Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1999 word(s) 1999 2021-05-07 10:44:34

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Andryukov, B. Seaweed Sulfated Polysaccharides. Encyclopedia. Available online: https://encyclopedia.pub/entry/9384 (accessed on 02 July 2024).
Andryukov B. Seaweed Sulfated Polysaccharides. Encyclopedia. Available at: https://encyclopedia.pub/entry/9384. Accessed July 02, 2024.
Andryukov, Boris. "Seaweed Sulfated Polysaccharides" Encyclopedia, https://encyclopedia.pub/entry/9384 (accessed July 02, 2024).
Andryukov, B. (2021, May 07). Seaweed Sulfated Polysaccharides. In Encyclopedia. https://encyclopedia.pub/entry/9384
Andryukov, Boris. "Seaweed Sulfated Polysaccharides." Encyclopedia. Web. 07 May, 2021.
Seaweed Sulfated Polysaccharides
Edit

Seaweed sulfated polysaccharides (PSs) has wide therapeutic potential, including anticoagulant, thrombolytic, and fibrinolytic activities, opens up new possibilities for their study in experimental and clinical trials. These natural compounds can be important complementary drugs for the recovery from hemostasis disorders due to their natural origin, safety, and low cost compared to synthetic drugs.

seaweed polysaccharides fucoidans carrageenans ulvans anticoagulants thrombolytics fibrinolytics thrombin inhibitors hemostasis disorders blood coagulation COVID-19 associated coagulopathy SARS-CoV-2

1. Introduction

Hemostasis disorders play an important role in the pathogenesis, clinical manifestations, and outcome of COVID-19. First of all, the hemostasis system suffers due to a complicated and severe course of COVID-19. In the initial stage of the disease, a significant number of COVID-19 patients develop signs of hypercoagulation, and in the later stages, signs of disseminated intravascular coagulation (DIC) syndrome. Coagulation disorders and their clinical manifestations have a number of differences from the classic DIC syndrome, and are named COVID-associated coagulopathy [1][2][3].

Patients with severe COVID-19 have a tendency toward thrombotic complications in the venous and arterial systems, which is the leading cause of death in this disease [1][4][5][6].

The treatment of COVID-19 patients and, in particular, correction of the hemostasis disorders is a difficult problem. Despite the data on the effectiveness of heparin drugs in reducing mortality in severe COVID-19 patients, and despite thromboprophylaxis [7][8], the development of venous and arterial thromboembolic complications has been reported. There is also a problem with the optimal dosage of anticoagulant agent [5][9][10][11]. Thus, there are a number of unresolved issues regarding the use of these drugs.

In this regard, there is a need to use the effective anticoagulants and antithrombotic drugs, as well as their optimal dosages, in the treatment of COVID-19 patients and for the prevention of thrombotic complications [7][12][13].

Despite the success achieved in SARS-CoV-2 therapy, the development of more effective treatment regimens for this infection with the inclusion of anticoagulants, thrombolytics, and fibrinolytics, as well as the search for new effective drugs of this pharmacological group, does not lose relevance. The wide therapeutic potential of seaweed sulfated polysaccharides (PSs), including anticoagulant, thrombolytic, and fibrinolytic activities, opens up a new possibilities for their study in experimental and clinical trials.

2. Treatment Strategies of COVID-19-Induced Hypercoagulation and the Potency of Seaweed Sulfated PSs for the Correction of Hemostasis Disorders

