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 + 3551 word(s) 3551 2022-03-21 04:34:25 |
2 format correct Meta information modification 3551 2022-03-21 10:43:48 |

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.
Asakura, H. SARS-CoV-2 Vaccination-Associated Coagulopathy. Encyclopedia. Available online: https://encyclopedia.pub/entry/20796 (accessed on 18 May 2024).
Asakura H. SARS-CoV-2 Vaccination-Associated Coagulopathy. Encyclopedia. Available at: https://encyclopedia.pub/entry/20796. Accessed May 18, 2024.
Asakura, Hidesaku. "SARS-CoV-2 Vaccination-Associated Coagulopathy" Encyclopedia, https://encyclopedia.pub/entry/20796 (accessed May 18, 2024).
Asakura, H. (2022, March 21). SARS-CoV-2 Vaccination-Associated Coagulopathy. In Encyclopedia. https://encyclopedia.pub/entry/20796
Asakura, Hidesaku. "SARS-CoV-2 Vaccination-Associated Coagulopathy." Encyclopedia. Web. 21 March, 2022.
SARS-CoV-2 Vaccination-Associated Coagulopathy
Edit

Coronavirus Disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is frequently complicated by thrombosis. In some cases of severe COVID-19, fibrinolysis may be markedly enhanced within a few days, resulting in fatal bleeding. In the treatment of COVID-19, attention should be paid to both coagulation activation and fibrinolytic activation. Various thromboses are known to occur after vaccination with SARS-CoV-2 vaccines. Vaccine-induced immune thrombotic thrombocytopenia (VITT) can occur after adenovirus-vectored vaccination, and is characterized by the detection of anti-platelet factor 4 antibodies by enzyme-linked immunosorbent assay and thrombosis in unusual locations such as cerebral venous sinuses and visceral veins. Treatment comprises high-dose immunoglobulin, argatroban, and fondaparinux. Some VITT cases show marked decreases in fibrinogen and platelets and marked increases in D-dimer, suggesting the presence of enhanced-fibrinolytic-type disseminated intravascular coagulation with a high risk of bleeding. In the treatment of VITT, evaluation of both coagulation activation and fibrinolytic activation is important, adjusting treatments accordingly to improve outcomes.

COVID-19 SARS-CoV-2 vaccine coagulopathy fibrinolysis enhanced-fibrinolytic-type DIC nafamostat

1. Introduction

The novel coronavirus disease 2019 (COVID-19), which was first identified in Wuhan in December 2019, is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of the end of January 2022, approximately two years had passed since the beginning of the COVID-19 pandemic, with the number of infected people worldwide exceeding 3.5 billion and the number of deaths exceeding 550 million at that time. COVID-19 is often complicated by coagulopathy and thrombosis. Though immobility of severely ill patients needs to be considered [1], the incidence of thrombosis appears high among severe COVID-19 cases [2][3].
SARS-CoV-2 vaccines have been developed by many countries to prevent the spread of the infection and reduce the severity of COVID-19. However, it is known that vaccination rarely causes coagulopathy [4].
In both COVID-19-associated coagulopathy and SARS-CoV-2 vaccination-associated coagulopathy, attention to both thrombosis and bleeding is required. While much attention has been given to thrombosis and coagulation activation, almost never have reports discussed bleeding and fibrinolysis. Researchers would therefore like to highlight and discuss these relatively neglected factors. A summarizing figure is shown in (Figure 1).
Figure 1. Summarizing figure. COVID-19 causes coagulation activation depending on its severity. In addition, some cases of severe COVID-19 have markedly increased fibrinolysis. Thrombosis appears as the main symptom when coagulation activation exceeds fibrinolytic activation. Conversely, bleeding appears as the main symptom when fibrinolytic activation exceeds coagulation activation. After SARS-CoV-2 vaccination, VITT rarely occurs. In most cases, coagulation activation exceeds fibrinolytic activation and thrombosis occurs, but in some cases, bleeding appears when fibrinolytic activation exceeds coagulation activation. Abbreviations: COVID-19, coronavirus disease 2019; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; VITT, vaccine-induced immune thrombotic thrombocytopenia.

2. SARS-CoV-2 Vaccination-Associated Coagulopathy

2.1. Safety of Vaccination in Persons with Coagulation Abnormalities

Safe administration of vaccines to patients with coagulation abnormalities involves two considerations: first, whether the physical invasion associated with vaccination will cause bleeding in patients with bleeding tendencies or hemorrhagic diseases; and second, whether patients with hemorrhagic or thrombotic diseases will experience exacerbation or flare-up of the underlying disease.

2.1.1. Preventing Bleeding Due to Vaccination in Patients with Coagulation Abnormalities

Vaccination of patients on antiplatelet and anticoagulant medications (warfarin and DOAC) is certainly possible. However, adequate compression of the vaccination site should be performed after vaccination [5]. If the PT-INR is above the therapeutic range, vaccination should be postponed until the PT-INR is back within the therapeutic range [6], and in DOAC-treated patients, vaccination is best avoided when blood levels of DOACs are high.
Vaccination after prophylaxis treatment is recommended in patients with hemophilia who are receiving regular prophylaxis treatment and in patients with severe von Willebrand disease.
Thrombocytopenia/dysfunctional platelets do not interfere with vaccination, but adequate compression of the vaccination site after vaccination is advisable.
In chronic DIC (e.g., cases with aortic aneurysms and vascular malformations), the invasiveness of vaccination may cause severe bleeding and should be discussed with the attending physician [7].

