Skip to main content

Efficacy of antithrombin in preclinical and clinical applications for sepsis-associated disseminated intravascular coagulation

Abstract

Antithrombin (AT) is known as an important physiological anticoagulant. AT inactivates thrombin and multiple other coagulation factors, thereby strongly inhibiting the over-activation of the coagulation system during disseminated vascular coagulation (DIC). AT also suppresses the pro-inflammatory reactions that are promoted through protease-activated receptor-1 during sepsis. One of the unique characteristics of AT is the conformational change it undergoes when binding to heparin-like molecules. The anticoagulant function is greatly accelerated after AT binds to externally administered heparin in the circulating blood. Meanwhile, AT also binds to syndecan-4 on the cell surface under physiological conditions, thereby contributing to local antithrombogenicity. The binding of AT and syndecan-4 upregulates prostaglandin I2 production, downregulates pro-inflammatory cytokine production, and suppresses the leukocyte-endothelial interaction. Other than these activities, recent preclinical studies have reported that AT might inhibit neutrophil necrotic cell death and the ejection of neutrophil extracellular traps. Together, these effects may lead to the attenuation of inflammation by decreasing the level of damage-associated molecular patterns. Although a number of animal studies have demonstrated a survival benefit of AT, the clinical benefit has long been argued since the effect of high-dose AT was denied in 2001 in a large-scale randomized controlled trial targeting patients with severe sepsis. However, recent clinical studies examining the effects of a supplemental dose of AT in patients with sepsis-associated DIC have revealed that AT is potentially effective for DIC resolution and survival improvement without increasing the risk of bleeding. Since DIC is still a major threat during sepsis, the optimal method of identifying this promising drug needs to be identified.

Introduction

As was written in a recent review by Hunt [1], the fundamental strategy for caring for patients with sepsis-associated disseminated intravascular coagulation (DIC) is the management of underlying infection. During septic DIC, thrombus formation is driven by activated coagulation, the impairment of anticoagulant mechanisms including the antithrombin (AT) system, and compromised fibrin removal arising from the depression of the fibrinolytic system [2]. Microvascular thrombosis contributes to diminished oxygen delivery and subsequent organ dysfunction. Accordingly, anticoagulant therapy is expected to play some role in alleviating this dangerous condition [3]. In the early 2000s, some large-scale randomized controlled trials (RCTs) targeting severe sepsis were conducted [4]-[6], but none of the anticoagulants that were examined are currently available for clinical use. Actually, sepsis-associated DIC, instead of severe sepsis, might be an appropriate target for these anticoagulant therapies. Indeed, some subgroup analyses of subjects with sepsis-associated DIC in the above-mentioned RCTs have revealed effects on mortality [7],[8]. However, the effects of these anticoagulants on septic DIC have not been examined in the well-qualified studies. Recently, a small-sized but properly designed RCT succeeded in demonstrating the efficacy of a physiological dose of AT for DIC resolution [9]. Following this report, an analysis using a nationwide administrative database in Japan revealed a positive effect of physiological AT use on mortality. Under these circumstances, the “Harmonized guidance for DIC” has been released by the International Society on Thrombosis and Haemostasis (ISTH) [10]. In this guidance, AT is graded as “potentially recommended”. Therefore, our present and future tasks will be to search for appropriate measures for AT use and to accumulate sufficient evidence.

Review

Preclinical evaluation

AT is a vitamin K-independent glycoprotein with a molecular weight of approximately 59 kDa and is one of the major natural anticoagulants that have been aggressively studied [8],[11],[12]. AT inhibits thrombin in a 1:1 fashion and leads to the formation of a thrombin-antithrombin complex (TAT), thereby inactivating the enzymatic activity of thrombin and leading to its elimination from the circulation. Therefore, the inactivation of thrombin is considered to be a rational therapeutic strategy for DIC. Apart from anticoagulation, the anti-inflammatory function of AT can also be explained by the neutralization of thrombin. Thrombin has been implicated in the inflammatory cascade [13]; specifically, it increases leukocyte rolling and adhesion [14] by increasing the expression of endothelial P- and E-selectin [15],[16] and intercellular adhesion molecule-1 (ICAM-1) [16], thereby promoting leukocyte recruitment. Thrombin also elicits an inflammatory reaction through its receptor, protease activated receptor (PAR)-1, on the cell surface [17]. PAR-1 is known to play pivotal roles in the activation of inflammation [18] by inducing the production of pro-inflammatory cytokines and chemokines by the endothelium [16],[19]. Thus, the anti-inflammatory function of AT is thought to depend, at least in part, on the blockage of the effects of PAR-1.

Another cell-modulating activity of AT is induced after binding with its specific receptor syndecan-4, a type of heparin-like glycosaminoglycan (GAG), on various types of cell surfaces [20]. One major property of AT is thought to be its ability to stimulate prostacyclin production by endothelial cells through the binding of AT and syndecan-4 [21]. Prostacyclin exerts an anti-inflammatory function by blocking neutrophil tethering on the vascular endothelium [22] and by downregulating the production of pro-inflammatory cytokines [23]. When AT and syndecan-4 bind together on neutrophils, monocytes, and lymphocytes, their interactions with the endothelium are suppressed. The downregulation of P-selectin may also be involved in the vessel wall integrity-preserving properties of AT [24].

