@article{MTMT:34754128, title = {mRNA-LNP COVID-19 Vaccine Lipids Induce Complement Activation and Production of Proinflammatory Cytokines: Mechanisms, Effects of Complement Inhibitors, and Relevance to Adverse Reactions}, url = {https://m2.mtmt.hu/api/publication/34754128}, author = {Bakos, Tamás and Mészáros, Tamás and Kozma, Gergely Tibor and Berényi, Petra and Facskó, Réka and Farkas, Henriette and Dézsi, László and Heirman, Carlo and de Koker, Stefaan and Schiffelers, Raymond and Glatter, Kathryn Anne and Radovits, Tamás and Szénási, Gábor and Szebeni, János}, doi = {10.3390/ijms25073595}, journal-iso = {INT J MOL SCI}, journal = {INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES}, volume = {25}, unique-id = {34754128}, issn = {1661-6596}, abstract = {A small fraction of people vaccinated with mRNA–lipid nanoparticle (mRNA-LNP)-based COVID-19 vaccines display acute or subacute inflammatory symptoms whose mechanism has not been clarified to date. To better understand the molecular mechanism of these adverse events (AEs), here, we analyzed in vitro the vaccine-induced induction and interrelations of the following two major inflammatory processes: complement (C) activation and release of proinflammatory cytokines. Incubation of Pfizer-BioNTech’s Comirnaty and Moderna’s Spikevax with 75% human serum led to significant increases in C5a, sC5b-9, and Bb but not C4d, indicating C activation mainly via the alternative pathway. Control PEGylated liposomes (Doxebo) also induced C activation, but, on a weight basis, it was ~5 times less effective than that of Comirnaty. Viral or synthetic naked mRNAs had no C-activating effects. In peripheral blood mononuclear cell (PBMC) cultures supplemented with 20% autologous serum, besides C activation, Comirnaty induced the secretion of proinflammatory cytokines in the following order: IL-1α < IFN-γ < IL-1β < TNF-α < IL-6 < IL-8. Heat-inactivation of C in serum prevented a rise in IL-1α, IL-1β, and TNF-α, suggesting C-dependence of these cytokines’ induction, although the C5 blocker Soliris and C1 inhibitor Berinert, which effectively inhibited C activation in both systems, did not suppress the release of any cytokines. These findings suggest that the inflammatory AEs of mRNA-LNP vaccines are due, at least in part, to stimulation of both arms of the innate immune system, whereupon C activation may be causally involved in the induction of some, but not all, inflammatory cytokines. Thus, the pharmacological attenuation of inflammatory AEs may not be achieved via monotherapy with the tested C inhibitors; efficacy may require combination therapy with different C inhibitors and/or other anti-inflammatory agents.}, year = {2024}, eissn = {1422-0067}, orcid-numbers = {Bakos, Tamás/0000-0002-0569-8343; Facskó, Réka/0000-0002-5611-0597; Farkas, Henriette/0000-0003-2929-1721; Dézsi, László/0000-0002-4190-9793; Szénási, Gábor/0000-0002-7350-6091} } @{MTMT:34754067, title = {Evaluation of the Acute Anaphylactoid Reactogenicity of Nanoparticle-Containing Medicines and Vaccines Using the Porcine CARPA Model}, url = {https://m2.mtmt.hu/api/publication/34754067}, author = {Szebeni, János}, booktitle = {Characterization of Nanoparticles Intended for Drug Delivery}, doi = {10.1007/978-1-0716-3786-9_23}, unique-id = {34754067}, year = {2024}, pages = {229-243} } @misc{MTMT:34521339, title = {mRNA-LNP COVID-19 vaccine lipids induce low level complement activation and production of proinflammatory cytokines: Mechanisms, effects of complement inhibitors, and relevance to adverse reactions}, url = {https://m2.mtmt.hu/api/publication/34521339}, author = {Bakos, Tamás and Mészáros, Tamás and Kozma, Gergely Tibor and Petra, Berényi and Facskó, Réka and Henriette, Farkas and Dézsi, László and Carlo, Heirman and Stefaan, de Koker and Raymond, Schiffelers