• No results found

Optimising diagnosis and treatment of coagulopathy in severely injured trauma patients - General introduction and outline of the thesis

N/A
N/A
Protected

Academic year: 2021

Share "Optimising diagnosis and treatment of coagulopathy in severely injured trauma patients - General introduction and outline of the thesis"

Copied!
14
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Optimising diagnosis and treatment of coagulopathy in severely injured trauma

patients

Balvers, K.

Publication date

2016

Document Version

Final published version

Link to publication

Citation for published version (APA):

Balvers, K. (2016). Optimising diagnosis and treatment of coagulopathy in severely injured

trauma patients.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)
(3)

GENERAL INTRODUCTION

Trauma has a profound impact on public health around the world. Yearly approximately 5 million people die due to traumatic injury, which is 1 out of every 3 severely injured

patients1. Therefore, improving survival after trauma is a major challenge in which

timely therapy is of great importance. Although increased knowledge about the mechanisms and pathophysiology of traumatic injury to the human body have led to improved trauma care, surgical procedures, and critical care management over the last decades, still a large proportion of patients die after trauma. Better understanding of how the injury and treatment affect the outcome after trauma may result in a decreased mortality. However, in this field there is still a lot of work to do.

In trauma patients, massive haemorrhage is one of the leading causes of mortality.

Exsanguination accounts for more than 30% of mortality in trauma patients2. The main

part of the treatment of massive haemorrhage is to stop the bleeding. However, the development of trauma-induced coagulopathy (TIC) hampers this and exacerbates the bleeding. Therefore, treatment of coagulopathy is a cornerstone in achieving haemostasis and in therapy of bleeding trauma patients.

COAGULOPATHY

Coagulopathy is a condition of the blood in which the blood`s ability to coagulate is impaired. However, the term coagulopathy can relate to several divers conditions. Intensivists associate coagulopathy with disseminated intravascular coagulopathy (DIC), which is characterized by an increased tendency of clotting of the blood, also known as hypercoagulopathy, which is thought to contribute to organ failure and late mortality. Trauma surgeons interpret coagulopathy as a diminished clotting function, also known as hypocoagulopathy, which is associated with early mortality. Additionally, several terms in literature are used to refer to the same condition. Terms such as acute traumatic coagulopathy (ATC), early coagulopathy of trauma (ECT), trauma-induced coagulopathy (TIC), and the acute coagulopathy of trauma-shock (ACoTS) are commonly used. Both the various interpretations and terms used for coagulopathy, illustrate the lack of knowledge on the dynamics of the coagulation process in trauma. In this thesis we will further discuss coagulopathy after trauma. The term in this thesis used for coagulopathy is trauma-induced coagulopathy (TIC) and refers to a diminished clotting function, also knowns as a hypocoagulable state, upon arrival at the Emergency Department.

(4)

TRAUMA-INDUCED COAGULOPATHY

Almost 25% of the severely injured trauma patients have developed hypocoagulopathy

on arrival to the Emergency Department3, 4. Compared to trauma patients without

coagulopathy, patients with TIC have a fourfold higher risk for mortality. Early mortality is determined by a hypocoagulable state and bleeding to death, whereas late mortality is determined by a hypercoagulable state and the development of multiple organ

failure2, 5.

The hypocoagulable state increases the risk for bleeding and exacerbates blood loss. This early mortality by haemorrhage is one of the leading causes of death in

trauma patients, but it is also the most preventable cause of death6-8. Treatment of

coagulopathy is a cornerstone in achieving haemostasis and in therapy of bleeding trauma patients, as controlling the bleeding by a surgical procedure is not possible without a good functioning clotting system. However, overtreatment of TIC may result in a hypercoagulable state, which is associated with the development of multiple organ failure and late mortality. Therefore, to treat TIC adequately, knowledge about the pathophysiology and dynamics of coagulopathy in the course of severe trauma is required.

PATHOGENESIS OF TRAUMA-INDUCED COAGULOPATHY

Conventional theory holds that early TIC was caused by hypothermia, acidosis and dilution, also known as the lethal triad. Hypothermia and acidosis result in the dysfunction of clotting enzymes, whereas administration of resuscitation fluids dilutes

the concentration of clotting factors in blood9, 10. TIC results in an increased blood

loss with exacerbation of hypovolemic shock and concomitant decreased perfusion of

organs, which leads to hypothermia, acidosis and subsequently death2-4, 11.