2.1.2. Vaccination and Thrombotic/Hemorrhagic Disease Exacerbations and Relapses

In patients with immune thrombocytopenia (ITP), the relapse of ITP with vaccination represents a matter of concern. In fact, vaccination of patients with ITP resulted in a ≧20% decrease in platelet count from baseline in about half of patients [8]. However, no reports have suggested that vaccination should be withdrawn in patients with ITP. Risk factors for ITP relapse with vaccination include ongoing treatment, old age [9], and post-splenectomy status or a long history of prior treatment [10]. In terms of platelets, low platelet counts are reportedly associated with a higher risk of relapse [9], and platelet counts often decrease rapidly in patients with normal platelet count [11]. Regardless of the platelet count, attention should be paid to transition of the platelet count in ITP patients. The nadir of platelet counts in ITP patients has been observed around 7–10 days after vaccination [10]. Reports have also recommended measuring platelet counts on days 3–7 after vaccination to confirm the presence or absence of ITP flare-up [12]. Some reports have described ITP cases after vaccination in which the patients were able to receive additional vaccinations without problems [13].
Furthermore, a decrease in platelet count greater than 50% from baseline was observed in 1.0% of healthy controls [9], suggesting that a significant number of patients after vaccination may experience de novo ITP, including cases in which clinical symptoms do not develop and go unnoticed [9].
In patients with paroxysmal nocturnal hemoglobinuria (PNH) or atypical hemolytic uremic syndrome using eculizumab or ravulizumab, vaccination within one week of eculizumab administration and within four weeks of ravulizumab administration is recommended to maintain blood levels of these agents [14]. Thus far, no reports have described an increased risk of relapse of the primary disease after vaccination in specific diseases or a need to refrain from vaccination, and the benefits of vaccination are considered to outweigh the disadvantages. Adequate consultation with the patient and careful observation of changes in coagulation studies are important.

2.2. Novel Clotting Abnormalities after Vaccination

Vaccines are being developed and marketed around the world by a number of manufacturers. The number of vaccinations worldwide has exceeded 10 billion, and more than 50% of the global population had completed the required number of vaccinations as of the end of January 2022 [15]. However, the occurrence of thrombosis and bleeding after vaccination remains problematic.

2.2.1. Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT)

A series of reports have described thrombosis with thrombocytopenia after vaccination with the adenovirus-vectored vaccine (ChAdOx1 nCoV-19) [16][17][18][19]. Similar reports have been made for Ad26.COV2.S, which is an adenovirus vector type vaccine [20][21]. Thrombi occurring after adenovirus-vectored vaccinations have also been characterized as occurring in unusual sites, such as cerebral venous sinuses and visceral veins (e.g., portal vein) [16].
Thrombosis with thrombocytopenia following administration of adenovirus vector vaccines has been referred to as VITT, as well as vaccine-induced prothrombotic immune thrombocytopenia and thrombosis with thrombocytopenia syndrome [22]. VITT is currently the most commonly used term and is considered by the authors as the most appropriate term for describing this condition.

2.2.2. Thrombotic/Bleeding Disorders Other Than VITT

In addition to the adenovirus vector vaccines, ChAdOx1-nCoV-19 and Ad26.COV2.S, the mRNA vaccines, BNT162b2 and mRNA-1273, have been reported to cause various thrombotic/hemorrhagic diseases. Thrombotic/hemorrhagic adverse effects of SARS-CoV-2 vaccines, including those of the adenovirus vector type and the mRNA type, are described below. The diseases that should be differentiated from VITT are shown in Table 1.
Table 1. Diseases that should be differentiated from VITT and their key considerations.
Disease Name Abbreviation Important Clinical and Laboratory Findings
Heparin-induced thrombocytopenia HIT History of exposure to heparin, 4T’s score
Thrombotic microangiopathy TMA Appearance of schizocytes (peripheral blood smear), marked decrease in haptoglobin
Thrombotic thrombocytopenic purpura TTP A type of TMA with markedly reduced ADAMTS13 activity with ADAMTS13 inhibitor
Immune thrombocytopenia ITP Diagnosis of exclusion. Increased megakaryocytes in bone marrow and positive antiplatelet antibodies assist in diagnosis
Antiphospholipid antibody syndrome APS Positive for at least one of the following antibodies: lupus anticoagulant; anticardiolipin antibody; and anti-β2 GPI antibody
Paroxysmal nocturnal hemoglobinuria PNH Hemolysis (normocytic anemia, elevated reticulocyte, elevated indirect bilirubin, elevated LDH, decreased haptoglobin),
presence of PNH type-cells (CD55/59-negative)
Disseminated intravascular coagulation DIC PT, APTT, fibrinogen, FDP, D-dimer, AT, TAT, PIC, plasminogen, α2PI
Abbreviations: VITT, vaccine-induced immune thrombotic thrombocytopenia; TMA, thrombotic microangiopathy; ADAMTS13, a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13; GP, glycoprotein; LDH, lactate dehydrogenase; PT, prothrombin time; APTT, activated partial thromboplastin time; FDP, fibrin/fibrinogen degradation products; AT, antithrombin; TAT, thrombin-antithrombin complex; PIC, plasmin-αplasmin2 inhibitor complex; α2PI, α2 plasmin inhibitor.
(1)
Hemorrhagic disease
Complications such as hematuria, extensive petechial hemorrhage, subarachnoid hemorrhage [23], immune thrombocytopenia [24][25][26][27][28], Evans syndrome [29], acquired hemophilia [30], and factor XIII inhibitor [31] have been reported with the use of adenovirus vector and mRNA vaccines. In particular, when platelet transfusion is performed for hemorrhagic disease, examination to rule out VITT should always be performed, because with the presence of VITT in the background, the underlying condition may be aggravated after platelet transfusion.
Since thrombosis after vaccination initially drew worldwide attention, there is a tendency to focus on thrombosis, but it is important to remember that hemorrhagic side effects may also occur.
(2)
Thrombotic disease
Reports have described carotid artery thrombosis and aortic thrombotic complications [32], deep vein thrombosis [33][34], myocardial infarction [35], adrenal infarction/hemorrhage [36][37], and cerebral infarction/intracranial hemorrhage [38] following adenovirus vector and mRNA vaccinations. Examination to rule out VITT should always be conducted, not only in the case of hemorrhagic events, but also in the case of thrombotic events, due to the concern that a background of VITT may exacerbate the underlying condition with heparin administration.
(3)
Thrombophilia with low platelet count
Numerous reports have described thrombotic thrombocytopenic purpura [39][40][41] and DIC [42]. The exclusion of VITT is still important to determine an accurate treatment strategy.
According to a systematic review of cardiovascular and hematological events after SARS-CoV-2 vaccination, these abnormalities tended to be slightly more frequent among women and young people. Adenovirus-vectored vaccines, ChAdOx1-nCoV-19 and for Ad26.COV2.S, were associated with higher rates of thrombosis and thrombocytopenia, while mRNA vaccines, BNT162b2 and mRNA-1273, were associated with higher rates of cardiac injury in a higher proportion of cases [43]. Although accurate evaluation is difficult due to differences in the number of vaccinations, age group, race, and other factors among manufacturers, certain tendencies in thrombotic/hemorrhagic adverse effects may exist depending on the type of vaccine. Although this remains a subject for further study, knowing which manufacturer’s vaccine a patient received is still important in actual clinical practice.