The treatment of COVID-19 patients, and in particular hemostasis disorders, is challenging and a difficult issue, since the pathogenesis of the disorders is not completely clear.
Addressing the need for the use of anticoagulants, their optimal dosage in the prevention and treatment of patients with COVID-19, and the control of anticoagulant therapy are important issues. Heparin drugs are often used as anticoagulants in the treatment of patients with COVID-19. There is unfractionated heparin (UFH)—first-generation preparations called heparin, which are a mixture of PSs with a molecular weight (MW) in the range of 2–30 kDa (with a predominance of high-molecular fractions of glycosaminoglycan), and low-molecular-weight heparin (LMWH), obtained by chemical or enzymatic hydrolysis of UFH and having a molecular weight of 3–7 kDa.
The mechanism of anticoagulant action of heparin is to inhibit the activity of thrombin (clotting factor IIa), which catalyzes the conversion of fibrinogen to fibrin and some other reactions in the hemostatic system. The antithrombin activity of heparin depends on the presence of the plasma protein antithrombin III (AT III). When heparin binds to AT III, conformational changes occur in the latter molecule, which allow it to quickly connect to the active center of thrombin and other serine proteases (factors IXa, Xa, XIa and XIIa, kallikrein, and plasmin) [14]. Therefore, heparin inhibits thrombosis, contributing to the inactivation of thrombin by its physiological inhibitor AT III. In the presence of heparin, the inactivation of thrombin by antithrombin III is accelerated about 1000-fold. Slightly less important for the manifestation of the anticoagulant effect of heparin is the “heparin II cofactor” (HC II), the second heparin-dependent thrombin inhibitor other than AT III, which neutralizes thrombin only at high concentrations of heparin in the blood plasma [15][16].
Both UFH and LMWH form complexes with AT III. However, if the UFH-AT III complex equally inhibits thrombin (factor IIa) and factor Xa, as well as other enzyme blood-clotting factors (Hageman factor, factors IX, XI, XII, etc.), then the anti-Xa activity prevails in LMWH [14][17][18][19].
Among the direct acting parenteral anticoagulants, it is recommended to give preference to LMWH compared to UFH, since the inhibitory activity of LMWH is stronger toward factor Xa, and LMWH rarely causes such complications as heparin—induced thrombocytopenia and osteoporosis. Qian et al. [20] observed a reduction of treatment duration in patients with acute exacerbation of chronic obstructive pulmonary disease receiving ventilatory support using LMWH. ISTH interim guidance recommends LMWH as a first-line therapy for the prophylaxis of VTE in hospitalized patients with COVID-19 [8]. The synthetic drug fondaparinux has also been shown to be effective for the prevention of VTE in older acute medical patients and in reducing total mortality [21].
According to Tang et al. [7], LMWH contributed to a decrease in mortality in patients with sepsis-induced coagulopathy. The same author has shown the effectiveness of enoxaparin for prophylactic coagulation abnormalities in COVID-19 patients.
With regard to the dosage of heparin, a prophylactic dose of LMWH is recommended for the prevention of VTE in hospitalized patients with COVID-19, and a therapeutic dose of LMWH is recommended for patients with significantly elevated D-dimer concentrations [22].
Therefore, taking into account the pathogenesis, prevention, and treatment regimens for severe complications of COVID-19, LMWH are included in the treatment protocols for all hospitalized patients. LMWH is also recommended for use in the outpatient setting. The administration of LMWH, the duration of its use, and the dose should be determined taking into account the risk factors for each patient in combination with laboratory monitoring.
Prospective clinical trials are ongoing to confirm the benefits of using anticoagulants to improve the survival of COVID-19-patients.
In addition, direct oral anticoagulants are used in COVID-19 patient therapy. However, as Testa et al. [23] have shown, when used concomitantly with antiviral agents, the latter, especially those that interact with P-glycoprotein and/or cytochrome P450-based metabolic pathways, can alter the pharmacokinetic and pharmacodynamic profiles, hence altering their anticoagulant activity and increasing the risk of bleeding. In this regard, although direct oral anticoagulants are convenient for the outpatient management of COVID-19 patients, caution is recommended, given their interaction with other drugs used to treat COVID-19 [24][25].
The use of heparins as anticoagulants is also indicated when taking into account the additional mechanisms of their action. In particular, heparin exhibits anti-inflammatory activity by inhibiting the recruitment of neutrophils into tissues, binding and neutralizing inflammatory cytokines and acute phase proteins, and potentially having a protective effect on the endothelium [26][27][28][29]. It also has been hypothesized that heparin may interfere with the interaction between the virus and the host cell via a nonspecific ion bond, and thus may contribute to reducing the frequency of infected cells in COVID-19 [27]. A number of studies have reported that the use of heparins in critically ill COVID-19 patients contributed to a decrease in the production of elevated cytokines (IL-6 and TNF-α) [30]. Additionally, heparin protects the endothelium [31]. There are also data that heparin interacts with spike proteins of several viruses, including the SARS-CoV-2 spike protein receptor-binding domain, suggesting that it may be able to modulate protein’s interactions with the endothelium [32]. Due to these characteristics, LMWH remains as the best choice of anticoagulant for hospitalized patients with severe COVID-19.
However, despite evidence that heparin in prophylactic doses is effective in reducing mortality in severe COVID-19 patients, a number of studies have reported a high incidence of venous and even arterial thromboembolic complications (deep vein thrombosis, pulmonary embolism or in situ thrombosis in the pulmonary arteries, and arterial thrombosis), despite thromboprophylaxis, which raises the issue of increasing anticoagulant doses. Prospective studies are currently in progress to try to answer this question [5][33][34].
White et al. [35] also reported heparin resistance in COVID-19. According to the authors, out of 14 patients with COVID-19 associated coagulopathy and with a high risk of thrombosis, who were treated with LMWH or UFH, resistance to UFH was documented in 8/10 (80%) patients, and suboptimal peak anti-Xa following therapeutic LMWH in 5/5 (100%) patients.