However, nowadays it is suggested that early development of TIC is caused by external factors, like hypothermia, dilution and acidosis, in combination with a response of the body to tissue injury. After tissue injury, endothelial cell activation results in the initiation of the pro-inflammatory response system and the triggering of thrombo-thrombomodulin complexes. These complexes activate protein C, also known as the protein C pathway. Activated protein C inhibits clotting factors V and VIII thereby reducing the clotting function. However, in trauma, the presence of shock and sustained hypoperfusion, causes an increased release of thrombo-thrombomodulin complexes, which results in a widespread protein C activation and an impaired clot formation. Additionally, besides the fact that clotting factor V and VIII are inhibited by activated protein C,

(5)

protein C depletes plasminogen inhibitors, including (PAI-1). Normally, plasminogen inhibitors have the function to inhibit the formation of plasminogen in to plasmin. The breakdown of the fibrin network and subsequently the clot is thereby prevented. However, depletion of plasminogen inhibitors by protein C results in an increased clot breakdown, also known as hyperfibrinolysis. This hypocoagulable effect is further enhanced by the release of small molecules of heparin-substances after endothelial damage. Together this aggravates a hypocoagulable state, blood loss and subsequently

increases haemorrhagic-related deaths12-16. Figure 1 illustrates the pathogenesis of

coagulopathy after trauma.

At the same time, consumption of activated protein C occurs after trauma. This depletion of protein C is a potential mechanism for the development of hypercoagulopathy. Due to depletion of protein C, clotting factor V and VIII and plasminogen inhibitors are no longer inhibited, which may result in a hypercoagulable state. It is suggested that this hypercoagulable state is associated with the formation of micro-thrombi, also known

as DIC, and the formation of multiple organ failure after trauma17-20. Also, depletion

of protein C leads potentially to an impaired immune response and subsequently a

higher risk of infectious diseases12-16, 21-23. In line with this, previous studies reported

lower levels of protein C levels in patients with sepsis and ventilator-associated

pneumonia in critically ill trauma patients14, 24. This indicates that the immune system

and the coagulation system are interlinked and that activation of the immune system is associated with a pro-coagulant effect in trauma patients. However, which mediators are responsible for this, is unknown. Results of previous studies suggest that a prompt release of microparticles, which are vesicles which are shed into the bloodstream by cells under conditions of stress, are associated with both a coagulant and a

pro-inflammatory immune responses25-29. However, whether this also applies to trauma

patients remains to be determined. Hypo- and hypercoagulopathy after trauma poses a challenge to the trauma team, with the need for awareness and timely treatment of the lethal triad while avoiding unnecessary transfusion. The diagnosis and treatment of TIC is a cornerstone in this process.

DIAGNOSIS OF TIC

Early detection and identification of trauma patients with coagulopathy is required to optimise therapy. Activated partial thromboplastin time (aPTT), prothrombin time (PT), the international normalized ratio (INR), platelet count, fibrinogen and d-dimer are conventional clotting tests, which are used frequently in the clinical setting. However, the use of these tests is rather based on tradition than on evidence based medicine supporting the use of these tests in trauma setting. Conventional clotting tests are

(6)

FIGURE 1:

(7)

very time-consuming as results become available after at least 40-60 minutes. Also, these tests reflect only a part of the clotting profile. Thereby, these tests have minimal

impact on transfusion practice in bleeding trauma patients30-33. Although these tests are

commonly used to evaluate and to predict bleeding, these tests are originally designed to diagnose coagulation disorders and to evaluate anticoagulant medication. Therefore, transfusion practice is currently more an empiric procedure than based upon adequate clotting tests. This is alarming, as conventional clotting tests do not allow for correct diagnosis of TIC and hence no targeted therapy is possible. In conclusion, no adequate diagnostic and monitoring tools for coagulopathy in trauma patients are available nowadays.

Viscoelatic Heamostatic Assays (VHA), like thromboelastometry (ROTEM®) and

thromboelastography (TEG®), are rapid tests which reflect the whole coagulation

status. Within 5-10 minutes a first impression of the clotting function is visualized. VHA tests provide an impression for global haemostasis, including the measurement of the total coagulation process from clot formation until clot breakdown. Therefore the use of these VHA assays may be a valuable alternative for diagnosing and monitoring

of the effectivity of treatment of TIC in bleeding trauma patients33-38. However, clear

reference values for coagulopathy in trauma patients still need to be determined, as the manufacturer has provided only general reference values. Furthermore, it is unknown what the monitoring capacity of these VHA assays is and whether implementation of these tests results in optimization of transfusion by avoiding transfusion unnecessary blood products and pro-coagulants. Additionally, it remains to be determined whether VHA assays can be used to provide targeted transfusion in trauma patients and what the triggers and targets are for transfusion. Currently, the suspicion of bleeding, hypovolemia and the haemoglobin level are frequently used as triggers for transfusion, however it is unknown whether these factors predict bleeding adequately and are able to use as a transfusion target. Therefore, adequate and rapid diagnostic tools for coagulopathy are required to optimise and monitor treatment of coagulopathic trauma patients.