3.3. Pathophysiology of VITT

VITT is characterized by thrombosis occurring at unusual sites such as cerebral venous sinuses and visceral veins (thrombosis in common sites such as venous thromboembolism is also seen), decreased platelet count, and coagulation abnormalities (e.g., elevated D-dimer) occurring 4–28 days after vaccination. In addition, a positive (usually strongly positive) result from enzyme-linked immunosorbent assay (ELISA) for anti-platelet factor 4 (PF4) antibodies is the basis for a definitive diagnosis, even though heparin is not used. As such, the condition of VITT is considered similar to immune (spontaneous) heparin-induced thrombocytopenia (HIT) [44]. An important point is that measurement of anti-PF4 antibodies by chemiluminescent immunoassay (CLIA) or latex agglutination will result in false-negative results, rendering definitive diagnosis impossible with these two methods [22][45]. Importantly, anti-PF4 antibodies should be measured by ELISA. However, false-negative results with the CLIA and latex agglutination methods can also be used as one basis for VITT diagnosis [46].
Although the frequency of VITT is extremely low, ranging from a few to ten cases per million [47], it is important to note that this condition is often fatal when present.
The mechanism of VITT is thought to be the formation of autoantibodies against the complex of PF4 and free DNA or the coating protein of adenoviruses and binding of the Fc portion of autoantibodies to the Fc receptor on the platelet membrane, which may induce platelet activation and aggregation. In addition, prothrombotic microparticles are released from activated platelets and thrombin production is promoted. PF4 binds to heparin sulfate and chondroitin sulfate on vascular endothelial cells, and binding of autoantibodies to these sites activates endothelial cells. Tissue factor expression from endothelial cells is also thought to activate coagulation [16] (Figure 2). Although 67% (29/43) of healthy controls are positive for anti-PF4 antibodies on day 22 after the first dose of ChAdOx1-nCoV-19, no positive patients showed high antibody titers or developed VITT [48]. When evaluating the anti-PF4 antibody (by ELISA), attention must be paid not only to the positive or negative result, but also to the antibody titer. The prevalence of anti-PF4 antibodies is 1.0–6.6% [49][50] in pre-vaccinated populations. A significant number of post-vaccination anti-PF4 antibody-positive individuals are likely to have had antibodies prior to vaccination, making routine anti-PF4 antibody testing of asymptomatic individuals less useful [51].
Figure 2. Mechanism of VITT development in adenovirus vector vaccines. Immunoglobulin G autoantibodies against the complex of platelet factor 4 and free DNA in the vaccine result in platelet activation and aggregation. In addition, the release of microparticles and activation of coagulation by vascular endothelial cells are thought to be mechanisms underlying thrombus formation in VITT. Abbreviations: VITT, vaccine-induced immune thrombotic thrombocytopenia; PF4, platelet factor 4.
Post-vaccination cerebral venous sinus thrombosis has also been reported with BNT162b2 [52][53][54]. However, in all such cases, platelet counts were normal and anti-PF4 antibodies were not detected, therefore, VITT is not a consideration.
The mRNA vaccines work by delivering mRNA (for the formation of spike proteins) protected by lipid nanoparticles to ribosomes in human cells, causing the production of spike proteins [55]. S protein produced by mRNA vaccine is not exactly the same as the S protein of SARS-CoV-2. For example, uridine of mRNA is converted to pseudouridine [56][57] for more efficiency and safety [58]. The mechanism of mRNA vaccine-induced thrombosis may resemble that of COVID-19 [54], although the details are as yet unknown.

3.4. Diagnosis of VITT

If a patient presents with any of the symptoms listed in Table 2 within 4–28 days after vaccination (day 0 being the date of vaccination), VITT should be suspected because of the possibility of new thrombosis. Coagulation tests for PT, APTT, fibrinogen, and D-dimer (FDP) are mandatory, and PT and APTT alone are insufficient. Since some VITT patients present with DIC and bleeding tendency, measuring TAT (a marker of coagulation activation) and PIC, plasminogen, and α2PI (as markers of fibrinolysis activation) at the same time is advisable [59].
Table 2. Clinical features indicative of VITT.
Clinical Findings
(1) Onset 4–28 days after vaccination (counting the day of vaccination as day 0)
(2) Symptoms suggestive of stroke (unilateral facial palsy, unilateral motor palsy, language disorder, joint parallax, hemispheric neglect, etc.)
(3) Symptoms suggestive of cerebral venous sinus thrombosis (persistent headache, visual disturbance, seizure, nausea and vomiting, psychiatric symptoms, etc.)
(4) Symptoms suggestive of visceral vein thrombosis (persistent abdominal pain, nausea and vomiting, etc.)
(5) Symptoms suggestive of deep vein thrombosis or pulmonary thromboembolism (pain and swelling in lower limbs, chest and back pain, shortness of breath, etc.)
(6) Hemorrhagic tendencies such as hemorrhagic infarction, petechial hemorrhage, and mottled hemorrhage can also occur.
Abbreviations: VITT, vaccine-induced immune thrombotic thrombocytopenia. (Modified from References [60][61][62]).
As mentioned above, measurement of anti-PF4 antibodies by ELISA is of paramount importance, as almost all cases of VITT are positive and show a high titer. However, if VITT is strongly suspected, waiting for the test results may take too long, and initiation of empirical treatment is advisable.
A marked increase in D-dimer to more than four times the upper limit of normal alongside a decreased platelet count is highly suggestive of VITT, but a marked increase in D-dimer is not essential for diagnosis. This is because some VITT patients show only mild elevations in D-dimer.
Another report proposed a “pre-VITT syndrome” in which severe headache at 5–20 days after adenovirus-vectored vaccination should be considered a sign of cerebral venous sinus thrombosis [63]. In fact, although no thrombosis was found on imaging, VITT was suspected early based on clinical symptoms and blood test findings (such as thrombocytopenia and elevated D-dimer), and treatment for VITT was started early, resulting in discharge from hospital in less than one week [64][65]. Notably, VITT is known to occur after administration of adenovirus vector-type COVID-19 vaccines. However, on day 10 after inoculation with the Gardasil 9 vaccine against human papillomavirus (inactivated vaccine), thrombocytopenia similar to that in VITT, venous thrombosis, elevated D-dimer, and the presence of anti-PF4-polyanion complex antibody have been reported [66]. Similar pathologies may have been observed with other vaccinations, but have simply gone unnoticed until now.
Initiating treatment is important when VITT is suspected, as the mortality rate for VITT was an extremely high 50% when the disease was first recognized, although this has now reportedly improved to 22% and even 5% [60]. This is probably due to increased awareness of the disease and earlier detection. The FAPIC score has been devised as a prognostic score looking at fibrinogen, age, platelet count, presence of intracerebral hemorrhage, and presence of cerebral venous sinus thrombosis [67]: fibrinogen < 150 mg/dL; age ≤ 60 years; platelets < 25,000/μL, intracerebral hemorrhage, and cerebral venous sinus thrombosis all represent prognostic factors for poor clinical outcome. This is a simple scoring system, but does not address treatment options for each stratified risk level. In addition, fibrinogen and platelets are suspected to be involved in enhanced-fibrinolytic-type DIC, as described below.