There is also the problem of anticoagulants’ optimal dose strategy. For example, a reduction in mortality in hospitalized patients with COVID-19 is shown when using a therapeutic, but not a preventive, dose of anticoagulants [12][13][36].

Taking into account the high frequency of thromboembolic complications, the International Society on Thrombosis and Haemostasis’ guidelines continue to recommend thromboprophylaxis for all hospitalized patients, and in most patients with COVID-19, thromboprophylaxis should be continued after hospital discharge [9].

The Expert Group of American Society of Hematology recommended the use of prophylactic-intensity anticoagulation compared to moderate-intensity or therapeutic-intensity anticoagulants for critical COVID-19-related patients or acute COVID-19 patients who do not have a confirmed VTE [37]. Taking into consideration their activation and their role in the clot formations, platelets are a potential target for the prevention of complications in SARS-CoV-2 infection [38][39][40][41]. The most commonly used antiplatelet agents are aspirin, clopidogrel, lopinavir/ritonavir, vorapaxar, and dipyridamole [41][42][43][44]. Among the antiplatelet agents, aspirin and clopidogrel are associated with decreased risk of ARDS, as well as decreased mortality among critically ill COVID-19 patients [43][44].

Vorapaxar and dipyridamole are considered promising antiplatelet agents for the treatment of COVID-19. Vorapaxar exhibits its antiplatelet activity through the antagonism of protease-activated receptor 1 (PAR-1), which plays an important role in thrombin-induced platelet aggregation, and is associated with blood clotting and inflammation [45].

For example, Liu et al. [42] used dipyridamole with a positive clinical effect. However, these authors note that there are a number of questions regarding the use of antiplatelet drugs in COVID-19 treatment, and therefore additional clinical trials are needed [42].

The data presented above indicate that anticoagulant and antithrombotic therapy play a key role in the treatment of hemostasis disorders in COVID-19, but there are still a number of unresolved issues regarding the use of these drugs. Thus, the optimal effective anticoagulant and antithrombotic agents, as well as their doses, remain uncertain.

A separate problem is the interaction of antithrombotic and anticoagulant drugs with specific drugs in COVID-19 treatments, since not all drugs are compatible. An individual approach to the patient is recommended, aimed at the optimal risk/benefit ratio of various antithrombotic strategies, taking into account the underlying hypercoagulable state.

Therefore, the treatment of patients and prevention of thrombosis in COVID-19 requires solving issues related to the need to find new effective anticoagulants, as well as their optimal dosage. In this regard, the search for new effective anticoagulants remains relevant. In this aspect, the sulfated PSs of seaweed are of interest.

3. Conclusions

Seaweed sulfated polysaccharides (PS) possess anticoagulant, antithrombotic, and fibrinolytic properties. These activities depends on the structural features (content and position of sulfate groups on the main chain of the PSs, molecular weight, monosaccharide composition and type of glycosidic bonds, the degree of PS chain branching, etc.). Sulfated PSs act on the hemostasis system both directly and indirectly and affect the factors of the extrinsic and intrinsic coagulation pathways, as well as the final stage of coagulation or the conversion of fibrinogen to fibrin under the thrombin influence. Anticoagulant activity of PS can be associated with plasma AT III, and antithrombotic activity—with an increase in the level of t-PA and competitive binding to the t-PA-PAI-1 complex.