The balance between the anticoagulation effect and the anti-inflammatory cellular function is dominantly regulated by heparins. AT contains a heparin-binding domain, and its anticoagulation activity is maximized by several orders of magnitude after binding with heparin in the bloodstream (Figure 1). This binding of heparin to the lysyl side chains of AT induces a nonreversible conformational change leading to a high affinity towards thrombin. Since the latter effect is more desired in sepsis, the coadministration of AT and heparin might not be a good choice for the treatment of patients with sepsis and not associated with DIC [7].

Figure 1
figure 1

Multifactorial functions of antithrombin in circulating blood and on the cell surface. The interactions between antithrombin (AT) and the endothelium are shown in the figure. The affinity of antithrombin to thrombin and its enzymatic inhibition are increased by the binding of the heparin-binding site of AT to syndecan-4 on the cell surface or externally administered heparins. Thrombin loses its coagulant activity after the formation of a thrombin-antithrombin complex. Other than thrombin, AT inactivates factors Xa and IXa. As for its anti-inflammatory function, AT inactivates thrombin, thereby attenuating the cellular reactions through the activation of protease-activated receptor (PAR)-1.

Recently, the mechanisms involved in the pathological derangement of coagulation in patients with sepsis have become increasingly clear. Tissue factor (TF) is a key factor in thrombin generation during sepsis [25]. However, recent studies have elucidated that damage-associated molecular patterns (DAMPs) from necrotic cells and neutrophil extracellular traps (NETs) and their components also play major roles in the activation of the coagulation system [26],[27]. NETs activate coagulation by expressing tissue factor [28] and stimulate platelets via histones [29], which are also strong promoters of the extrinsic pathway [30]. Interestingly, recent studies have reported that antithrombin suppresses necrotic cell death and NET formation in the animal and in vitro studies [31],[32].

In animal models, the effect of AT substitution on survival was first reported by Triantaphyllopoulos [33] in a lipopolysaccharide (LPS)-induced rabbit model of sepsis. Similar effects were reported in an Escherichia coli-induced primate model of sepsis by Taylor et al. [34]. Other than these reports, the protective effect of AT on mortality has been reported in various models of sepsis [35],[36]. In our rat model, all the rats died when treated with a continuous infusion of 10.0 mg/kg of LPS over 24 h, but 50% of the rats survived when treated with low-dose AT and all the rats survived when treated with high-dose AT [37]. These results were adopted as the fundamental basis of the clinical studies.

Clinical evaluation

A pharmacological dose of recombinant activated protein C [4], high-dose AT [5], and tissue factor pathway inhibitor [6] are widely known to have failed to demonstrate a survival benefit among patients with severe sepsis in large-scale RCTs. Some reasons have been suggested to explain the failures of these trials. First, even if the anti-inflammatory effects of anticoagulants have been advocated in animal and ex vivo studies, anticoagulant therapies seem to be effective only in the septic patients with DIC, but not in those without DIC, in a clinical setting. Subgroup analyses performed in subjects with DIC in the KyberSept trial [5] and PROWESS trial [38] revealed improvements in survival [7],[8]. The second reason is that “pharmacological” or “high-dose” anticoagulants may cause bleeding. For example, treatment with high-dose AT was correlated with a significant hemorrhagic tendency. The incidence of total bleeding events was reported to be 12.8% in the control group and 22.0% in the AT-treated group (relative risk [RR], 1.71 [95% confidence interval [CI], 1.42–2.06]). Thus, increased bleeding events might diminish the beneficial effects of AT. Third, the concomitant use of heparin might interfere with the effect of AT. As a matter of fact, nearly 70% of high-dose antithrombin-treated patients received heparin. Hoffmann et al. [39],[40] analyzed the data from KyberSept and reported increased bleeding risks associated with antithrombin plus concomitant heparin, compared with antithrombin alone. Regarding this issue, we will introduce the latest studies examining the above problems in the following section.

While a high dose can be harmful, an insufficient dose will be ineffective. So far, we have conducted two nonrandomized multi-institutional post-marketing surveys to determine the optimal AT dose. In the first survey, a total of 729 sepsis-associated DIC patients with an AT activity of 70% or lower were analyzed. Among these patients, AT was substituted at a dose of either 1,500 IU/day (n = 650) or 3,000 IU/day (n = 79) for three consecutive days. The dose selection was made by the attending physicians based primarily on each patient's condition, and the baseline AT activity level was lower among the patients who received 3,000 IU/day. As a result, the survival of the patients who had received 1,500 IU/day was 65.2%, while that of the patients who received 3,000 IU/day was 74.7%, and a logistic regression analysis showed that the supplemented AT dose of 3,000 IU/day contributed to a better survival outcome (odds ratio [OR], 1.912; P = 0.026) [41]. Whether this supplemental dose of AT increased the bleeding risk remained unclear because a placebo control was not included in this post-marketing survey. Nevertheless, since the incidence was 6.52% (major bleeding, 1.71%), which was even lower than that of the control group (major bleeding, 5.7%) in the KyberSept trial [5], we believe that AT supplementation for septic DIC patients is an acceptable treatment.