TREATMENT OF TIC

Over the last decades, research efforts in the field of transfusion practice in trauma patients have been directed towards treatment of the principle drivers of the lethal triad, including hypothermia, acidosis and coagulopathy. Therefore, supportive care in trauma consists of prevention of hypothermia and the administration of fewer fluids. From this point of view, prevention of hypothermia and a restricted fluid policy have become part of standard trauma care. Additionally, transfusion practice has evolved from

(8)

A

B

FIGURE 2: VHAs like ROTEM® (A) and TEG® (B)39, 40

administration of red blood cells (RBCs) towards earlier administration of fresh frozen

plasma (FFPs) and platelets (PLTs) to red blood cells (RBCs)41-44. A RBC:FFP:platelet ratio

of 1:1:1 is suggested to be the closest approximation of whole blood and contributes to

the achievement of haemostasis thereby decreasing mortality33, 41, 45, 46, 47-51.

In order to obtain a balanced ratio of blood products, massive transfusion protocols (MTPs) are increasingly being used in trauma care. MTPs attempt to provide rapid and standardized issuing of blood products in a 1:1:1 ratio and aim to reduce

time-to transfusion by keeping pre-thawed plasma available47-50. However, evidence for a

beneficial effect of implementation of the MTP on obtaining a balanced transfusion ratio, coagulation profiles and overall survival is still lacking. Furthermore, it is unclear whether implementation of an MTP results in an increased incidence of overtransfusion of blood products. Overtransfusion is still a frequently observed phenomenon and is

associated with adverse events like sepsis and multiple organ failure21-23.

Alternative transfusion strategies with early balanced resuscitation in order to control TIC and to decrease traumatic bleeding while avoiding unnecessary transfusion are therefore required. An alternative to empiric use of ratios may be transfusion practice

(9)

guided by VHA assays, as these tests are rapid and reflect the coagulation status adequately. Furthermore, these tests have shown promising results in their ability to

detect and to monitor coagulopathy34, 37, 38. As results become available within 5-10

minutes after initiating the VHA assays, these tests may be used to guide transfusion of blood products, pro-coagulant and antifibrinolytic agents. Subsequently, VHA assays could be incorporated in a transfusion algorithm, which supports rapid clinical management of severely injured trauma patients. However, the additional value of the use of VHA assays in trauma resuscitation has to be determined.

AIM AND OUTLINE OF THE THESIS

This thesis focusses on knowledge gaps in the field of diagnosis and treatment of TIC in severely injured trauma patients. In order to explore potential diagnostic tools for TIC and to investigate potential strategies to optimise treatment of TIC, the Academic Medical Center of Amsterdam has been participating in the International Trauma Research Network (INTRN) since 2012. The INTRN is a consortium of 6 European Level-1 trauma centres, which received funding from the European Union Framework Programme 7 (FP7) to perform research in the field of coagulopathy after trauma. This thesis is partly established by collaboration with INTRN and by using a large database of trauma patients. The aim of this thesis is to evaluate diagnostic tools for TIC and to investigate which transfusion strategy is associated with the best outcome after trauma. The first part of this thesis focusses on optimising diagnosis of TIC, whereas the second part of this thesis focusses on optimising treatment of TIC.

PART 1 DIAGNOSIS OF COAGULOPATHY

• Chapter 1 provides a narrative review of the utility of ROTEM® and TEG® to detect

coagulopathy in critically ill non-bleeding patients.

• Chapter 2 assesses the predictive value of hypercoagulopathy detected by ROTEM®

for the development of multiple organ failure.

• Chapter 3 determines the association between the haemoglobin level and the neurologic outcome of patients after traumatic brain injury.

• Chapter 4 investigates the role of microparticles in mediating the immune response following trauma.

TREATMENT OF COAGULOPATHY

• Chapter 5 gives a systematic overview of the risk factors related to coagulopathy and transfusion practice for adverse outcome after major trauma.