The combination of the anticoagulant, antithrombotic, and fibrinolytic properties allows us to consider seaweed sulfated PSs as alternative sources of new anticoagulant and antithrombotic compounds. In this regard, and along with the low toxicity and relative low cost of their production, sulfated PSs are promising for clinical use and represent an alternative to heparin. The wide therapeutic potential of seaweed sulfated PSs, including anticoagulant, thrombolytic, and fibrinolytic activities, opens up new possibilities for their study in experimental and clinical trials. These natural compounds can be important complementary drugs in the fight against coronaviruses due to their natural origin, safety, and low cost compared to synthetic drugs. However, further investigations are needed regarding the use of seaweed PSs as potential anticoagulants, thrombolytics, and fibrinolytics aimed to correct hemostasis disorders in COVID-19 patients.

References

  1. Belen-Apak, F.B.; Sarıalioğlu, F. Pulmonary intravascular coagulation in COVID-19: Possible pathogenesis and recommendations on anticoagulant/thrombolytic therapy. J. Thromb. Thrombolysis 2020, 50, 278–280.
  2. Iba, T.; Levy, J.H.; Levi, M.; Connors, J.M.; Thachil, J. Coagulopathy of coronavirus disease. Crit. Care Med. 2020, 48, 1358–1364.
  3. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847.
  4. Berkman, S.A.; Tapson, V.F. COVID-19 and Its Implications for Thrombosis and Anticoagulation. Semin Respir Crit. Care Med. 2021, 42, 316–326.
  5. Klok, F.A.; Kruip, M.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.; Kant, K.M.; Kaptein, H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V.; et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020, 191, 145–147.
  6. Tan, B.K.; Mainbourg, S.; Friggeri, A.; Bertoletti, L.; Douplat, M.; Dargaud, Y.; Grange, C.; Lobbes, H.; Provencher, S.; Lega, J.C. Arterial and venous thromboembolism in COVID-19: A study-level meta-analysis. Thorax 2021, 23.
  7. Tang, N.; Bai, H.; Chen, X.; Gong, J.; Li, D.; Sun, Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020, 18, 1094–1099.
  8. Thachil, J.; Tang, N.; Gando, S.; Falanga, A.; Cattaneo, M.; Levi, M.; Clark, C.; Iba, T. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J. Thromb. Haemost. 2020, 18, 1023–1026.
  9. Kollias, A.; Kyriakoulis, K.G.; Dimakakos, E.; Poulakou, G.; Stergiou, G.S.; Syrigos, K. Thromboembolic risk and anticoagulant therapy in COVID-19 patients: Emerging evidence and call for action. Br. J. Haematol. 2020, 189, 846–847.
  10. Jonmarker, S.; Hollenberg, J.; Dahlberg, M.; Stackelberg, O.; Litorell, J.; Everhov, Å.H.; Järnbert-Pettersson, H.; Söderberg, M.; Grip, J.; Schandl, A.; et al. Dosing of thromboprophylaxis and mortality in critically ill COVID-19 patients. Crit. Care 2020, 24, 653.
  11. Kharma, N.; Roehrig, S.; Shible, A.A.; Elshafei, M.S.; Osman, D.; Elsaid, I.M.; Mustafa, S.F.; Aldabi, A.; Smain, O.A.M.; Lance, M.D. Anticoagulation in critically ill patients on mechanical ventilation suffering from COVID-19 disease, The ANTI-CO trial: A structured summary of a study protocol for a randomized controlled trial. Trials 2020, 21, 769.
  12. Lemos, A.C.B.; do Espírito Santo, D.A.; Salvetti, M.C.; Gilio, R.N.; Agra, L.B.; Pazin-Filho, A.; Miranda, C.H. Therapeutic versus prophylactic anticoagulation for severe COVID-19: A randomized phase II clinical trial (HESACOVID). Thromb. Res. 2020, 196, 359–366.