Since the survival difference did not reach statistical significance and the effect of AT was more prominent in severer cases (lower baseline AT activity) in the first survey, we conducted a second survey in septic DIC patients with baseline AT activities of less than 40%. A total of 307 patients (259 patients received 1,500 IU/day and 48 patients received 3,000 IU/day) were investigated in this second survey. The results demonstrated a significantly higher rate of DIC resolution (66.7% vs. 45.2%, P = 0.007) and a better survival outcome (77.1% vs. 56.4%, P = 0.010) among the patients supplemented with 3,000 IU/day of AT. Bleeding events were observed in 6.96% (major bleeding, 3.04%) of the patients supplemented with 1,500 IU/day and 6.52% (major bleeding, 4.35%) of the patients supplemented with 3,000 IU/day; this difference was not significant [42].

From these results, we assumed that AT supplementation at a sufficient dose could be expected to demonstrate a favorable effect. If so, which dose is most likely to be sufficient? The recovered AT activity level may provide a clue as to the most appropriate dose. The mean AT activity level in the patients supplemented with 3,000 IU/day recovered to within the normal range (>80%), while that of patients supplemented with 1,500 IU/day never reached the normal range in either survey in the second survey. In the first survey, the baseline AT activity was approximately 50%, and it rose up to above 80% in the patients supplemented with 1,500 IU/day. So, roughly 1,500 IU/day may be adequate if the initial AT activity level is above 50%; however, if the initial AT activity level is less than 50%, 3,000 IU/day will likely be needed. However, the Japanese health-care system allows the use of 3,000 IU/day of AT only in limited, severe cases. Thus, combination therapy with AT and recombinant thrombomodulin is now attracting attention [43].

Almost simultaneously with the above-mentioned surveys, the Japanese Association for Acute Medicine (JAAM) performed a multicenter placebo-controlled RCT in 60 septic DIC patients with baseline antithrombin levels of 50%–80%. The treatment group (30 cases) received a supplemental dose of AT (30 IU/kg/day × 3 days), and the mean AT activity recovered to 107.6% ± 24.5% in the treatment group, while it remained around 60% in the placebo group. The DIC resolution rate was 53.3% (16/30) in the treatment group, which was more than double that in the control group (20.0%, 6/30), and a significant improvement in DIC resolution was observed [9].

Following these reports, Tagami et al. [44] performed an analysis using a nationwide administrative database in Japan. A total of 9,075 patients with severe pneumonia and DIC were categorized into an antithrombin group (2,663 cases) and a control group (6,412 cases). Propensity score matching created a matched cohort of 2,194 pairs of patients with and without antithrombin treatment. The results demonstrated that standard AT supplementation (1,500–3,000 IU/day × 3 days) was associated with a 9.9% (95% CI, 3.5%–16.3%) reduction in 28-day mortality. Multiple logistic regression analyses showed an association between AT use and the 28-day mortality (adjusted odds ratio, 0.85 [95% CI, 0.75–0.97]). Based on the above-mentioned evidence, a supplemental dose of AT is assumed to be effective in patients with sepsis-associated DIC. However, to confirm this hypothesis, an adequately powered RCT will be required.

One topic in anticoagulant therapy is the development of recombinant AT. Only plasma-derived AT is presently available in Japan, but the development of recombinant AT is currently underway. A phase 3 trial has just been completed, and the results will be published in the near future.

Usefulness as a biomarker

We have introduced the therapeutic characteristics of AT. However, AT can also be used as a biomarker for DIC. A reduced plasma level of AT in patients with DIC is a well-known fact [45]. Furthermore, decreased AT activity is reportedly correlated with the severity and outcome of patients [46]. Therefore, AT activity has become a popular test in patients with coagulopathy in Japan. The mechanisms responsible for the decrease in AT during sepsis are considered to be as follows: the consumption of AT during activated coagulation [47], the decreased synthesis of AT in the liver [47],[48], the degradation of AT by neutrophil elastase [49],[50], and the leakage of AT to the extravascular space. The reduced AT activity level results in a decreased ability to undergo thrombin inactivation, leading to the further acceleration of the coagulation system.

We previously reported that the AT activity level is approximately 80% of normal in septic patients without organ dysfunction, decreasing to approximately 60% in patients with severe sepsis, and 40% in patients with full-blown DIC [51]. Others have reported that the diagnostic value of an area under the receiver operating characteristic curve (AUC) of the AT activity exceeded 0.8 for the prediction of patient outcome [52],[53]. Thus, we think that AT is an excellent discriminator of the severity of sepsis. Recently, Choi et al. [54] reported a significant correlation between AT and the DIC score in patients with sepsis, suggesting that AT is a good indicator of DIC severity. Indeed, AT had a significant prognostic power in a Kaplan-Meier analysis, showing a higher hazard ratio than conventional coagulation markers, such as D-dimer. Similar observations were reported in patients with conditions other than sepsis, such as multiple trauma or major surgery [45],[55].