3.5. Treatment of VITT (Attention to Fibrinolytic Pathophysiology)

The treatment strategy is based on the treatment of HIT, and includes discontinuation of heparin (although heparin may reportedly be used if HIT can be reliably excluded [60]), high-dose immunoglobulin therapy, anticoagulants other than heparin (argatroban, fondaparinux, DOACs, etc.), and steroids have all been mentioned [68][69].
However, in some VITT patients, in addition to a marked increase in D-dimer, marked decreases in platelet count and fibrinogen have been observed (Figure 3) [16][70], suggesting a change in coagulation markers as in enhanced-fibrinolytic-type DIC [71]. If such patients are reflexively treated with anticoagulant therapy, major bleeding may result, and rigorous evaluation of coagulation and fibrinolytic markers is thus necessary. In fact, a clinical trial of argatroban for DIC was conducted in Japan more than 30 years ago, but was discontinued after the occurrence of bleeding side effects in many patients. In retrospect, researchers believe that argatroban was more likely to cause bleeding in patients with enhanced-fibrinolytic-type DIC, and should not be prescribed without caution in patients with enhanced-fibrinolytic-type DIC. The characteristics of suppressed-fibrinolytic-type DIC, enhanced-fibrinolytic-type DIC, and VITT are summarized in Table 3.
Figure 3. Relationship between VITT and fibrinolytic pathophysiology. Researchers plotted fibrinogen on the horizontal axis and D-dimer on the vertical axis for 30 definitive VITT cases (using data from Reference [67]) in which both fibrinogen and D-dimer values were known. The lower the level of fibrinogen, the higher the level of D-dimer. In particular, patients with blue dotted squares showed a decrease in fibrinogen and a marked increase in D-dimer, suggesting the complication of enhanced-fibrinolytic-type DIC (a subject for further investigation). If anticoagulants are administered to the same intensity as in other cases, the risk of bleeding is considered high. To confirm the diagnosis of enhanced-fibrinolytic-type DIC, measurement of not only the coagulation activation marker TAT, but also the fibrinolysis activation marker PIC is essential. Patients with markedly decreased α2PI are at high risk of bleeding. Importantly, in enhanced-fibrinolytic-type DIC, the increase in FDP is more prominent than the increase in D-dimer (discrepancy between FDP and D-dimer). Antithrombin, as a coagulation inhibitor, does not often decrease, even in patients with a marked decrease in α2PI, a fibrinolytic inhibitor (except in patients with reduced hepatic reserve). Abbreviations: VITT, vaccine-induced immune thrombotic thrombocytopenia; DIC, disseminated intravascular coagulation; TAT, thrombin–antithrombin complex; PIC, plasmin–α2 plasmin inhibitor complex; α2 PI, α2 plasmin inhibitor; FDP, fibrin/fibrinogen degradation products.
Table 3. Laboratory findings in typical cases of suppressed/enhanced-fibrinolytic-type DIC and VITT.
Disease Suppressed-Fibrinolytic-Type DIC Enhanced-Fibrinolytic-Type DIC VITT
Underlying disease/cause Severe sepsis APL, aortic aneurysm, prostate cancer, etc. Adenovirus vector type
vaccination
Pathophysiology Activation of coagulation and mild fibrinolysis activation Activation of coagulation and enhanced fibrinolysis Antibodies against PF4 are mediated
platelet and coagulation activation
Main symptom Organ damage Bleeding Headache, abdominal pain, etc.
Examination findings Platelet count Decreased Decreased Decreased
PT Prolonged Normal to prolonged Normal to prolonged *
APTT Prolonged Slightly shortened to prolonged Normal to prolonged *
Fibrinogen Normal to elevated Decreased Significantly reduced to normal
D-dimer Increased Increased Increased
FDP Increased Markedly increased Increased—markedly increased *
TAT Increased Increased Increased *
PIC Slightly increased Markedly increased Increased—markedly increased *
Medical treatment Anticoagulant therapy Anticoagulant therapy ± antifibrinolytic therapy Anticoagulant therapy other than heparin, high-dose immunoglobulin therapy, etc.
* Author’s guess due to insufficient information. Abbreviations: DIC, disseminated intravascular coagulation; VITT, vaccine-induced immune thrombotic thrombocytopenia; APL, acute promyelocytic leukemia; PF4, platelet factor 4; PT, prothrombin time; APTT, activated partial thromboplastin time; FDP, fibrin/fibrinogen degradation products; TAT, thrombin–antithrombin complex; PIC, plasmin–α2 plasmin inhibitor complex.
Tranexamic acid has also been reported to save lives [72], but the use of tranexamic acid in VITT should be considered with care, given the risk of exacerbating thrombosis. Anti-PF4 antibodies persist for at least four months after the end of VITT treatment [73], but advantages [73] and disadvantages [74] are seen with respect to VITT flare-ups, so careful follow-up may be necessary.
In addition, no relapse of VITT occurred after a second additional dose of vaccine was administered to 40 patients who had developed VITT after the first dose. In many cases, mRNA-type vaccines, BNT162b2 and mRNA-1273, had been administered as the second dose, suggesting that immune responses to spike proteins are not involved in the pathogenesis of VITT. From the perspective of protection against infection, a second vaccination should be administered even after the onset of VITT [75].
Cerebral venous sinus thrombosis occurring after vaccination with BNT162b2 (but not after vaccination with ChAdOx1-nCoV-19) did not show a decreased platelet count or anti-PF4 antibodies, was not VITT, and was treated with heparins [53]. Which manufacturer’s vaccine was administered is an important piece of information for determining the course of treatment.