  13. Paranjpe, I.; Fuster, V.; Lala, A.; Russak, A.J.; Glicksberg, B.S.; Levin, M.A.; Charney, A.W.; Narula, J.; Fayad, Z.A.; Bagiella, E.; et al. Association of treatment dose anticoagulation within-hospital survival among hospitalized patients with COVID-19. J. Am. Coll. Cardiol. 2020, 76, 122–124.
  14. Spadarella, G.; Di Minno, A.; Donati, M.B.; Mormile, M.; Ventre, I.; Di Minno, G. From unfractionated heparin to pentasaccharide: Paradigm of rigorous science growing in the understanding of the in vivo thrombin generation. Blood Rev. 2020, 39, 10061.
  15. Hirsh, J.; Raschke, R. Heparin and low-molecular-weight heparin: The Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest 2004, 126, 188S–203S.
  16. Rezaie, A.R. Prothrombin protects factor Xa in the prothrombinase complex from inhibition by the heparin-antithrombin complex. Blood 2001, 97, 2308–2313.
  17. Bianchini, P.; Liverani, L.; Spelta, F.; Mascellani, G.; Parma, B. Variability of Heparins and Heterogeneity of Low Molecular Weight Heparins. Semin Thromb Hemost. 2007, 33, 496–502.
  18. Navarese, E.P.; De Luca, G.; Castriota, F.; Kozinski, M.; Gurbel, P.A.; Gibson, C.M.; Andreotti, F.; Buffon, A.; Siller-Matula, J.M.; Sukiennik, A.; et al. Low-molecular-weight heparins vs. unfractionated heparin in the setting of percutaneous coronary intervention for ST-elevation myocardial infarction: A meta-analysis. J. Thromb. Haemost. 2011, 9, 1902–1915.
  19. Padilla, A.; Gray, E.; Pepper, D.S.; Barrowcliffe, T.W. Inhibition of thrombin generation by heparin and low molecular weight (LMW) heparins in the absence and presence of platelet factor 4 (PF4). Br. J. Haematol. 1992, 82, 406–413.
  20. Qian, Y.; Xie, H.; Tian, R.; Yu, K.; Wang, R. Efficacy of low molecular weight heparin in patients with acute exacerbation of chronic obstructive pulmonary disease receiving ventilatory support. COPD 2014, 11, 171–176.
  21. Cohen, A.T.; Davidson, B.L.; Gallus, A.S.; Lassen, M.R.; Prins, M.H.; Tomkowski, W.; Turpie, A.G.; Egberts, J.F.; Lensing, A.W.; ARTEMIS Investigators. Efficacy and safety of fondaparinux for the prevention of venous thromboembolism in older acute medical patients: Randomised placebo controlled trial. BMJ 2006, 332, 325–329.
  22. Al-Ani, F.; Chehade, S.; Lazo-Langner, A. Thrombosis risk associated with COVID-19 infection. A scoping review. Thromb. Res. 2020, 192, 152–160.
  23. Testa, S.; Prandoni, P.; Paoletti, O.; Morandini, R.; Tala, M.; Dellanoce, C.; Giorgi-Pierfranceschi, M.; Betti, M.; Danz, G.B.; Pan, A.; et al. Direct oral anticoagulant plasma levels’ striking increase in severe COVID-19 respiratory syndrome patients treated with antiviral agents: The Cremona experience. J. Thromb. Haemost. 2020, 18, 1320–1323.
  24. University of Liverpool. COVID-19 Drug Interactions. Available online: (accessed on 23 April 2020).
  25. Marietta, M.; Ageno, W.; Artoni, A.; De Candia, E.; Gresele, P.; Marchetti, M.; Marcucci, R.; Tripodi, A. COVID-19 and haemostasis: A position paper from Italian society on thrombosis and haemostasis (SISET). Blood Transfus. 2020, 18, 167–169.
  26. Young, E. The anti-inflammatory effects of heparin and related compounds. Thromb. Res. 2008, 122, 743–752.
  27. Thachil, J. The versatile heparin in COVID-19. J. Thromb. Haemost. 2020, 18, 1020–1022.
  28. Tichelaar, Y.I.; Kluin-Nelemans, H.J.; Meijer, K. Infections and inflammatory diseases as risk factors for venous thrombosis. A systematic review. Thromb. Haemost. 2012, 107, 827–837.
  29. Poterucha, T.J.; Libby, P.; Goldhaber, S.Z. More than an anticoagulant: Do heparins have direct anti-inflammatory effects? Thromb. Haemost. 2017, 117, 437–444.
  30. Atallah, B.; Mallah, S.I.; AlMahmeed, W. Anticoagulation in COVID-19. Eur. Heart J. Cardiovasc. Pharmacother. 2020, 6, 260–261.