and Kathryn, Anne Glatter and Radovits, Tamás and Szénási, Gábor and Szebeni, János}, unique-id = {34521339}, abstract = {Messenger RNA-containing lipid nanoparticles (mRNA-LNPs) enabled widespread COVID-19 vaccination with a small fraction of vaccine recipients displaying acute or sub-acute inflammatory symptoms. The molecular mechanism of these adverse events (AEs) remains undetermined. Here we report that the mRNA-LNP vaccine, Comirnaty, triggers low-level complement (C) activation and production of inflammatory cytokines, which may be key underlying processes of inflammatory AEs. In serum, Comirnaty and the control PEGylated liposome (Doxebo) caused different rises of C split products, C5a, sC5b-9, Bb and C4d, indicating stimulation of the classical pathway of C activation mainly by the liposomes, while a stronger stimulation of the alternative pathway was equal with the vaccine and the liposomes. Spikevax had similar C activation as Comirnaty, but viral or synthetic mRNAs had no such effect. In autologous serum-supplemented peripheral blood mononuclear cell (PBMC) cultures, Comirnaty caused increases in the levels of sC5b-9 and proinflammatory cytokines in the following order: IL-1α < IFN-γ < IL-1β < TNF-α < IL-6 < IL-8, whereas heatinactivation of serum prevented the rises of IL-1α, IL-1β, and TNF-α. Clinical C inhibitors, Soliris and Berinert, suppressed vaccine-induced C activation in serum but did not affect cytokine production when applied individually. These findings suggest that the PEGylated lipid coating of mRNA-LNP nanoparticles can trigger C activation mainly via the alternative pathway, which may be causally related to the induction of some, but not all inflammatory cytokines. While innate immune stimulation is essential for the vaccine’s efficacy, concurrent production of C- and PBMC-derived inflammatory mediators may contribute to some of the AEs. Pharmacological attenuation of harmful cytokine production using C inhibitors likely requires blocking the C cascade at multiple points.}, year = {2024}, pages = {&}, orcid-numbers = {Bakos, Tamás/0000-0002-0569-8343; Facskó, Réka/0000-0002-5611-0597; Dézsi, László/0000-0002-4190-9793; Szénási, Gábor/0000-0002-7350-6091} } @misc{MTMT:34747635, title = {COMPARISON OF LIPOSOMAL DRUG-INDUCED BLOOD PRESSURE CHANGES IN MICE AND RATS}, url = {https://m2.mtmt.hu/api/publication/34747635}, author = {Szénási, Gábor and Bakos, Tamás and Őrfi, Erik and Mészáros, Tamás and Hricisák, László and Rosivall, László and Hamar, Péter and Benyó, Zoltán and Szebeni, János and Dézsi, László}, unique-id = {34747635}, abstract = {Introduction: Liposomal drugs administered intravenously (i.v.) can induce IgEindependent side effects known as infusion reaction, and also termed as complement activation-related pseudoallergy (CARPA). Aim: We aimed to reveal the basic mechanisms of blood pressure changes after i.v. injection of amphotericin B-containing liposomes (Abelcet, rats: 10 mg/kg; mice: 30 mg/kg). Methods: Male NMRI mice, and wild type or thromboxane prostanoid receptor deficient (TP KO) mice on C57Bl/6 background, as well as male Wistar rats (anesthetized with pentobarbital) were used (n=6-8/group). Mean arterial blood pressure was continuously monitored. Blood was collected at 0, 1, 3 10 and 30 min in rats, and from separate groups of mice at 3-5 min after treatment. Plasma C3a and thromboxane B2 (TXB2) concentrations were assayed using ELISA. Blood count was obtained using a hematology analyzer (Abacus Vet5). Results: Abelcet caused transient hypertension in mice (10-15 min) and transient hypotension in rats (20-40 min). Abelcet resulted in leucopenia and thrombocytopenia, and increased plasma complement C3a and TXB2 concentrations in both species. Complement depletion with cobra venom factor (CVF) lengthened