References

  1. Geerts, W.; Selby, R. Prevention of Venous Thromboembolism in the ICU. Chest 2003, 124, 357S–363S.
  2. Horiuchi, H.; Morishita, E.; Urano, T.; Yokoyama, K. The Questionnaire-survey Joint Team on The COVID-19-related thrombosis COVID-19-Related Thrombosis in Japan: Final Report of a Questionnaire-Based Survey in 2020. J. Atheroscler. Thromb. 2021, 28, 406–416.
  3. Kerbikov, O.; Orekhov, P.; Borskaya, E.; Nosenko, N. High incidence of venous thrombosis in patients with moderate-to-severe COVID-19. Int. J. Hematol. 2021, 113, 344–347.
  4. Hunter, P.R. Thrombosis after COVID-19 vaccination. BMJ 2021, 373, n958.
  5. Perrin, G.; Le Beller, C.; Darnige, L.; Khider, L.; Smadja, D.M.; Louet, A.L.-L.; Planquette, B.; Lebeaux, D.; Sanchez, O.; Sabatier, B.; et al. Intramuscular Vaccination in Adults with Therapeutic Anticoagulation in the Era of COVID-19 Vaccines Outbreak: A Practical Review. TH Open 2021, 5, e166–e170.
  6. Ogawa, H.; Asakura, H. Consideration of Tranexamic Acid Administration to COVID-19 Patients. Physiol. Rev. 2020, 100, 1595–1596.
  7. Yamada, S.; Asakura, H. Therapeutic Strategies for Disseminated Intravascular Coagulation Associated with Aortic Aneurysm. Int. J. Mol. Sci. 2022, 23, 2396.
  8. Jiang, D.; Portuguese, A.J.; Weatherford, A.; Garcia, D.; Gernsheimer, T. Platelet trends after Covid-19 vaccination in patients with chronic or persistent immune thrombocytopenia. Am. J. Hematol. 2021, 96, E472–E474.
  9. Visser, C.; Swinkels, M.; van Werkhoven, E.D.; Croles, F.N.N.; Noordzij-Nooteboom, H.S.; Eefting, M.; Last-Koopmans, S.M.; Idink, C.; Westerweel, P.E.; Santbergen, B.; et al. COVID-19 vaccination in patients with immune thrombocytopenia. Blood Adv. 2022, 6, 1637–1644.
  10. Lee, E.-J.; Beltrami-Moreira, M.; Al-Samkari, H.; Cuker, A.; DiRaimo, J.; Gernsheimer, T.; Kruse, A.; Kessler, C.M.; Kruse, C.; Leavitt, A.D.; et al. SARS-CoV-2 vaccination and ITP in patients with de novo or preexisting ITP. Blood 2022, 139, 1564–1574.
  11. Crickx, E.; Moulis, G.; Ebbo, M.; Terriou, L.; Briantais, A.; Languille, L.; Limal, N.; Guillet, S.; Michel, M.; Mahevas, M.; et al. Safety of anti-SARS-CoV-2 vaccination for patients with immune thrombocytopenia. Br. J. Haematol. 2021, 195, 703–705.
  12. Kuter, D.J. Exacerbation of immune thrombocytopenia following COVID-19 vaccination. Br. J. Haematol. 2021, 195, 365–370.
  13. Immune thrombocytopenia associated with COVID-19 mRNA vaccine tozinameran—a clinical case and global pharmacovigilance data. Swiss Med Wkly. 2021, 151, w30084.
  14. Gerber, G.F.; Yuan, X.; Yu, J.; Cher, B.A.Y.; Braunstein, E.M.; Chaturvedi, S.; Brodsky, R.A. COVID-19 vaccines induce severe hemolysis in paroxysmal nocturnal hemoglobinuria. Blood 2021, 137, 3670–3673.
  15. Our World in Data. Coronavirus (COVID-19) Vaccinations. Available online: https://ourworldindata.org/covid-vaccinations (accessed on 28 February 2022).
  16. Greinacher, A.; Thiele, T.; Warkentin, T.E.; Weisser, K.; Kyrle, P.A.; Eichinger, S. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N. Engl. J. Med. 2021, 384, 2092–2101.
  17. Schultz, N.H.; Sørvoll, I.H.; Michelsen, A.E.; Munthe, L.A.; Lund-Johansen, F.; Ahlen, M.T.; Wiedmann, M.; Aamodt, A.-H.; Skattør, T.H.; Tjønnfjord, G.E.; et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N. Engl. J. Med. 2021, 384, 2124–2130.
  18. Scully, M.; Singh, D.; Lown, R.; Poles, A.; Solomon, T.; Levi, M.; Goldblatt, D.; Kotoucek, P.; Thomas, W.; Lester, W. Pathologic Antibodies to Platelet Factor 4 after ChAdOx1 nCoV-19 Vaccination. N. Engl. J. Med. 2021, 384, 2202–2211.
  19. Tiede, A.; Sachs, U.J.; Czwalinna, A.; Werwitzke, S.; Bikker, R.; Krauss, J.K.; Donnerstag, F.G.; Weißenborn, K.; Höglinger, G.U.; Maasoumy, B.; et al. Prothrombotic immune thrombocytopenia after COVID-19 vaccination. Blood 2021, 138, 350–353.
  20. See, I.; Su, J.R.; Lale, A.; Woo, E.J.; Guh, A.Y.; Shimabukuro, T.T.; Streiff, M.B.; Rao, A.K.; Wheeler, A.P.; Beavers, S.F.; et al. US Case Reports of Cerebral Venous Sinus Thrombosis with Thrombocytopenia After Ad26.COV2.S Vaccination, March 2 to April 21, 2021. JAMA 2021, 325, 2448–2456.
  21. Muir, K.-L.; Kallam, A.; Koepsell, S.A.; Gundabolu, K. Thrombotic Thrombocytopenia after Ad26.COV2.S Vaccination. N. Engl. J. Med. 2021, 384, 1964–1965.
