CC BY 4.0 · TH Open 2023; 07(01): e42-e55
DOI: 10.1055/a-2000-6576
Original Article

In vitro Effect of Dalteparin and Argatroban on Hemostasis in Critically Ill Sepsis Patients with New-Onset Thrombocytopenia

Søren Nygaard
1   Department of Clinical Biochemistry, Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark
2   Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
,
Christine L. Hvas
2   Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
3   Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
,
4   Faculty of Health, Aarhus University, Aarhus, Denmark
,
Kasper Adelborg
1   Department of Clinical Biochemistry, Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark
2   Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
5   Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
6   Department of Clinical Biochemistry, Gødstrup Regional Hospital, Herning, Denmark
› Author Affiliations
Funding The study was fully supported by the Aarhus University Research Foundation (fund license no.: AU FF-E-2020-7-20).

Abstract

Thrombocytopenia is common among critically ill sepsis patients, while they also hold an increased risk for thromboembolic events. Thus, the choice of anticoagulant prophylaxis for this patient population is challenging. We investigated the in vitro effect of low-molecular-weight heparin (dalteparin) and direct thrombin inhibitor (argatroban) on the hemostasis in blood from sepsis patients with new-onset thrombocytopenia. Thrombocytopenia was defined as a platelet count drop of ≥30% and/or from >100 × 109/L to 30 to 100 × 109/L within 24 hours prior to inclusion. We included five healthy individuals and ten patients. Analyses of thrombin generation (Calibrated Automated Thrombogram), thrombin-antithrombin (TAT) complex levels, prothrombin fragment 1+2 (F1+2), and rotational thromboelastometry (ROTEM) were performed. Based on dose–response relationships investigated in healthy blood, patient samples were spiked with prophylactic (0.25 IU/mL) and therapeutic (0.75 IU/mL) dalteparin and low (0.25 µg/mL) and high (0.50 µg/mL) argatroban concentrations, each with a sample without anticoagulant. In patients, the endogenous thrombin potential was markedly lower in therapeutic dalteparin samples than in samples without anticoagulant [median (range): 29 (0–388) vs. 795 (98–2121) nM × min]. In high argatroban concentration samples, thrombin lag time was longer than in samples without anticoagulant [median (range): 15.5 (10.5–20.2) versus 5.3 (2.8–7.3) min]. Dalteparin and argatroban both increased clotting time but did not affect maximum clot firmness in the ROTEM INTEM assay. Six patients had elevated TAT and eight patients had elevated F1 + 2. In conclusion, dalteparin mainly affected the amount of thrombin generated and argatroban delayed clot initiation in critically ill sepsis patients with new-onset thrombocytopenia. Neither anticoagulant affected clot strength.

Authors' Contributions

The study was conceptualized by K.A. and AM.H. All authors were involved in the design of the study. The screening and inclusion process was made feasible by C.L.H. and performed by S.N. The medical record data extraction, laboratory analyses, statistics, graphics, and data management were performed by S.N. K.A. had the primary responsibility of the study and supervised the process from start to finish. S.N. wrote the first draft of the manuscript, which was reviewed by all authors. All authors approved the manuscript prior to submission.