We conducted a multi-institutional observational study and analyzed samples from 78 sepsis patients with coagulopathy. Data collection was begun within 48 h after the platelet count first decreased to less than 150,000/mm3, and sequential changes in the coagulation markers, including AT activity, protein C activity, fibrin and fibrinogen degradation products (FDP), D-dimer, thrombin-antithrombin complex, plasmin α2-antiplasmin complex, soluble fibrin, and total plasminogen activator inhibitor-1, were evaluated. The results showed that the changes in hemostatic molecular markers were associated with the onset of organ dysfunction beginning at an early stage of sepsis and that AT activity and protein C activity exhibited the highest predictive values among these parameters [56]. Yanagida et al. [57] and Oshiro et al. [58] reported similar results for trauma patients.

Although the measurement of AT activity is valuable for estimating the severity and outcome of patients, it has not been included in major diagnostic criteria since the measurement of AT activity cannot be performed all day in many local laboratories [59]. In fact, all the major scoring systems consist of some of four routine laboratory tests: platelet count, prothrombin time (PT), fibrin-related marker level, and fibrinogen [60],[61]. Although still controversial [62], Egi et al. [63] suggested that the inclusion of AT activity in the diagnostic criteria may provide a better diagnostic performance.

The other unique feature of antithrombin activity is its usefulness as a prognostic indicator after antithrombin supplementation. We evaluated the efficacy of measuring AT activity in 192 septic DIC patients supplemented with AT. A logistic regression analysis indicated that not only the baseline AT activity level but also the ΔAT activity level (the AT value on day 3 − the AT value on day 0) was related to patient outcome. Furthermore, the results revealed that an increase in AT activity had the largest contribution to patient survival (Table 1).

Table 1 Relationship between outcome (28-day survival) and various factors using the stepwise method of logistic regression analysis

In summary, the sensitivity and/or specificity of AT activity for predicting the morbidity and mortality of septic DIC is superior to that of global coagulation tests, and we recommend that AT activity be included in future diagnostic criteria for septic DIC. We also recommend examining sequential changes in this marker when supplementation therapy is performed. The baseline AT value may also help to determine appropriate candidates for anticoagulant therapy [41].

Conclusions

AT, an important natural anticoagulant, inhibits over-activated coagulation and inflammation during sepsis through multifactorial pathways. However, its activity decreases significantly in sepsis-associated DIC. Although high-dose antithrombin administration has failed to provide a survival benefit, a supplementation dose that restores AT activity to within a normal range is expected to be useful. Currently, the “global guidance for the diagnosis and treatment of DIC” rates AT substitution as “potentially recommended,” meaning that AT can be used but that “further clinical study is required to prove its efficacy”.

Abbreviations

APC:

activated protein C

AT:

antithrombin

AUC:

area under the receiver operating characteristic curve

DAMPs:

damage-associated molecular patterns

DIC:

disseminated vascular coagulation

FDP:

fibrin/fibrinogen degradation products

GAGs:

glycosaminoglycans

ICAM-1:

intercellular adhesion molecule-1

ISTH:

International Society on Thrombosis and Haemostasis

JAAM:

Japanese Association for Acute Medicine

LPS:

lipopolysaccharide

NETs:

neutrophil extracellular traps

PAR:

protease-activated receptor

PT:

prothrombin time

RCTs:

randomized controlled trials

RR:

relative risk

TAT:

thrombin-antithrombin complex

TF:

tissue factor

References

  1. Hunt BJ: Bleeding and coagulopathies in critical care. N Engl J Med 2014, 370: 847-859. 10.1056/NEJMra1208626

    Article  CAS  PubMed  Google Scholar 

  2. Levi M, van der Poll T: Inflammation and coagulation. Crit Care Med 2010,38(Suppl):S26-S34. 10.1097/CCM.0b013e3181c98d21

    Article  CAS  PubMed  Google Scholar 

  3. Iba T, Gando S, Thachil J: Anticoagulant therapy for sepsis-associated disseminated intravascular coagulation: the view from Japan. J Thromb Haemost 2014, 12: 1010-1019. 10.1111/jth.12596

    Article  CAS  PubMed  Google Scholar 

  4. Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, Gårdlund B, Marshall JC, Rhodes A, Artigas A, Payen D, Tenhunen J, Al-Khalidi HR, Thompson V, Janes J, Macias WL, Vangerow B, Williams MD: Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med 2012, 366: 2055-2064. 10.1056/NEJMoa1202290