  22. Favaloro, E.J. Laboratory testing for suspected COVID-19 vaccine–induced (immune) thrombotic thrombocytopenia. Int. J. Lab. Hematol. 2021, 43, 559–570.
  23. Waraich, A.; Williams, G. Haematuria, a widespread petechial rash, and headaches following the Oxford AstraZeneca ChAdOx1 nCoV-19 Vaccination. BMJ Case Rep. 2021, 14, e245440.
  24. Paulsen, F.-O.; Schaefers, C.; Langer, F.; Frenzel, C.; Wenzel, U.; Hengel, F.E.; Bokemeyer, C.; Seidel, C. Immune Thrombocytopenic Purpura after vaccination with COVID-19 Vaccine (ChAdOx1 nCov-19). Blood 2021, 138, 996–999.
  25. Choi, P.Y.-I.; Hsu, D.; Tran, H.A.; Tan, C.W.; Enjeti, A.; Chen, V.M.Y.; Chong, B.H.; Curnow, J.; Pepperell, D.; Bird, R. Immune thrombocytopenia following vaccination during the COVID-19 pandemic. Haematologica 2021.
  26. Tarawneh, O.; Tarawneh, H. Immune thrombocytopenia in a 22-year-old post COVID-19 vaccine. Am. J. Hematol. 2021, 96, E133–E134.
  27. Nakamura, T.; Morodomi, Y.; Kanaji, S.; Okamura, T.; Nagafuji, K.; Kanaji, T. Detection of anti-GPIbα autoantibodies in a case of immune thrombocytopenia following COVID-19 vaccination. Thromb. Res. 2021, 209, 80–83.
  28. Okada, Y.; Sakai, R.; Sato-Fitoussi, M.; Nodera, M.; Yoshinaga, S.; Shibata, A.; Kurasawa, T.; Kondo, T.; Amano, K. Potential Triggers for Thrombocytopenia and/or Hemorrhage by the BNT162b2 Vaccine, Pfizer-BioNTech. Front. Med. 2021, 8, 751598.
  29. Hidaka, D.; Ogasawara, R.; Sugimura, S.; Fujii, F.; Kojima, K.; Nagai, J.; Ebata, K.; Okada, K.; Kobayashi, N.; Ogasawara, M.; et al. New-onset Evans syndrome associated with systemic lupus erythematosus after BNT162b2 mRNA COVID-19 vaccination. Int. J. Hematol. 2021, 115, 424–427.
  30. Radwi, M.; Farsi, S. A case report of acquired hemophilia following COVID-19 vaccine. J. Thromb. Haemost. 2021, 19, 1515–1518.
  31. Shimoyama, S.; Kanisawa, Y.; Ono, K.; Souri, M.; Ichinose, A. First and fatal case of autoimmune acquired factor XIII /13 deficiency after COVID -19/ SARS-CoV -2 vaccination. Am. J. Hematol. 2021, 97, 243–245.
  32. Goereci, Y.; Kleineberg, N.N.; Madlener, M.; Neuschmelting, H.; Fink, G.R.; Warnke, C.; Stetefeld, H. Successful treatment of thromboses of major arteries after ChAdOx1 nCov-19 vaccination. Neurol. Res. Pract. 2021, 3, 1–4.
  33. Andraska, E.A.; Kulkarni, R.; Chaudhary, M.; Sachdev, U. Three cases of acute venous thromboembolism in females after vaccination for coronavirus disease 2019. J. Vasc. Surg. Venous Lymphat. Disord. 2021, 10, 14–17.
  34. Carli, G.; Nichele, I.; Ruggeri, M.; Barra, S.; Tosetto, A. Deep vein thrombosis (DVT) occurring shortly after the second dose of mRNA SARS-CoV-2 vaccine. Intern. Emerg. Med. 2021, 16, 803–804.
  35. D’Agostino, V.; Caranci, F.; Negro, A.; Piscitelli, V.; Tuccillo, B.; Fasano, F.; Sirabella, G.; Marano, I.; Granata, V.; Grassi, R.; et al. A Rare Case of Cerebral Venous Thrombosis and Disseminated Intravascular Coagulation Temporally Associated to the COVID-19 Vaccine Administration. J. Pers. Med. 2021, 11, 285.
  36. Varona, J.F.; García-Isidro, M.; Moeinvaziri, M.; Ramos-López, M.; Fernández-Domínguez, M. Pri-mary adrenal insufficiency associated with Oxford-AstraZeneca ChAdOx1 nCoV-19 vaccine-induced immune thrombotic thrombocytopenia (VITT). Eur J Intern. Med. 2021, 91, 90–92.
  37. Blauenfeldt, R.A.; Kristensen, S.R.; Ernstsen, S.L.; Kristensen, C.C.H.; Simonsen, C.Z.; Hvas, A. Thrombocytopenia with acute ischemic stroke and bleeding in a patient newly vaccinated with an adenoviral vector-based COVID-19 vaccine. J. Thromb. Haemost. 2021, 19, 1771–1775.
  38. Shimazawa, R.; Ikeda, M. Potential adverse events in Japanese women who received tozinameran (BNT162b2, Pfizer-BioNTech). J. Pharm. Policy Pract. 2021, 14, 46.
  39. Kirpalani, A.; Garabon, J.; Amos, K.; Patel, S.; Sharma, A.P.; Ganesan, S.L.; Barton, M.; Cacciotti, C.; Leppington, S.; Bakovic, L.; et al. Thrombotic thrombocytopenic purpura temporally associated with BNT162b2 vaccination in an adolescent successfully treated with caplacizumab. Br. J. Haematol. 2021, 196, e11–e14.
  40. de Bruijn, S.; Maes, M.; De Waele, L.; Vanhoorelbeke, K.; Gadisseur, A. First report of a de novo iTTP episode associated with an mRNA-based anti-COVID-19 vaccination. J. Thromb. Haemost. 2021, 19, 2014–2018.