Supplementary Material



Publication History

Received: 02 September 2022

Accepted: 14 December 2022

Accepted Manuscript online:
18 December 2022

Article published online:
30 January 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Levi M, Poll Tv. Coagulation in patients with severe sepsis. Semin Thromb Hemost 2015; 41 (01) 9-15
  • 2 Kinasewitz GT, Yan SB, Basson B. et al; PROWESS Sepsis Study Group. Universal changes in biomarkers of coagulation and inflammation occur in patients with severe sepsis, regardless of causative micro-organism [ISRCTN74215569]. Crit Care 2004; 8 (02) R82-R90
  • 3 Ogura H, Gando S, Saitoh D. et al; Japanese Association for Acute Medicine Sepsis Registry (JAAMSR) Study Group. Epidemiology of severe sepsis in Japanese intensive care units: a prospective multicenter study. J Infect Chemother 2014; 20 (03) 157-162
  • 4 Venkata C, Kashyap R, Farmer JC, Afessa B. Thrombocytopenia in adult patients with sepsis: incidence, risk factors, and its association with clinical outcome. J Intensive Care 2013; 1 (01) 9
  • 5 Vanderschueren S, De Weerdt A, Malbrain M. et al. Thrombocytopenia and prognosis in intensive care. Crit Care Med 2000; 28 (06) 1871-1876
  • 6 Bachler M, Asmis LM, Koscielny J. et al. Thromboprophylaxis with argatroban in critically ill patients with sepsis: a review. Blood Coagul Fibrinolysis 2022; 33 (05) 239-256
  • 7 Alberio L, Angelillo-Scherrer A, Asmis L. et al. Recommendations on the use of anticoagulants for the treatment of patients with heparin-induced thrombocytopenia in Switzerland. Swiss Med Wkly 2020; 150: w20210
  • 8 Beiderlinden M, Treschan TA, Görlinger K, Peters J. Argatroban anticoagulation in critically ill patients. Ann Pharmacother 2007; 41 (05) 749-754
  • 9 Saugel B, Phillip V, Moessmer G, Schmid RM, Huber W. Argatroban therapy for heparin-induced thrombocytopenia in ICU patients with multiple organ dysfunction syndrome: a retrospective study. Crit Care 2010; 14 (03) R90
  • 10 Lewis BE, Wallis DE, Berkowitz SD. et al; ARG-911 Study Investigators. Argatroban anticoagulant therapy in patients with heparin-induced thrombocytopenia. Circulation 2001; 103 (14) 1838-1843
  • 11 Fu S, Yu S, Wang L, Ma X, Li X. Unfractionated heparin improves the clinical efficacy in adult sepsis patients: a systematic review and meta-analysis. BMC Anesthesiol 2022; 22 (01) 28
  • 12 Singer M, Deutschman CS, Seymour CW. et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA 2016; 315 (08) 801-810
  • 13 Schulman S, Kearon C. Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost 2005; 3 (04) 692-694
  • 14 Lim W. Using low molecular weight heparin in special patient populations. J Thromb Thrombolysis 2010; 29 (02) 233-240
  • 15 Alhenc-Gelas M, Jestin-Le Guernic C, Vitoux JF, Kher A, Aiach M, Fiessinger JN. Fragmin-Study Group. Adjusted versus fixed doses of the low-molecular-weight heparin fragmin in the treatment of deep vein thrombosis. Thromb Haemost 1994; 71 (06) 698-702
  • 16 Colucci G, Nagler M, Klaus N, Conte T, Giabbani E, Alberio L. Practical guidelines for argatroban and bivalirudin in patients with heparin-induced thrombocytopenia. J Transl Sci 2015; 1: 37-42
  • 17 Ostrowski SR, Søgaard OS, Tolstrup M. et al. Inflammation and platelet activation after COVID-19 vaccines - possible mechanisms behind vaccine-induced immune thrombocytopenia and thrombosis. Front Immunol 2021; 12: 779453
  • 18 Lundbech M, Krag AE, Christensen TD, Hvas AM. Thrombin generation, thrombin-antithrombin complex, and prothrombin fragment F1+2 as biomarkers for hypercoagulability in cancer patients. Thromb Res 2020; 186: 80-85
  • 19 Moreno RP, Metnitz PG, Almeida E. et al; SAPS 3 Investigators. SAPS 3–From evaluation of the patient to evaluation of the intensive care unit. Part 2: Development of a prognostic model for hospital mortality at ICU admission. Intensive Care Med 2005; 31 (10) 1345-1355
  • 20 Quan H, Li B, Couris CM. et al. Updating and validating the Charlson comorbidity index and score for risk adjustment in hospital discharge abstracts using data from 6 countries. Am J Epidemiol 2011; 173 (06) 676-682
  • 21 Taylor Jr FB, 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 (05) 1327-1330
  • 22 Larsen JB, Aggerbeck MA, Granfeldt A, Schmidt M, Hvas AM, Adelborg K. Disseminated intravascular coagulation diagnosis: positive predictive value of the ISTH score in a Danish population. Res Pract Thromb Haemost 2021; 5 (08) e12636
  • 23 Adelborg K, Larsen JB, Hvas AM. Disseminated intravascular coagulation: epidemiology, biomarkers, and management. Br J Haematol 2021; 192 (05) 803-818