    Article  CAS  PubMed  Google Scholar 

  5. Warren BL, Eid A, Singer P, Pillay SS, Carl P, Novak I, Chalupa P, Atherstone A, Pénzes I, Kübler A, Knaub S, Keinecke HO, Heinrichs H, Schindel F, Juers M, Bone RC, Opal SM: Caring for the critically ill patient. High-dose antithrombin III in severe sepsis: a randomized controlled trial. JAMA 2001, 286: 1869-1878. 10.1001/jama.286.15.1869

    Article  CAS  PubMed  Google Scholar 

  6. Abraham E, Reinhart K, Opal S, Demeyer I, Doig C, Rodriguez AL, Beale R, Svoboda P, Laterre PF, Simon S, Light B, Spapen H, Stone J, Seibert A, Peckelsen C, De Deyne C, Postier R, Pettilä V, Artigas A, Percell SR, Shu V, Zwingelstein C, Tobias J, Poole L, Stolzenbach JC, Creasey AA: Efficacy and safety of tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis: a randomized controlled trial. J Am Med Assoc 2003, 290: 238-247. 10.1001/jama.290.2.238

    Article  CAS  Google Scholar 

  7. Dhainaut JF, Yan SB, Joyce DE, Pettilä V, Basson B, Brandt JT, Sundin DP, Levi M: Treatment effects of drotrecogin alfa (activated) in patients with severe sepsis with or without overt disseminated intravascular coagulation. J Tromb Haemost 2004, 2: 1924-1933. 10.1111/j.1538-7836.2004.00955.x

    Article  CAS  Google Scholar 

  8. Kienast J, Juers M, Wiedermann CJ, Hoffmann JN, Ostermann H, Strauss R, Keinecke HO, Warren BL, Opal SM: Treatment effects of high-dose antithrombin without concomitant heparin in patients with severe sepsis with or without disseminated intravascular coagulation. J Tromb Haemost 2006, 4: 90-97. 10.1111/j.1538-7836.2005.01697.x

    Article  CAS  Google Scholar 

  9. Gando S, Saitoh D, Ishikura H, Ueyama M, Otomo Y, Oda S, Kushimoto S, Tanjoh K, Mayumi T, Ikeda T, Iba T, Eguchi Y, Okamoto K, Ogura H, Koseki K, Sakamoto Y, Takayama Y, Shirai K, Takasu O, Inoue Y, Mashiko K, Tsubota T, Endo S: A randomized, controlled, multicenter trial of the effects of antithrombin on disseminated intravascular coagulation in patients with sepsis. Crit Care 2013, 17: R297.

  10. Wada H, Thachil J, Di Nisio M, Mathew P, Kurosawa S, Gando S, Kim HK, Nielsen JD, Dempfle CE, Levi M, Toh CH: The Scientific Standardization Committee on DIC of the International Society on Thrombosis Haemostasis: Guidance for diagnosis and treatment of DIC from harmonization of the recommendations from three guidelines. J Thromb Haemost 2013, 11: 761-767. 10.1111/jth.12155

    Article  CAS  Google Scholar 

  11. Iba T, Kidokoro A: High-dose antithrombin therapy for sepsis: mechanisms of action. Shock 2002, 18: 389-394. 10.1097/00024382-200211000-00001

    Article  PubMed  Google Scholar 

  12. Hoffmann JN, Muhlbayer D, Jochum M, Inthorn D: Effect of long-term and high-dose antithrombin supplementation on coagulation and fibrinolysis in patients with severe sepsis. Crit Care Med 2004, 32: 1851-1859. 10.1097/01.CCM.0000139691.54108.1F

    Article  CAS  PubMed  Google Scholar 

  13. Moulin F, Pearson JM, Schultze AE: Thrombin is a distal mediator of lipopolysaccharide-induced liver injury in the rat. J Surg Res 1996, 65: 149-158. 10.1006/jsre.1996.0358

    Article  CAS  PubMed  Google Scholar 

  14. Moser R, Groscurth P, Fehr J: Promotion of transendothelial neutrophil passage by human thrombin. J Cell Sci 1990, 96: 737-744.

    CAS  PubMed  Google Scholar 

  15. Shankar R, de la Motte CA, Poptic EJ, DiCorleto PE: Thrombin receptor activating peptides differentially stimulate platelet derived growth factor production, monocytic cell adhesion, and E-selectin expression in human umbilical vein endothelial cells. J Biol Chem 1994, 269: 13936-13941.

    CAS  PubMed  Google Scholar 

  16. Sugama Y, Tiruppathi C, Offakidevi K, Andersen TT, Fenton JW II, Malik AB: Thrombin-induced expression of endothelial P-selectin and intracellular adhesion molecule-1: a mechanism for stabilizing neutrophil adhesion. J Cell Biol 1992, 119: 935-944. 10.1083/jcb.119.4.935

    Article  CAS  PubMed  Google Scholar 

  17. Coughlin SR: Thrombin signalling and protease-activated receptors. Nature 2000, 407: 258-264. 10.1038/35025229

    Article  CAS  PubMed  Google Scholar 

  18. Ossovskaya VS, Bunnett NW: Protease-activated receptors: contribution to physiology and disease. Physiol Rev 2004, 84: 579-621. 10.1152/physrev.00028.2003