the hypertensive effect in mice and abolished the hypotensive effect in rats. Pretreatment with DF2593A (10 mg/kg, i.v.), a C5a receptor (C5aR) antagonist, lengthened the hypertensive effect of Abelcet in mice and elicited a small decrease in the hypotensive effect in rats. Inhibition of C3a receptors with SB290157 (10 mg/kg) attenuated the hypertension in mice and enhanced the hypotension in rats, but both effects were rather small. Macrophage depletion with clodronate liposomes in mice lengthened the hypertensive effect similarly to CVF. Pre-treatment with GdCl3 to inhibit macrophages slightly attenuated the hypotensive effect of Abelcet in rats. Inhibition of mast cell activation by cromolyn or C48/80, decreased the hypotensive response in rats, but induced delayed hypotension in mice, respectively. Inhibition of platelet activation using eptifibatide, a platelet glycoprotein IIb/IIIa (GPIIb/IIIa) receptor inhibitor, lengthened the hypertensive effect in mice, but hardly affected the responses in rats. Abelcet did not change blood pressure in TP KO mice and led to a delayed hypertension after pretreatment with CVF. Conclusion: Our results suggest that complement activation has a small contribution to liposomal drug-induced hypotension, and both macrophages and mast cells contribute to the release of vasoactive mediators in rats. The early hypertensive effect of TXA2 release in mice wasindependent from complement activation, and wasreversed with some delay mainly by the activation of C5aR. Both macrophages and platelets are substantially implicated in the latter effect.}, year = {2023}, pages = {73-73}, orcid-numbers = {Szénási, Gábor/0000-0002-7350-6091; Bakos, Tamás/0000-0002-0569-8343; Őrfi, Erik/0000-0002-2190-0708; Hricisák, László/0000-0001-8320-2166; Rosivall, László/0000-0002-9809-3879; Hamar, Péter/0000-0002-1095-3564; Benyó, Zoltán/0000-0001-6015-0359; Dézsi, László/0000-0002-4190-9793} } @article{MTMT:34095902, title = {Eculizumab suppresses zymosan-induced release of inflammatory cytokines IL-1α, IL-1β, IFN-γ and IL-2 in autologous serum-substituted PBMC cultures: Relevance to cytokine storm in Covid-19}, url = {https://m2.mtmt.hu/api/publication/34095902}, author = {Bakos, Tamás and Kozma, Gergely Tibor and Szebeni, János and Szénási, Gábor}, doi = {10.1016/j.biopha.2023.115294}, journal-iso = {BIOMED PHARMACOTHER}, journal = {BIOMEDICINE & PHARMACOTHERAPY}, volume = {166}, unique-id = {34095902}, issn = {0753-3322}, year = {2023}, eissn = {1950-6007}, orcid-numbers = {Bakos, Tamás/0000-0002-0569-8343; Szénási, Gábor/0000-0002-7350-6091} } @article{MTMT:34058342, title = {Insights into the Structure of Comirnaty Covid-19 Vaccine: A Theory on Soft, Partially Bilayer-Covered Nanoparticles with Hydrogen Bond-Stabilized mRNA–Lipid Complexes}, url = {https://m2.mtmt.hu/api/publication/34058342}, author = {Szebeni, János and Kiss, Bálint and Bozó, Tamás and Turjeman, Keren and Levi-Kalisman, Yael and Barenholz, Yechezkel and Kellermayer, Miklós}, doi = {10.1021/acsnano.2c11904}, journal-iso = {ACS NANO}, journal = {ACS NANO}, volume = {17}, unique-id = {34058342}, issn = {1936-0851}, year = {2023}, eissn = {1936-086X}, pages = {13147-13157}, orcid-numbers = {Kiss, Bálint/0000-0002-1595-0426; Bozó, Tamás/0000-0002-2643-0661; Levi-Kalisman, Yael/0000-0002-2764-2738; Kellermayer, Miklós/0000-0002-5553-6553} } @article{MTMT:34039197, title = {Role of anti-polyethylene glycol (PEG) antibodies in the allergic reactions to PEG-containing Covid-19 vaccines: Evidence for immunogenicity of PEG}, url = {https://m2.mtmt.hu/api/publication/34039197}, author = {Kozma, Gergely Tibor and Mészáros, Tamás and Berényi, Petra and Facskó, Réka and Patkó, Zsófia Panna and Oláh, Csaba Zsolt and Nagy, Adrienne and Fülöp, Tamás Gyula and Glatter, Kathryn Anne and