Efficacy and Safety of Tranexamic Acid in Prehospital Traumatic Hemorrhagic Shock: Outcomes of the Cal-PAT Study

Michael M Neeki, Fanglong Dong, Jake Toy, Reza Vaezazizi, Joe Powell, Nina Jabourian, Alex Jabourian, David Wong, Richard Vara, Kathryn Seiler, Troy W Pennington, Joe Powell, Chris Yoshida-McMath, Shanna Kissel, Katharine Schulz-Costello, Jamish Mistry, Matthew S Surrusco, Karen R O'Bosky, Daved Van Stralen, Daniel Ludi, Karl Sporer, Peter Benson, Eugene Kwong, Richard Pitts, John T Culhane, Rodney Borger, Michael M Neeki, Fanglong Dong, Jake Toy, Reza Vaezazizi, Joe Powell, Nina Jabourian, Alex Jabourian, David Wong, Richard Vara, Kathryn Seiler, Troy W Pennington, Joe Powell, Chris Yoshida-McMath, Shanna Kissel, Katharine Schulz-Costello, Jamish Mistry, Matthew S Surrusco, Karen R O'Bosky, Daved Van Stralen, Daniel Ludi, Karl Sporer, Peter Benson, Eugene Kwong, Richard Pitts, John T Culhane, Rodney Borger

Abstract

Introduction: The California Prehospital Antifibrinolytic Therapy (Cal-PAT) study seeks to assess the safety and impact on patient mortality of tranexamic acid (TXA) administration in cases of trauma-induced hemorrhagic shock. The current study further aimed to assess the feasibility of prehospital TXA administration by paramedics within the framework of North American emergency medicine standards and protocols.

Methods: This is an ongoing multi-centered, prospective, observational cohort study with a retrospective chart-review comparison. Trauma patients identified in the prehospital setting with signs of hemorrhagic shock by first responders were administered one gram of TXA followed by an optional second one-gram dose upon arrival to the hospital, if the patient still met inclusion criteria. Patients administered TXA make up the prehospital intervention group. Control group patients met the same inclusion criteria as TXA candidates and were matched with the prehospital intervention patients based on mechanism of injury, injury severity score, and age. The primary outcomes were mortality, measured at 24 hours, 48 hours, and 28 days. Secondary outcomes measured included the total blood products transfused and any known adverse events associated with TXA administration.

Results: We included 128 patients in the prehospital intervention group and 125 in the control group. Although not statistically significant, the prehospital intervention group trended toward a lower 24-hour mortality rate (3.9% vs 7.2% for intervention and control, respectively, p=0.25), 48-hour mortality rate (6.3% vs 7.2% for intervention and control, respectively, p=0.76), and 28-day mortality rate (6.3% vs 10.4% for intervention and control, respectively, p=0.23). There was no significant difference observed in known adverse events associated with TXA administration in the prehospital intervention group and control group. A reduction in total blood product usage was observed following the administration of TXA (control: 6.95 units; intervention: 4.09 units; p=0.01).

Conclusion: Preliminary evidence from the Cal-PAT study suggests that TXA administration may be safe in the prehospital setting with no significant change in adverse events observed and an associated decreased use of blood products in cases of trauma-induced hemorrhagic shock. Given the current sample size, a statistically significant decrease in mortality was not observed. Additionally, this study demonstrates that it may be feasible for paramedics to identify and safely administer TXA in the prehospital setting.

Conflict of interest statement

Conflicts of Interest: By the WestJEM article submission agreement, all authors are required to disclose all affiliations, funding sources and financial or management relationships that could be perceived as potential sources of bias. No author has professional or financial relationships with any companies that are relevant to this study. There are no conflicts of interest or sources of funding to declare.

Figures

Figure
Figure
Patient exclusion flow chart that compares patient selection in the prehospital intervention group and control group. TXA, tranexamic acid; EMS, emergency medical services.

