranexamic acid on fibrinolysis evaluated by thromboelastography during cardiac surgery: a randomised, controlled study. Eur J Anaesthesiol. 2014;31:491–8. doi: 10.1097/EJA.0000000000000051.
    1. Couturier R, Rubatti M, Credico C, Louvain-Quintard V, Anerkian V, Doubine S, et al. Continuous or discontinuous tranexamic acid effectively inhibits fibrinolysis in children undergoing cardiac surgery with cardiopulmonary bypass. Blood Coagul Fibrinolysis Int J Haemost Thromb. 2014;25:259–65. doi: 10.1097/MBC.0000000000000051.
    1. Yee BE, Wissler RN, Zanghi CN, Feng C, Eaton MP. The effective concentration of tranexamic acid for inhibition of fibrinolysis in neonatal plasma in vitro. Anesth Analg. 2013;117:767–72. doi: 10.1213/ANE.0b013e3182a22258.
    1. Manji RA, Grocott HP, Leake J, Ariano RE, Manji JS, Menkis AH, et al. Seizures following cardiac surgery: the impact of tranexamic acid and other risk factors. Can J Anaesth J Can Anesth. 2012;59:6–13. doi: 10.1007/s12630-011-9618-z.
    1. Martin K, Knorr J, Breuer T, Gertler R, Macguill M, Lange R, et al. Seizures after open heart surgery: comparison of ε-aminocaproic acid and tranexamic acid. J Cardiothorac Vasc Anesth. 2011;25:20–5. doi: 10.1053/j.jvca.2010.10.007.
    1. Murkin JM, Falter F, Granton J, Young B, Burt C, Chu M. High-dose tranexamic Acid is associated with nonischemic clinical seizures in cardiac surgical patients. Anesth Analg. 2010;110:350–3. doi: 10.1213/ANE.0b013e3181c92b23.
    1. Jiménez JJ, Iribarren JL, Brouard M, Hernández D, Palmero S, Jiménez A, et al. Safety and effectiveness of two treatment regimes with tranexamic acid to minimize inflammatory response in elective cardiopulmonary bypass patients: a randomized double-blind, dose-dependent, phase IV clinical trial. J Cardiothorac Surg. 2011;6:138. doi: 10.1186/1749-8090-6-138.
    1. CRASH-2 trial collaborators. Shakur H, Roberts I, Bautista R, Caballero J, Coats T, et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomised, placebo-controlled trial. Lancet Lond Engl. 2010;376:23–32. doi: 10.1016/S0140-6736(10)60835-5.
    1. Charbit B, Mandelbrot L, Samain E, Baron G, Haddaoui B, Keita H, et al. The decrease of fibrinogen is an early predictor of the severity of postpartum hemorrhage. J Thromb Haemost JTH. 2007;5:266–73. doi: 10.1111/j.1538-7836.2007.02297.x.
    1. Cortet M, Deneux-Tharaux C, Dupont C, Colin C, Rudigoz R-C, Bouvier-Colle M-H, et al. Association between fibrinogen level and severity of postpartum haemorrhage: secondary analysis of a prospective trial. Br J Anaesth. 2012;108:984–9. doi: 10.1093/bja/aes096.
    1. Biron-Andréani C, Morau E, Schved J-F, Hédon B, Dechaud H. Amniotic fluid embolism with haemostasis complications: primary fibrinogenolysis or disseminated intravascular coagulation? Pathophysiol Haemost Thromb. 2003;33:170–1. doi: 10.1159/000077826.
    1. Annecke T, Geisenberger T, Kürzl R, Penning R, Heindl B. Algorithm-based coagulation management of catastrophic amniotic fluid embolism. Blood Coagul Fibrinolysis Int J Haemost Thromb. 2010;21:95–100. doi: 10.1097/MBC.0b013e328332cfe2.
    1. Svanberg L, Astedt B, Nilsson IM. Abruptio placentae--treatment with the fibrinolytic inhibitor tranexamic acid. Acta Obstet Gynecol Scand. 1980;59:127–30. doi: 10.3109/00016348009154628.
    1. van Geffen M, Loof A, Lap P, Boezeman J, Laros-van Gorkom BAP, Brons P, et al. A novel hemostasis assay for the simultaneous measurement of coagulation and fibrinolysis. Hematol Amst Neth. 2011;16:327–36.
    1. Delavenne X, Montbel A, Hodin S, Zufferey P, Basset T. Quantification of total and unbound tranexamic acid in human plasma by ultrafiltration liquid chromatography/tandem mass spectrometry: Application to pharmacokinetic analysis. J Pharm Biomed Anal. 2014;91:32–6. doi: 10.1016/j.jpba.2013.12.005.

Source: PubMed

3
Abonner