  • 24 Iba T, Di Nisio M, Thachil J. et al. Revision of the Japanese Association for Acute Medicine (JAAM) disseminated intravascular coagulation (DIC) diagnostic criteria using antithrombin activity. Crit Care 2016; 20: 287
  • 25 Iba T, Nisio MD, Levy JH, Kitamura N, Thachil J. New criteria for sepsis-induced coagulopathy (SIC) following the revised sepsis definition: a retrospective analysis of a nationwide survey. BMJ Open 2017; 7 (09) e017046
  • 26 Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform 2009; 42 (02) 377-381
  • 27 Hacquard M, Perrin J, Lelievre N, Vigneron C, Lecompte T. Inter-individual variability of effect of 7 low molecular weight antithrombin-dependent anticoagulants studied in vitro with calibrated automated thrombography. Thromb Res 2011; 127 (01) 29-34
  • 28 Robert S, Ghiotto J, Pirotte B. et al. Is thrombin generation the new rapid, reliable and relevant pharmacological tool for the development of anticoagulant drugs?. Pharmacol Res 2009; 59 (03) 160-166
  • 29 Feuring M, Wehling M, Schultz A. Dalteparin dose-dependently increases ROTEM(®) thrombelastography parameters only at supratherapeutic anti-factor Xa levels: an in vitro study. Clin Exp Pharmacol Physiol 2011; 38 (11) 783-786
  • 30 Thomas O, Larsson A, Tynngård N, Schött U. Thromboelastometry versus free-oscillation rheometry and enoxaparin versus tinzaparin: an in-vitro study comparing two viscoelastic haemostatic tests' dose-responses to two low molecular weight heparins at the time of withdrawing epidural catheters from ten patients after major surgery. BMC Anesthesiol 2015; 15: 170
  • 31 Schaden E, Schober A, Hacker S, Spiss C, Chiari A, Kozek-Langenecker S. Determination of enoxaparin with rotational thrombelastometry using the prothrombinase-induced clotting time reagent. Blood Coagul Fibrinolysis 2010; 21 (03) 256-261
  • 32 Ustinov NB, Zav'yalova EG, Kopylov AM. Effect of thrombin inhibitors on positive feedback in the coagulation cascade. Biochemistry (Mosc) 2016; 81 (03) 242-248
  • 33 Engström M, Rundgren M, Schött U. An evaluation of monitoring possibilities of argatroban using rotational thromboelastometry and activated partial thromboplastin time. Acta Anaesthesiol Scand 2010; 54 (01) 86-91
  • 34 Beiderlinden M, Werner P, Bahlmann A. et al. Monitoring of argatroban and lepirudin anticoagulation in critically ill patients by conventional laboratory parameters and rotational thromboelastometry - a prospectively controlled randomized double-blind clinical trial. BMC Anesthesiol 2018; 18 (01) 18
  • 35 Guy S, Kitchen S, Maclean R, Van Veen JJ. Limitation of the activated partial thromboplastin time as a monitoring method of the direct thrombin inhibitor argatroban. Int J Lab Hematol 2015; 37 (06) 834-843
  • 36 Gosselin RC, King JH, Janatpour KA, Dager WE, Larkin EC, Owings JT. Comparing direct thrombin inhibitors using aPTT, ecarin clotting times, and thrombin inhibitor management testing. Ann Pharmacother 2004; 38 (09) 1383-1388
  • 37 Wolberg AS. Thrombin generation and fibrin clot structure. Blood Rev 2007; 21 (03) 131-142
  • 38 Zucker M, Seligsohn U, Salomon O, Wolberg AS. Abnormal plasma clot structure and stability distinguish bleeding risk in patients with severe factor XI deficiency. J Thromb Haemost 2014; 12 (07) 1121-1130
  • 39 Aird WC. Vascular bed-specific hemostasis: role of endothelium in sepsis pathogenesis. Crit Care Med 2001; 29 (Suppl. 07) S28-S34 , discussion S34–S35
  • 40 Hoppensteadt D, Tsuruta K, Cunanan J. et al. Thrombin generation mediators and markers in sepsis-associated coagulopathy and their modulation by recombinant thrombomodulin. Clin Appl Thromb Hemost 2014; 20 (02) 129-135
  • 41 Butenas S, Branda RF, van't Veer C, Cawthern KM, Mann KG. Platelets and phospholipids in tissue factor-initiated thrombin generation. Thromb Haemost 2001; 86 (02) 660-667
  • 42 Gerotziafas GT, Depasse F, Busson J, Leflem L, Elalamy I, Samama MM. Towards a standardization of thrombin generation assessment: the influence of tissue factor, platelets and phospholipids concentration on the normal values of Thrombogram-Thrombinoscope assay. Thromb J 2005; 3: 16
  • 43 Chowdary P, Hamid C, Slatter D. et al. Impaired platelet-dependent thrombin generation associated with thrombocytopenia is improved by prothrombin complex concentrates in vitro. Res Pract Thromb Haemost 2020; 4 (02) 334-342
  • 44 Hemker HC, Giesen P, Al Dieri R. et al. Calibrated automated thrombin generation measurement in clotting plasma. Pathophysiol Haemost Thromb 2003; 33 (01) 4-15
  • 45 Dargaud Y, Luddington R, Gray E. et al. Effect of standardization and normalization on imprecision of calibrated automated thrombography: an international multicentre study. Br J Haematol 2007; 139 (02) 303-309