(10)

• Chapter 6 emphasizes the detrimental effect of accidental hypothermia on mortality in coagulopathic trauma patients at admittance to the intensive care unit. • Chapter 7 studies the effect of the introduction of an MTP on the use of blood

products and transfusion ratios.

• Chapter 8 systematically determines alternatives for the transfusion of AB-plasma in massively bleeding patients

• Chapter 9 investigates which transfusion strategy is associated with best outcome in bleeding trauma patients

• Chapter 10 determines the response of ROTEM® to transfusion practice in bleeding

(11)

REFERENCES 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Haagsma JA, Graetz N, Bolliger I, Naghavi M, Higashi H, Mullany EC, et al. The global burden of injury: incidence, mortality, disability-adjusted life years and time trends from the Global Burden of Disease study 2013. Injury prevention : journal of the International Society for Child and Adolescent Injury Prevention. 2016;22(1):3-18.

Sauaia A, Moore FA, Moore EE, Moser KS, Brennan R, Read RA, et al. Epidemiology of trauma deaths: a reassessment. J Trauma. 1995;38(2):185-93.

Brohi K, Singh J, Heron M, Coats T. Acute traumatic coagulopathy. J Trauma. 2003;54(6):1127-30.

MacLeod JB, Lynn M, McKenney MG, Cohn SM, Murtha M. Early coagulopathy predicts mortality in trauma. J Trauma. 2003;55(1):39-44.

Krug EG, Sharma GK, Lozano R. The global burden of injuries. Am J Public Health. 2000;90(4):523-6.

Bellamy RF. The causes of death in conventional land warfare: implications for combat casualty care research. Mil Med 1984;149(2):55-62. Holcomb JB, McMullin NR, Pearse L, Caruso J, Wade CE, Oetjen-Gerdes L, et al. Causes of death in U.S. Special Operations Forces in the global war on terrorism: 2001-2004. Ann Surg 2007;245(6):986-91.

Esposito TJ, Sanddal ND, Hansen JD, Reynolds S. Analysis of preventable trauma deaths and inappropriate trauma care in a rural state. J Trauma. 1995;39(5):955-62.

Maegele M, Lefering R, Yucel N, Tjardes T, Rixen D, Paffrath T, et al. Early coagulopathy in multiple injury: an analysis from the German Trauma Registry on 8724 patients. Injury. 2007;38(3):298-304.

Wafaisade A, Wutzler S, Lefering R, Tjardes T, Banerjee M, Paffrath T, et al. Drivers of acute coagulopathy after severe trauma: a multivariate analysis of 1987 patients. Emerg Med J 2010;27(12):934-9.

Cosgriff N, Moore EE, Sauaia A, Kenny-Moynihan M, Burch JM, Galloway B. Predicting life-threatening coagulopathy in the massively transfused trauma patient: hypothermia and acidoses revisited. J Trauma. 1997;42(5):857-61.

Maegele M, Schochl H, Cohen MJ. An update on the coagulopathy of trauma. Shock. 2014;41 Suppl 1:21-5.

Brohi K, Cohen MJ, Ganter MT, Matthay MA, Mackersie RC, Pittet JF. Acute traumatic coagulopathy: initiated by hypoperfusion: modulated through the protein C pathway? Ann Surg 2007;245(5):812-8.

Cohen MJ, Call M, Nelson M, Calfee CS, Esmon CT, Brohi K, et al. Critical role of activated protein C in early coagulopathy and later organ failure, infection and death in trauma patients. Ann Surg 2012;255(2):379-85.

Kutcher ME, Xu J, Vilardi RF, Ho C, Esmon CT, Cohen MJ. Extracellular histone release in response to traumatic injury: implications for a compensatory role of activated protein C. J Trauma Acute Care Surg 2012;73(6):1389-94. Rehm M, Bruegger D, Christ F, Conzen P, Thiel M, Jacob M, et al. Shedding of the endothelial glycocalyx in patients undergoing major vascular surgery with global and regional ischemia. Circulation. 2007;116(17):1896-906.

Gando S, Nakanishi Y, Tedo I. Cytokines and plasminogen activator inhibitor-1 in posttrauma disseminated intravascular coagulation: relationship to multiple organ dysfunction syndrome. Crit Care Med 1995;23(11):1835-42.

Gando S, Nakanishi Y, Kameue T, Nanzaki S. Soluble thrombomodulin increases in patients with disseminated intravascular coagulation and in those with multiple organ dysfunction syndrome after trauma: role of neutrophil elastase. J Trauma. 1995;39(4):660-4.