    Article  CAS  PubMed  Google Scholar 

  19. Riewald M, Petrovan RJ, Donner A, Mueller BM, Ruf W: Activation of endothelial cell protease activated receptor 1 by the protein C pathway. Science 2002, 296: 1880-1882. 10.1126/science.1071699

    Article  CAS  PubMed  Google Scholar 

  20. Roemisch J, Gray E, Hoffmann JN, Wiedermann CJ: Antithrombin: a new look at the actions of a serine protease inhibitor. Blood Coagul Fibrinol 2002, 13: 657-670. 10.1097/00001721-200212000-00001

    Article  CAS  Google Scholar 

  21. Uchiba M, Okajima K: Antithrombin III (AT III) prevents LPS-induced pulmonary vascular injury: novel biological activity of AT III. Semin Thromb Hemost 1997, 23: 583-590. 10.1055/s-2007-996140

    Article  CAS  PubMed  Google Scholar 

  22. Koca U, Olguner C, Ozkardeşler S, Karci A, Coker C, Tuncel P, Taşdöĝen A, Duru S, Ulukuş C, Elar Z: Antithrombin III pretreatment reduces neutrophil recruitment into the lung in a rat model of abdominal sepsis. Acta Anaesthesiol Scand 2005, 49: 203-208. 10.1111/j.1399-6576.2004.00567.x

    Article  CAS  PubMed  Google Scholar 

  23. Zhou W, Hashimoto K, Goleniewska K, O'Neal JF, Ji S, Blackwell TS, Fitzgerald GA, Egan KM, Geraci MW, Peebles RS Jr: Prostaglandin I2 analogs inhibit proinflammatory cytokine production and T cell stimulatory function of dendritic cells. J Immunol 2007, 178: 702-710. 10.4049/jimmunol.178.2.702

    Article  CAS  PubMed  Google Scholar 

  24. Nishijima K, Kiryu J, Tsujikawa A, Honjo M, Nonaka A, Yamashiro K, Kamizuru H, Ieki Y, Tanihara H, Honda Y, Ogura Y: Inhibitory effects of antithrombin III on interactions between blood cells and endothelial cells during retinal ischemia-reperfusion injury. Invest Ophthalmol Vis Sci 2003, 44: 332-341. 10.1167/iovs.02-0493

    Article  PubMed  Google Scholar 

  25. Darbousset R, Thomas GM, Mezouar S, Frère C, Bonier R, Mackman N, Renné T, Dignat-George F, Dubois C, Panicot-Dubois L: Tissue factor-positive neutrophils bind to injured endothelial wall and initiate thrombus formation. Blood 2012, 120: 2133-2143. 10.1182/blood-2012-06-437772

    Article  CAS  PubMed  Google Scholar 

  26. Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A: Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 2007, 176: 231-241. 10.1083/jcb.200606027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P: Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 2007, 13: 463-469. 10.1038/nm1565

    Article  CAS  PubMed  Google Scholar 

  28. von Brühl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Köllnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S: Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012, 209: 819-835. 10.1084/jem.20112322

    Article  PubMed  PubMed Central  Google Scholar 

  29. Fuchs TA, Bhandari AA, Wagner DD: Histones induce rapid and profound thrombocytopenia in mice. Blood 2011, 118: 3708-3714. 10.1182/blood-2011-01-332676

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kambas K, Mitroulis I, Apostolidou E, Girod A, Chrysanthopoulou A, Pneumatikos I, Skendros P, Kourtzelis I, Koffa M, Kotsianidis I, Ritis K: Autophagy mediates the delivery of thrombogenic tissue factor to neutrophil extracellular traps in human sepsis. PLoS One 2012, 7: e45427.

  31. Iba T, Miki T, Hashiguchi N, Tabe Y, Nagaoka I: Combination of antithrombin and recombinant thrombomodulin modulates neutrophil cell-death and decreases circulating DAMPs levels in endotoxemic rats. Thromb Res 2014, 134: 169-173. 10.1016/j.thromres.2014.04.015

    Article  CAS  PubMed  Google Scholar 

  32. Iba T, Miki T, Hashiguchi N, Yamada A, Nagaoka I: Combination of antithrombin and recombinant thrombomodulin attenuates leukocyte-endothelial interaction and suppresses the increase of intrinsic damage-associated molecular patterns in endotoxemic rats. J Surg Res 2014, 187: 581-586. 10.1016/j.jss.2013.10.058

    Article  CAS  PubMed  Google Scholar 

  33. Triantaphyllopoulos DC: Effects of human antithrombin III on mortality and blood coagulation induced in rabbits by endotoxin. Thromb Haemost 1984, 51: 232-235.

    CAS  PubMed  Google Scholar 

  34. Taylor FB Jr, Emerson TE Jr, Jordan R, Chang AK, Blick KE: Antithrombin-III prevents the lethal effects of Escherichia coli infusion in baboons. Circ Shock 1988, 26: 227-235.

