4.7 Article

Insights in ChAdOx1 nCoV-19 vaccine-induced immune thrombotic thrombocytopenia

Journal

BLOOD
Volume 138, Issue 22, Pages 2256-2268

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2021013231

Keywords

-

Categories

Funding

  1. Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) [374031971, TRR240, 398967434, SFB/TR261, 25440785, SFB877, KFO306, 80750187 SFB841, DFG-FR4239/1-1, INST 2026/13-1 FUGG]
  2. European Union [840189]
  3. German Heart Foundation [F/34/18]
  4. Ministerium fur Wirtschaft, Arbeit und Gesundheit Mecklenburg Vorpommern (project COVIDPROTECT), Leibniz WissenschaftsCampus [ComBioCat W10/2018]
  5. Sudmeyer fund for kidney and vascular research (Sudmeyer-Stiftung fur Nierenund Gefassforschung)
  6. Gerhard Buchtemann fund
  7. Marie Curie Actions (MSCA) [840189] Funding Source: Marie Curie Actions (MSCA)

Ask authors/readers for more resources

The mechanism underlying vaccine-induced immune thrombotic thrombocytopenia (VITT) primarily involves the formation of PF4/vaccine complexes and the proinflammatory environment stimulated by the vaccine, leading to the production of high-titer anti-PF4 antibodies in the body and consequent thrombosis.
SARS-CoV-2 vaccine ChAdOx1 nCoV-19 (AstraZeneca) causes a thromboembolic complication termed vaccine-induced immune thrombotic thrombocytopenia (VITT). Using biophysical techniques, mouse models, and analysis of VITT patient samples, we identified determinants of this vaccine-induced adverse reaction. Super-resolution microscopy visualized vaccine components forming antigenic complexes with platelet factor 4 (PF4) on platelet surfaces to which anti-PF4 antibodies obtained from VITT patients bound. PF4/vaccine complex formation was charge-driven and increased by addition of DNA. Proteomics identified substantial amounts of virus production-derived T-REx HEK293 proteins in the ethylenediaminetetraacetic acid (EDTA)-containing vaccine. Injected vaccine increased vascular leakage in mice, leading to systemic dissemination of vaccine components known to stimulate immune responses. Together, PF4/vaccine complex formation and the vaccine-stimulated proinflammatory milieu trigger a pronounced B-cell response that results in the formation of high-in VITT patients. The resulting high-titer anti-PF4 antibodies potently activated platelets in avidity anti-PF4 antibodies the presence of PF4 or DNA and polyphosphate polyanions. Anti-PF4 VITT patient antibodies also stimulated neutrophils to release neutrophil extracellular traps (NETs) in a platelet PF4-dependent manner. Biomarkers of procoagulant NETs were elevated in VITT patient serum, and NETs were visualized in abundance by immunohistochemistry in cerebral vein thrombi obtained from VITT patients. Together, vaccine-induced PF4/adenovirus aggregates and proinflammatory reactions stimulate pathologic anti-PF4 antibody production that drives thrombosis in VITT. The data support a 2-step mechanism underlying VITT that resembles the pathogenesis of (autoimmune) heparin-induced thrombocytopenia.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available