(12)

REFERENCES 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 Disseminated intravascular coagulation and sustained systemic inflammatory response syndrome predict organ dysfunctions after trauma: application of clinical decision analysis. Ann Surg 1999;229(1):121-7. Paffrath T, Wafaisade A, Lefering R, Simanski C, Bouillon B, Spanholtz T, et al. Venous thromboembolism after severe trauma: incidence, risk factors and outcome. Injury. 2010;41(1):97-101.

Sauaia A, Moore FA, Moore EE, Haenel JB, Read RA, Lezotte DC. Early predictors of postinjury multiple organ failure. Arch Surg 1994;129(1):39-45.

Dewar DC, Tarrant SM, King KL, Balogh ZJ. Changes in the epidemiology and prediction of multiple-organ failure after injury. J Trauma Acute Care Surg 2013;74(3):774-9.

Frohlich M, Lefering R, Probst C, Paffrath T, Schneider MM, Maegele M, et al. Epidemiology and risk factors of multiple-organ failure after multiple trauma: an analysis of 31,154 patients from the TraumaRegister DGU. J Trauma Acute Care Surg 2014;76(4):921-7. Choi Q, Hong KH, Kim JE, Kim HK. Changes in plasma levels of natural anticoagulants in disseminated intravascular coagulation: high prognostic value of antithrombin and protein C in patients with underlying sepsis or severe infection. Annals of laboratory medicine. 2014;34(2):85-91.

Fujimi S, Ogura H, Tanaka H, Koh T, Hosotsubo H, Nakamori Y, et al. Increased production of leukocyte microparticles with enhanced expression of adhesion molecules from activated polymorphonuclear leukocytes in severely injured patients. J Trauma. 2003;54(1):114-9.

Mastronardi ML, Mostefai HA, Meziani F, Martinez MC, Asfar P, Andriantsitohaina R. Circulating microparticles from septic shock patients exert differential tissue expression of enzymes related to inflammation and oxidative stress. Crit Care Med 2011;39(7):1739-48.

Morel O, Toti F, Hugel B, Bakouboula B, Camoin-Jau L, Dignat-George F, et al. Procoagulant microparticles: disrupting the vascular homeostasis equation? Arterioscler Thromb Vasc Biol 2006;26(12):2594-604. Ogura H, Kawasaki T, Tanaka H, Koh T, Tanaka R, Ozeki Y, et al. Activated platelets enhance microparticle formation and platelet-leukocyte interaction in severe trauma and sepsis. J Trauma. 2001;50(5):801-9.

Ogura H, Tanaka H, Koh T, Fujita K, Fujimi S, Nakamori Y, et al. Enhanced production of endothelial microparticles with increased binding to leukocytes in patients with severe systemic inflammatory response syndrome. J Trauma. 2004;56(4):823-30.

Holcomb JB, Minei KM, Scerbo ML, Radwan ZA, Wade CE, Kozar RA, et al. Admission rapid thrombelastography can replace conventional coagulation tests in the emergency department: experience with 1974 consecutive trauma patients. Annals of surgery. 2012;256(3):476-86.

Dzik WH. Predicting hemorrhage using preoperative coagulation screening assays. Current hematology reports. 2004;3(5):324-30.

Park MS, Martini WZ, Dubick MA, Salinas J, Butenas S, Kheirabadi BS, et al. Thromboelastography as a better indicator of hypercoagulable state after injury than prothrombin time or activated partial thromboplastin time. J Trauma. 2009;67(2):266-75; discussion 75-6.

Davenport R, Manson J, De’Ath H, Platton S, Coates A, Allard S, et al. Functional definition and characterization of acute traumatic coagulopathy. Crit Care Med 2011;39(12):2652-8.

Johansson PI, Stensballe J. Effect of Haemostatic Control Resuscitation on mortality in massively bleeding patients: a before and after study. Vox Sang 2009;96(2):111-8.

(13)

REFERENCES 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 S, Doran C, Kirkman E. Utility of interim ROTEM((R)) values of clot strength, A5 and A10, in predicting final assessment of coagulation status in severely injured battle patients. Injury. 2013;44(5):593-9.

Rugeri L, Levrat A, David JS, Delecroix E, Floccard B, Gros A, et al. Diagnosis of early coagulation abnormalities in trauma patients by rotation thrombelastography. Journal of thrombosis and haemostasis : JTH. 2007;5(2):289-95.