    CAS  PubMed  Google Scholar 

  35. Emerson TE Jr, Fournel MA, Leach WJ, Redens TB: Protection against disseminated intravascular coagulation and death by antithrombin-III in the Escherichia coli endotoxemic rat. Circ Shock 1987, 21: 1-13.

    CAS  PubMed  Google Scholar 

  36. Dickneite G, Paques EP: Reduction of mortality with antithrombin III in septicemic rats: a study of Klebsiella pneumoniae-induced sepsis. Thromb Haemost 1993, 69: 98-102.

    CAS  PubMed  Google Scholar 

  37. Iba T, Yagi Y, Kidokoro A, Ohno Y, Tankaka H, Kaneshiro Y, Kaneko K: Protective effects of antithrombin III on organ dysfunction induced by the continuous infusion of endotoxin in rats. Jpn Assoc Acute Med 1998, 9: 45-52.

    Google Scholar 

  38. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, Fisher CJ Jr: Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med 2001, 344: 699-709. 10.1056/NEJM200103083441001

    Article  CAS  PubMed  Google Scholar 

  39. Hoffmann JN, Wiedermann CJ, Juers M, Ostermann H, Kienast J, Briegel J, Strauss R, Warren BL, Opal SM: Benefit/risk profile of high-dose antithrombin in patients with severe sepsis treated with and without concomitant heparin. Thromb Haemost 2006, 95: 850-856.

    CAS  PubMed  Google Scholar 

  40. Wiedermann CJ, Hoffmann JN, Juers M, Ostermann H, Kienast J, Briegel J, Strauss R, Keinecke HO, Warren BL, Opal SM: High-dose antithrombin III in the treatment of severe sepsis in patients with a high risk of death: efficacy and safety. Crit Care Med 2006, 34: 285-292. 10.1097/01.CCM.0000194731.08896.99

    Article  CAS  PubMed  Google Scholar 

  41. Iba T, Saito D, Wada H, Asakura H: Efficacy and bleeding risk of antithrombin supplementation in septic disseminated intravascular coagulation: a prospective multicenter survey. Thromb Res 2012, 130: e129-e133. 10.1016/j.thromres.2012.03.021

    Article  CAS  PubMed  Google Scholar 

  42. Iba T, Saitoh D, Wada H, Asakura H: Efficacy and bleeding risk of antithrombin supplementation in septic disseminated intravascular coagulation: a secondary survey. Crit Care 2014, 18: 497.

  43. Nakahara K, Okuse C, Adachi S, Suetani K, Kitagawa S, Okano M, Michikawa Y, Takagi R, Shigefuku R, Itoh F: Use of antithrombin and thrombomodulin in the management of disseminated intravascular coagulation in patients with acute cholangitis. Gut Liver 2013, 7: 363-370. 10.5009/gnl.2013.7.3.363

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tagami T, Matsui H, Horiguchi H, Fushimi K, Yasunaga H: Antithrombin and mortality in severe pneumonia patients with sepsis-associated disseminated intravascular coagulation: an observational nationwide study. J Thromb Haemost 2014, 12: 1470-1479.

    Article  CAS  PubMed  Google Scholar 

  45. Fourrier F, Chopin C, Goudemand J, Hendrycx S, Caron C, Rime A, Marey A, Lestavel P: Septic shock, multiple organ failure, and disseminated intravascular coagulation. Compared patterns of antithrombin III, protein C, and protein S deficiencies. Chest 1992, 101: 816-823. 10.1378/chest.101.3.816

    Article  CAS  PubMed  Google Scholar 

  46. Lauterbach R, Pawlik D, Radziszewska R, Wozniak J, Rytlewski K: Plasma antithrombin III and protein C levels in early recognition of late-onset sepsis in newborns. Eur J Pediatr 2006, 165: 585-589. 10.1007/s00431-006-0139-7

    Article  CAS  PubMed  Google Scholar 

  47. Rodeghiero F, Mannucci PM, Viganò S, Barbui T, Gugliotta L, Cortellaro M, Dini E: Liver dysfunction rather than intravascular coagulation as the main cause of low protein C and antithrombin III in acute leukemia. Blood 1984, 63: 965-969.

    CAS  PubMed  Google Scholar 

  48. Sié P, Letrenne E, Caranobe C, Genestal M, Cathala B, Boneu B: Factor II related antigen and antithrombin III levels as indicators of liver failure in consumption coagulopathy. Thromb Haemost 1982, 47: 218-220.