  31. Mitchell, W.B. Thromboinflammation in COVID-19 acute lung injury. Paediatr. Respir. Rev. 2020, 35, 20–24.
  32. Mycroft-West, C.; Su, D.; Elli, S.; Guimond, S.; Miller, G.; Turnbull, J.; Yates, E.; Guerrini, M.; Fernig, D.; Lima, M.; et al. The 2019 coronavirus (SARS-CoV-2) surface protein (Spike) S1 receptor binding domain undergoes conformational change upon heparin binding. BioRxiv 2020.
  33. Thomas, W.; Varley, J.; Johnston, A.; Symington, E.; Robinson, M.; Sheares, K.; Lavinio, A.; Besser, M. Thrombotic complications of patients admitted to intensive care with COVID-19 at a teaching hospital in the United Kingdom. Thromb. Res. 2020, 191, 76–77.
  34. Helms, J.; Tacquard, C.; Severac, F.; Leonard-Lorant, I.; Ohana, M.; Delabranche, X.; Merdji, H.; Clere-Jehl, R.; Schenck, M.; Fagot Gandet, F.; et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: A multicenter prospective cohort study. Intensive Care Med. 2020, 46, 1089–1098.
  35. White, D.; MacDonald, S.; Bull, T.; Hayman, M.; de Monteverde-Robb, R.; Sapsford, D.; Lavinio, A.; Varley, J.; Johnston, A.; Besser, M.; et al. Heparin resistance in COVID-19 patients in the intensive care unit. J. Thromb. Thrombolysis 2020, 50, 287–291.
  36. Bulanov, A.Y.; Roitman, E.V. New coronavirus infection, hemostasis and heparin dosing problems: It is important to say now. Tromboz Gemostaz i Reologiya 2020, 2, 11–18. (In Russian)
  37. Cuker, A.; Tseng, E.K.; Nieuwlaat, R.; Angchaisuksiri, P.; Blair, C.; Dane, K.; Davila, J.; DeSancho, M.T.; Diuguid, D.; Griffin, D.O.; et al. American Society of Hematology 2021 guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19. Blood Adv. 2021, 5, 872–888.
  38. McFadyen, J.D.; Stevens, H.; Peter, K. The Emerging Threat of (Micro) Thrombosis in COVID-19 and Its Therapeutic Implications. Circ. Res. 2020, 127, 571–587.
  39. Fox, S.E.; Akmatbekov, A.; Harbert, J.L.; Li, G.; Quincy Brown, J.; Vander Heide, R.S. Pulmonary and cardiac pathology in Covid-19: The first autopsy series from New Orleans. Lancet Respir. Med. 2020, 8, 681–686.
  40. Kow, C.S.; Hasan, S.S. The use of antiplatelet agents for arterial thromboprophylaxis in COVID-19. Rev. Esp. Cardiol. 2021, 74, 114–115.
  41. Ranucci, M.; Ballotta, A.; Di Dedda, U.; Bayshnikova, E.; Dei Poli, M.; Resta, M.; Falco, M.; Albano, G.; Menicanti, L. The procoagulant pattern of patients with COVID-19 acute respiratory distress syndrome. J. Thromb. Haemost. 2020, 18, 1747–1751.
  42. Liu, X.; Li, Z.; Liu, S.; Sun, J.; Chen, Z.; Jiang, M.; Zhang, Q.; Wei, Y.; Wang, X.; Huang, Y.Y.; et al. Potential therapeutic effects of dipyridamole in the severely ill patients with COVID-19. Acta Pharm. Sin. B 2020, 10, 1205–1215.
  43. Panka, B.A.; de Grooth, H.J.; Spoelstra-de Man, A.M.; Looney, M.R.; Tuinman, P.R. Prevention or treatment of ARDS with aspirin: A review of preclinical models and meta-analysis of clinical studies. Shock 2017, 47, 13–21.
  44. Reilly, J.P.; Christie, J.D. Linking genetics to ARDS pathogenesis: The role of the platelet. Chest 2015, 147, 585–586.
  45. Bikdeli, B.; Madhavan, M.V.; Gupta, A.; Jimenez, D.; Burton, J.R.; Der Nigoghossian, C.; Chuich, T.; Nouri, S.N.; Dreyfus, I.; Driggin, E.; et al. Global COVID-19 Thrombosis Collaborative Group. Pharmacological Agents Targeting Thromboinflammation in COVID-19: Review and Implications for Future Research. Thromb. Haemost. 2020, 120, 1004–1024.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 325
Revision: 1 time (View History)
Update Date: 07 May 2021
1000/1000
Video Production Service