Schochl H, Nienaber U, Hofer G, Voelckel W, Jambor C, Scharbert G, et al. Goal-directed coagulation management of major trauma patients using thromboelastometry (ROTEM)-guided administration of fibrinogen concentrate and prothrombin complex concentrate. Crit Care. 2010;14(2):R55. Schochl H, Nienaber U, Maegele M, Hochleitner G, Primavesi F, Steitz B, et al. Transfusion in trauma: thromboelastometry-guided coagulation factor concentrate-based therapy versus standard fresh frozen plasma-based therapy. Crit Care. 2011;15(2):R83. GmbH TI. Available from: https://www.rotem. de.

Heamonetics. Available from: http://teg. haemonetics.com/.

Holcomb JB, del Junco DJ, Fox EE, Wade CE, Cohen MJ, Schreiber MA, et al. The prospective, observational, multicenter, major trauma transfusion (PROMMTT) study: comparative effectiveness of a time-varying treatment with competing risks. JAMA Surg 2013;148(2):127-36.

Kutcher ME, Kornblith LZ, Narayan R, Curd V, Daley AT, Redick BJ, et al. A paradigm shift in trauma resuscitation: evaluation of evolving massive transfusion practices. JAMA Surg 2013;148(9):834-40.

Wafaisade A, Maegele M, Lefering R, Braun M, Peiniger S, Neugebauer E, et al. High plasma to red blood cell ratios are associated with lower mortality rates in patients receiving

multiple transfusion (4</=red blood cell units<10) during acute trauma resuscitation. J Trauma. 2011;70(1):81-8.

Zink KA, Sambasivan CN, Holcomb JB, Chisholm G, Schreiber MA. A high ratio of plasma and platelets to packed red blood cells in the first 6 hours of massive transfusion improves outcomes in a large multicenter study. Am J Surg 2009;197(5):565-70. Holcomb JB, Jenkins D, Rhee P, Johannigman J, Mahoney P, Mehta S, et al. Damage control resuscitation: directly addressing the early coagulopathy of trauma. J Trauma. 2007;62(2):307-10.

Holcomb JB, Tilley BC, Baraniuk S, Fox EE, Wade CE, Podbielski JM, et al. Transfusion of plasma, platelets, and red blood cells in a 1:1:1 vs a 1:1:2 ratio and mortality in patients with severe trauma: the PROPPR randomized clinical trial. JAMA. 2015;313(5):471-82. Ball CG, Dente CJ, Shaz B, Wyrzykowski AD, Nicholas JM, Kirkpatrick AW, et al. The impact of a massive transfusion protocol (1:1:1) on major hepatic injuries: does it increase abdominal wall closure rates? Can J Surg 2013;56(5):E128-E34.

Khan S, Allard S, Weaver A, Barber C, Davenport R, Brohi K. A major haemorrhage protocol improves the delivery of blood component therapy and reduces waste in trauma massive transfusion. Injury. 2013;44(5):587-92.

McDaniel LM, Neal MD, Sperry JL, Alarcon LH, Forsythe RM, Triulzi D, et al. Use of a massive transfusion protocol in nontrauma patients: activate away. J Am Coll Surg 2013;216(6):1103-9.

O’Keeffe T, Refaai M, Tchorz K, Forestner JE, Sarode R. A massive transfusion protocol to decrease blood component use and costs. Arch Surg 2008;143(7):686-90.

Schuster KM, Davis KA, Lui FY, Maerz LL, Kaplan LJ. The status of massive transfusion protocols in United States trauma centers:

(14)

REFERENCES

massive transfusion or massive confusion? Transfusion. 2010;50(7):1545-51.

Referenties

GERELATEERDE DOCUMENTEN

The B3LYP and PBE0 calculations of the absorption energies of the sulfide adduct m-PSP-SH, however, indicate that there are at least two charge-transfer states at lower

We examine two hypotheses implied by the theory: one, for any three players with two links present, the probability of a third link being present is increasing in the strength of

Rhodium Catalyzed Hydroformylation of Higher Alkenes using Amphiphilic Ligands.. Buhling, A.; Kamer, P.C.J.; van

Hence, an original owner is protected increasingly more in France, Germany, and the United States, and a natural metrics of her/his property rights on a movable good is the number

Thiss thesis focuses on the factors that are involved in the import of peroxisomall proteins in the yeast Saccharomyces cerevisiae (baker's yeast). The peroxisomall protein

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons.. In case of

Chapterr 2 Saccharomyces cerevisiae PTS1 receptor Pex5p interacts with 53 thee SH3 domain of the peroxisomal membrane protein. Pexl3pp in an unconventional, non-PXXP-related manner