    PubMed  Google Scholar 

  49. Jordan RE, Kilpatrick J, Nelson RM: Heparin promotes the inactivation of antithrombin by neutrophil elastase. Science 1987, 237: 777-779. 10.1126/science.3649921

    Article  CAS  PubMed  Google Scholar 

  50. Seitz R, Wolf M, Egbring R, Havemann K: The disturbance of hemostasis in septic shock: role of neutrophil elastase and thrombin, effects of antithrombin III and plasma substitution. Eur J Haematol 1989, 43: 22-28. 10.1111/j.1600-0609.1989.tb01246.x

    Article  CAS  PubMed  Google Scholar 

  51. Iba T, Yagi Y, Kidokoro A: The role of the endothelium in changes in procoagulant activity in sepsis. J Am Coll Surg 1998, 187: 321-329. 10.1016/S1072-7515(98)00177-X

    Article  CAS  PubMed  Google Scholar 

  52. Pettila V, Pentti J, Pettila M, Takkunen O, Jousela I: Predictive value of antithrombin III and serum C-reactive protein concentration in critically ill patients with suspected sepsis. Crit Care Med 2002, 30: 271-275. 10.1097/00003246-200202000-00001

    Article  CAS  PubMed  Google Scholar 

  53. Iba T, Kidokoro A, Fukunaga M, Sugiyama K, Kato H: Association between the severity of sepsis and the changes in hemostatic molecular markers and vascular endothelial damage markers. Shock 2005, 23: 25-29. 10.1097/01.shk.0000144422.32647.b6

    Article  CAS  PubMed  Google Scholar 

  54. 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. Ann Lab Med 2014, 34: 85-91. 10.3343/alm.2014.34.2.85

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Cauchie P, Cauchie C, Boudjeltia KZ, Carlier E, Deschepper N, Govaerts D, Migaud-Fressart M, Woodhams B, Brohée D: Diagnosis and prognosis of overt disseminated intravascular coagulation in a general hospital—meaning of the ISTH score system, fibrin monomers, and lipoprotein-C-reactive protein complex formation. Am J Hematol 2006, 81: 414-419. 10.1002/ajh.20597

    Article  CAS  PubMed  Google Scholar 

  56. Iba T, Gando S, Murata A, Kushimoto S, Saitoh D, Eguchi Y, Ohtomo Y, Okamoto K, Koseki K, Mayumi T, Ikeda T, Ishikura H, Ueyama M, Ogura H, Endo S, Shimazaki S: Predicting the severity of SIRS-associated coagulopathy with hemostatic molecular markers and vascular endothelial injury markers. J Trauma 2007, 63: 1093-1098. 10.1097/01.ta.0000251420.41427.d3

    Article  PubMed  Google Scholar 

  57. Yanagida Y, Gando S, Sawamura A, Hayakawa M, Uegaki S, Kubota N, Homma T, Ono Y, Honma Y, Wada T, Jesmin S: Normal prothrombinase activity, increased systemic thrombin activity, and lower antithrombin levels in patients with disseminated intravascular coagulation at an early phase of trauma: comparison with acute coagulopathy of trauma-shock. Surgery 2013, 154: 48-57. 10.1016/j.surg.2013.02.004

    Article  PubMed  Google Scholar 

  58. Oshiro A, Yanagida Y, Gando S, Henzan N, Takahashi I, Makise H: Hemostasis during the early stages of trauma: comparison with disseminated intravascular coagulation. Crit Care 2014, 18: R61.

  59. Favaloro EJ: Laboratory testing in disseminated intravascular coagulation. Semin Thromb Hemost 2010, 36: 458-467. 10.1055/s-0030-1254055

    Article  CAS  PubMed  Google Scholar 

  60. Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M: Scientific Subcommittee on Disseminated Intravascular Coagulation (DIC) of the International Society on Thrombosis and Haemostasis (ISTH): Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb Haemost 2001, 86: 1327-1330.

    CAS  PubMed  Google Scholar 

  61. Gando S, Iba T, Eguchi Y, Ohtomo Y, Okamoto K, Koseki K, Mayumi T, Murata A, Ikeda T, Ishikura H, Ueyama M, Ogura H, Kushimoto S, Saitoh D, Endo S, Shimazaki S: A multicenter, prospective validation of disseminated intravascular coagulation diagnostic criteria for critically ill patients: comparing current criteria. Crit Care Med 2006, 34: 625-631.

    Article  PubMed  Google Scholar 

  62. Oh D, Jang MJ, Lee SJ, Chong SY, Kang MS, Wada H: Evaluation of modified non-overt DIC criteria on the prediction of poor outcome in patients with sepsis. Thromb Res 2010, 126: 18-23. 10.1016/j.thromres.2009.12.008

    Article  CAS  PubMed  Google Scholar 

  63. Egi M, Morimatsu H, Wiedermann CJ, Tani M, Kanazawa T, Suzuki S, Matsusaki T, Shimizu K, Toda Y, Iwasaki T, Morita K: Non-overt disseminated intravascular coagulation scoring for critically ill patients: the impact of antithrombin levels. Thromb Haemost 2009, 101: 696-705.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A portion of this review was presented in the 40th Annual Meeting of the Japanese Society of Intensive Care Medicine.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Toshiaki Iba.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

IT and SD wrote the manuscript. Both authors read and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iba, T., Saitoh, D. Efficacy of antithrombin in preclinical and clinical applications for sepsis-associated disseminated intravascular coagulation. j intensive care 2, 66 (2014). https://doi.org/10.1186/s40560-014-0051-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40560-014-0051-6

Keywords