  41. Maayan, H.; Kirgner, I.; Gutwein, O.; Herzog-Tzarfati, K.; Rahimi-Levene, N.; Koren-Michowitz, M.; Blickstein, D. Acquired thrombotic thrombocytopenic purpura: A rare disease associated with BNT162b2 vaccine. J. Thromb. Haemost. 2021, 19, 2314–2317.
  42. Aladdin, Y.; Algahtani, H.; Shirah, B. Vaccine-Induced Immune Thrombotic Thrombocytopenia with Disseminated Intravascular Coagulation and Death following the ChAdOx1 nCoV-19 Vaccine. J. Stroke Cerebrovasc. Dis. 2021, 30, 105938.
  43. Al-Ali, D.; Elshafeey, A.; Mushannen, M.; Kawas, H.; Shafiq, A.; Mhaimeed, N.; Mhaimeed, O.; Mhaimeed, N.; Zeghlache, R.; Salameh, M.; et al. Cardiovascular and haematological events post COVID-19 vaccination: A systematic review. J. Cell. Mol. Med. 2021, 26, 636–653.
  44. Sharifian-Dorche, M.; Bahmanyar, M.; Sharifian-Dorche, A.; Mohammadi, P.; Nomovi, M.; Mowla, A. Vaccine-induced immune thrombotic thrombocytopenia and cerebral venous sinus thrombosis post COVID-19 vaccination; a systematic review. J. Neurol. Sci. 2021, 428, 117607.
  45. Vayne, C.; Rollin, J.; Gruel, Y.; Pouplard, C.; Galinat, H.; Huet, O.; Mémier, V.; Geeraerts, T.; Marlu, R.; Pernod, G.; et al. PF4 Immunoassays in Vaccine-Induced Thrombotic Thrombocytopenia. N. Engl. J. Med. 2021, 385, 376–378.
  46. Tiede, A.; Althaus, K.; Sachs, U.J.; Cooper, N.; Czwalinna, A.; Müller, J.; Pötzsch, B. PF4-Dependent Immunoassays in Patients with Vaccine-Induced Immune Thrombotic Thrombocytopenia: Results of an Interlaboratory Comparison. Thromb. Haemost. 2021, 121, 1622–1627.
  47. Cines, D.B.; Bussel, J.B. SARS-CoV-2 Vaccine-Induced Immune Thrombotic Thrombocytopenia. N. Engl. J. Med. 2021, 384, 2254–2256.
  48. Terpos, E.; Politou, M.; Ntanasis-Stathopoulos, I.; Karalis, V.; Merkouri, E.; Fotiou, D.; Gavriatopoulou, M.; Malandrakis, P.; Kastritis, E.; Trougakos, I.; et al. High Prevalence of Anti-PF4 Antibodies Following ChAdOx1 nCov-19 (AZD1222) Vaccination Even in the Absence of Thrombotic Events. Vaccines 2021, 9, 712.
  49. Hursting, M.J.; Pai, P.J.; McCracken, J.E.; Hwang, F.; Suvarna, S.; Lokhnygina, Y.; Bandarenko, N.; Arepally, G.M. Platelet Factor 4/Heparin Antibodies in Blood Bank Donors. Am. J. Clin. Pathol. 2010, 134, 774–780.
  50. Arepally, G.M.; Hursting, M.J. Platelet factor 4/heparin antibody (IgG/M/A) in healthy subjects: A literature analysis of commercial immunoassay results. J. Thromb. Thrombolysis 2008, 26, 55–61.
  51. Barefah, A.S.; Radhwi, O.O.; Alamri, S.S.; Alahwal, H.M.; Denetiu, I.; Almohammadi, A.T.; Bahashwan, S.M.; Qari, M.H.; Algaissi, A.; Alamer, E.; et al. Low clinical utility of testing for anti-platelet factor 4 in asymptomatic individuals after ChAdOx1 nCoV-19 vaccine. Int. J. Lab. Hematol. 2021.
  52. Yamaguchi, Y.; Kimihira, L.; Nagasawa, H.; Seo, K.; Wada, M. Cerebral Venous Sinus Thrombosis After BNT162b2 mRNA COVID-19 Vaccination. Cureus 2021, 13, e18775.
  53. Fan, B.E.; Shen, J.Y.; Lim, X.R.; Tu, T.M.; Chang, C.C.R.; Khin, H.S.W.; Koh, J.S.; Rao, J.P.; Lau, S.L.; Tan, G.B.; et al. Cerebral venous thrombosis post BNT162b2 mRNA SARS-CoV-2 vaccination: A black swan event. Am. J. Hematol. 2021, 96, E357–E361.
  54. Dias, L.; Soares-Dos-Reis, R.; Meira, J.; Ferrão, D.; Soares, P.R.; Pastor, A.; Gama, G.; Fonseca, L.; Fagundes, V.; Carvalho, M. Cerebral Venous Thrombosis after BNT162b2 mRNA SARS-CoV-2 vaccine. J. Stroke Cerebrovasc. Dis. 2021, 30, 105906.
  55. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N. Engl. J. Med. 2020, 383, 2603–2615.
  56. Mulligan, M.J.; Lyke, K.E.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Raabe, V.; Bailey, R.; Swanson, K.A.; et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature 2020, 586, 589–593.
  57. Corbett, K.S.; Edwards, D.K.; Leist, S.R.; Abiona, O.M.; Boyoglu-Barnum, S.; Gillespie, R.A.; Himansu, S.; Schäfer, A.; Ziwawo, C.T.; DiPiazza, A.T.; et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 2020, 586, 567–571.
  58. Karikó, K.; Buckstein, M.; Ni, H.; Weissman, D. Suppression of RNA Recognition by Toll-like Receptors: The Impact of Nucleoside Modification and the Evolutionary Origin of RNA. Immunity 2005, 23, 165–175.