References

    1. Injury and Violence, The Facts - 2014. Geneva, Switzerland: World Health Organization; 2014.
    1. WISQARS - Leading Causes of Death Reports, National and Regional, 1999–2014. Centers for Disease Control and Prevention; 2015. [Accessed Feb 23, 2016]. Available at:
    1. Data & Statistics (WISQARS™): Cost of Injury Reports. Centers for Disease Control and Prevention; 2014. [Accessed Feb 23, 2016].
    1. Brohi K, Singh J, Heron M, et al. Acute traumatic coagulopathy. J Trauma. 2003;54(6):1127–30.
    1. Niles SE, McLaughlin DF, Perkins JG, et al. Increased mortality associated with the early coagulopathy of trauma in combat casualties. J Trauma. 2008;64(6):1459–63. 1463–5.
    1. Kauvar D, Lefering R, Wade C. Impact of hemorrhage on trauma outcome: an overview of epidemiology, clinical presentations, and therapeutic considerations. J Trauma. 2006;60(6 Suppl):S3–11.
    1. Sauaia A, Moore F, Moore E, et al. Epidemiology of trauma deaths: a reassessment. J Trauma. 1995;38(2):185–93.
    1. Paudyal P, Smith J, Robinson M, et al. Tranexamic acid in major trauma: implementation and evaluation across South West England. Eur J Emerg Med. 2017;24(1):44–8.
    1. MacLeod JB, Lynn M, McKenney MG, et al. Early coagulopathy predicts mortality in trauma. J Trauma. 2003;55(1):39–44.
    1. Shackford SR, Mackersie RC, Holbrook TL, et al. The epidemiology of traumatic death. A population-based analysis. Arch Surg. 1993;128(5):571–5.
    1. Brown JB, Neal MD, Guyette FX, et al. Design of the Study of Tranexamic Acid during Air Medical Prehospital Transport (STAAMP) Trial: addressing the knowledge gaps. Prehosp Emerg Care. 2015;19(1):79–86.
    1. Shakur H, Roberts I, Bautista R, 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. 2010;376(9734):23–32.
    1. Roberts I, Shakur H, Afolabi A, et al. The importance of early treatment with tranexamic acid in bleeding trauma patients: an exploratory analysis of the CRASH-2 randomised controlled trial. Lancet. 2011;377(9771):1096–101.1101.e1–2.
    1. Roberts I, Shakur H, Coats T, et al. The CRASH-2 trial: a randomised controlled trial and economic evaluation of the effects of tranexamic acid on death, vascular occlusive events and transfusion requirement in bleeding trauma patients. Health Technol Assess. 2013;17(10):1–79.
    1. Morrison J, Dubose J, Rasmussen T, et al. Military Application of Tranexamic Acid in Trauma Emergency Resuscitation (MATTERs) Study. Arch Surg. 2012;147(2):113–9.
    1. Lipsky AM, Abracmovich A, Nadler R, et al. Transexamic acid in the prehospital setting: Israel Defense Forces’ initial experience. Injury. 2014;45(1):66–70.
    1. Vu EN, Schlamp RS, Wand RT, et al. Prehospital use of transexamic acid for hemorrhagic shock in primary and secondary air medical evacuation. Air Med J. 2013;32(5):289–92.
    1. Neeki MM, MacNeil C, Toy J, et al. Accuracy of perceived estimated travel time by EMS to a trauma center in San Bernardino County, California. West J Emerg Med. 2016;17(4):418–26.
    1. Raza I, Davenport R, Rourke C, et al. The incidence and magnitude of fibrinolytic activation in trauma patients. J Thromb Haemost. 2013;11(2):307–14.
    1. Cap AP, Baer DG, Orman JA, et al. Tranexamic acid for trauma patients: a critical review of the literature. J Trauma. 2011;71(1 Suppl):S9–14.
    1. Lawson JH, Murphy MP. Challenges for providing effective hemostasis in surgery and trauma. Semin Hematol. 2004;41(1 Suppl 1):55–64.
    1. Syrovets T, Lunov O, Simmet T. Plasmin as a proinflammatory cell activator. J Leukoc Biol. 2012;92(3):509–19.
    1. Jimenez JJ, Iribarren JL, Lorente L, et al. Tranexamic acid attenuates inflammatory response in cardiopulmonary bypass surgery through blockade of fibrinolysis: a case control study followed by a randomized double-blind controlled trial. Crit Care. 2007;11(6):R117.
    1. Toner RW, Pizzi L, Leas B, et al. Costs to hospitals of acquiring and processing blood in the US: a survey of hospital-based blood banks and transfusion services. Appl Health Econ Health Policy. 2011;9(1):29–37.
    1. Dowd NP, Karski JM, Cheng DC, et al. Pharmacokinetics of tranexamic acid during cardiopulmonary bypass. Anesthesiology. 2002;97(2):390–9.
    1. Sigaut S, Tremey B, Ouattara A, et al. Comparison of two doses of tranexamic acid in adults undergoing cardiac surgery with cardiopulmonary bypass. Anesthesiology. 2014;120(3):590–600.
    1. Ekback G, Axelsson K, Ryttberg L, et al. Tranexamic acid reduces blood loss in total hip replacement surgery. Anesth Analg. 2000;91(5):1124–30.
    1. Henry DA, Carless PA, Moxey AJ, et al. Anti-fibrinolytic use for minimising perioperative allogeneic blood transfusion. Cochrane Database Syst Rev. 2011;1886;16(3):CD00.
    1. Fiechtner BK, Nuttall GA, Johnson ME, et al. Plasma tranexamic acid concentrations during cardiopulmonary bypass. Anesth Analog. 2001;92(5):1131–6.
    1. Horrow JC, Van Riper DF, Strong MD, et al. The dose-response relationship of tranexamic acid. Anesthesiology. 1995;82(2):383–92.
    1. Pilbrant A, Schannong M, Vessman J. Pharmacokinetics and bioavailability of tranexamic acid. Euro J Clin Pharmacol. 1981;20(1):65–72.
    1. O Eriksson H, Kjellman H, Pilbrant A. Pharmacokinetics of tranexamic acid after intravenous administration to normal volunteers. Euro J Clin Pharmacol. 1974;7(5):375–80.
    1. Benoni G, Björkman S, Fredin H. Application of pharmacokinetic data from healthy volunteers for the prediction of plasma concentrations of tranexamic Acid in surgical patients. Clin Drug Invest. 1995;10(5):280–7.

Source: PubMed

3
Abonnieren