  59. Greinacher, A.; Langer, F.; Makris, M.; Pai, M.; Pavord, S.; Tran, H.; Warkentin, T.E. Vaccine-induced immune thrombotic thrombocytopenia (VITT)—Update on diagnosis and management considering different resources: Response to Comment from Yamada et al. J. Thromb. Haemost. 2022, 20, 542–543.
  60. Greinacher, A.; Langer, F.; Makris, M.; Pai, M.; Pavord, S.; Tran, H.; Warkentin, T.E. Vaccine-induced immune thrombotic thrombocytopenia (VITT): Update on diagnosis and management considering different resources. J. Thromb. Haemost. 2022, 20, 149–156.
  61. Nazy, I.; Sachs, U.J.; Arnold, D.M.; McKenzie, S.E.; Choi, P.; Althaus, K.; Ahlen, M.T.; Sharma, R.; Grace, R.F.; Bakchoul, T. Recommendations for the clinical and laboratory diagnosis of VITT against COVID-19: Communication from the ISTH SSC Subcommittee on Platelet Immunology. J. Thromb. Haemost. 2021, 19, 1585–1588.
  62. Bussel, J.B.; Connors, J.M.; Cines, D.B.; Cines, D.B.; Dunbar, C.E.; Michaelis, L.C.; Kreuziger, L.B.; Lee, A.Y.Y.; Pabinger-Fasching, I. Throm-Bosis with Thrombocytopenia Syndrome (Also Termed Vaccine Induced Thrombotic Thrombocytopenia). Version 1.7. Available online: https://www.hematology.org/covid-19/vaccine-induced-immune-thrombotic-thrombocytopenialast (accessed on 17 December 2021).
  63. Salih, F.; Schönborn, L.; Kohler, S.; Franke, C.; Möckel, M.; Dörner, T.; Bauknecht, H.C.; Pille, C.; Graw, J.A.; Alonso, A.; et al. Vaccine-Induced Thrombocytopenia with Severe Headache. N. Engl. J. Med. 2021, 385, 2103–2105.
  64. Khuhapinant, A.; Rungjirajittranon, T.; Suwanawiboon, B.; Chinthammitr, Y.; Ruchutrakool, T. Successful venous thromboprophylaxis in a patient with vaccine-induced immune thrombotic thrombocytopenia (VITT): A case report of the first reported case in Thailand. Thromb. J. 2021, 19, 1–5.
  65. Kennedy, V.E.; Wong, C.C.; Hong, J.M.; Peng, T.A.; Brondfield, S.; Reilly, L.M.; Cornett, P.; Leavitt, A.D. VITT following Ad26.COV2.S vaccination presenting without radiographically demonstrable thrombosis. Blood Adv. 2021, 5, 4662–4665.
  66. Johansen, S.; Lægreid, I.J.; Ernstsen, S.L.; Azrakhsh, N.A.; Kittang, A.O.; Lindås, R.; Gjertsen, B.T.; Vetti, N.; Mørtberg, T.V.; Sørvoll, I.H.; et al. Thrombosis and thrombocytopenia after HPV vaccination. J. Thromb. Haemost. 2021, 20, 700–704.
  67. Hwang, J.; Park, S.H.; Lee, S.W.; Lee, S.B.; Lee, M.H.; Jeong, G.H.; Kim, M.S.; Kim, J.Y.; Koyanagi, A.; Jacob, L.; et al. Predictors of mortality in thrombotic thrombocytopenia after adenoviral COVID-19 vaccination: The FAPIC score. Eur. Heart J. 2021, 42, 4053–4063.
  68. Klok, F.A.; Pai, M.; Huisman, M.V.; Makris, M. Vaccine-induced immune thrombotic thrombocytopenia. Lancet Haematol. 2021, 9, e73–e80.
  69. Arepally, G.M.; Ortel, T.L. Vaccine-induced immune thrombotic thrombocytopenia: What we know and do not know. Blood 2021, 138, 293–298.
  70. Thaler, J.; Ay, C.; Gleixner, K.V.; Hauswirth, A.W.; Cacioppo, F.; Grafeneder, J.; Quehenberger, P.; Pabinger, I.; Knöbl, P. Successful treatment of vaccine-induced prothrombotic immune thrombocytopenia (VIPIT). J. Thromb. Haemost. 2021, 19, 1819–1822.
  71. Asakura, H. Classifying types of disseminated intravascular coagulation: Clinical and animal models. J. Intensiv. Care 2014, 2, 20.
  72. Wilting, F.N.; Kotsopoulos, A.M.; Platteel, A.C.; van Oers, J.A. Intracerebral Hemorrhage and Thrombocytopenia After AstraZeneca COVID-19 Vaccine: Clinical and Diagnostic Challenges of Vaccine-Induced Thrombotic Thrombocytopenia. Cureus 2021, 13, e17637.
  73. Nicolson, P.L.; Montague, S.J.; Smith, C.W.; Lodwick, C.S.; Stoneley, C.; Roberts, M.; Watson, S.P.; Lowe, G.C.; Lester, W.A. Anti-PF4 levels of patients with VITT do not reduce 4 months following AZD1222 vaccination. medRxiv 2021.
  74. Thaler, J.; Jilma, P.; Samadi, N.; Roitner, F.; Mikušková, E.; Kudrnovsky-Moser, S.; Rettl, J.; Preiss, R.; Quehenberger, P.; Pabinger, I.; et al. Long-term follow-up after successful treatment of vaccine-induced prothrombotic immune thrombocytopenia. Thromb. Res. 2021, 207, 126–130.
  75. Lacy, J.; Pavord, S.; Brown, K.E. VITT and Second Doses of Covid-19 Vaccine. N. Engl. J. Med. 2022, 386, 95.
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: 879
Entry Collection: COVID-19
Revisions: 2 times (View History)
Update Date: 21 Mar 2022
1000/1000