Radovits, Tamás and Merkely, Béla Péter and Szebeni, János}, doi = {10.1016/j.vaccine.2023.06.009}, journal-iso = {VACCINE}, journal = {VACCINE}, volume = {41}, unique-id = {34039197}, issn = {0264-410X}, year = {2023}, eissn = {1873-2518}, pages = {4561-4570}, orcid-numbers = {Facskó, Réka/0000-0002-5611-0597; Merkely, Béla Péter/0000-0001-6514-0723} } @misc{MTMT:33993634, title = {The COVID-19 mRNA vaccine Comirnaty induces anaphylactic shock in an anti-PEG hyperimmune large animal model: Role of complement activation in cardiovascular, hematological and inflammatory mediator changes}, url = {https://m2.mtmt.hu/api/publication/33993634}, author = {Barta, András Bálint and Radovits, Tamás and Dobos, Attila Balázs and Kozma, Gergely Tibor and Mészáros, Tamás and Berényi, Petra and Facskó, Réka and Fülöp, Tamas Gyula and Merkely, Béla and Szebeni, János}, unique-id = {33993634}, year = {2023}, pages = {541479} } @article{MTMT:33856424, title = {Comparative in vitro and in vivo Evaluation of Different Iron Oxide-Based Contrast Agents to Promote Clinical Translation in Compliance with Patient Safety}, url = {https://m2.mtmt.hu/api/publication/33856424}, author = {Unterweger, Harald and Janko, Christina and Folk, Tamara and Cicha, Iwona and Stelczerné Kovács, Noémi and Gyebnár, Gyula and Horváth, Ildikó and Máthé, Domokos and Zheng, Kang H. and Coolen, Bram F. and Stroes, Erik and Szebeni, János and Alexiou, Christoph and Dézsi, László and Lyer, Stefan}, doi = {10.2147/IJN.S402320}, journal-iso = {INT J NANOMED}, journal = {INTERNATIONAL JOURNAL OF NANOMEDICINE}, volume = {18}, unique-id = {33856424}, issn = {1176-9114}, abstract = {Introduction: One of the major challenges in the clinical translation of nanoparticles is the development of formulations combining favorable efficacy and optimal safety. In the past, iron oxide nanoparticles have been introduced as an alternative for gadolinium-containing contrast agents; however, candidates available at the time were not free from adverse effects.Methods: Following the development of a potent iron oxide-based contrast agent SPIONDex, we now performed a systematic comparison of this formulation with the conventional contrast agent ferucarbotran and with ferumoxytol, taking into consideration their physicochemical characteristics, bio-and hemocompatibility in vitro and in vivo, as well as their liver imaging properties in rats.Results: The results demonstrated superior in vitro cyto-, hemo-and immunocompatibility of SPIONDex in comparison to the other two formulations. Intravenous administration of ferucarbotran or ferumoxytol induced strong complement activation-related pseu-doallergy in pigs. In contrast, SPIONDex did not elicit any hypersensitivity reactions in the experimental animals. In a rat model, comparable liver imaging properties, but a faster clearance was demonstrated for SPIONDex.Conclusion: The results indicate that SPIONDex possess an exceptional safety compared to the other two formulations, making them a promising candidate for further clinical translation.}, keywords = {COMPLEMENT ACTIVATION; Magnetic Resonance Imaging; NANOPARTICLES; MRI; CARPA; Nanomedicine}, year = {2023}, eissn = {1178-2013}, pages = {2071-2086}, orcid-numbers = {Unterweger, Harald/0000-0002-8335-0692; Janko, Christina/0000-0001-5705-6329; Cicha, Iwona/0000-0002-7399-5307; Dézsi, László/0000-0002-4190-9793} } @article{MTMT:33760499, title = {Covid-19 vaccines elicit effective IgG responses in an elderly thymus cancer patient with chemotherapy}, url = {https://m2.mtmt.hu/api/publication/33760499}, author = {Koller, Ákos and Szebeni, János}, doi = {10.1080/21645515.2023.2188035}, journal-iso = {HUM VACC IMMUNOTHER}, journal = {HUMAN VACCINES & IMMUNOTHERAPEUTICS}, volume = {19}, unique-id = {33760499}, issn = {2164-5515}, year = {2023}, eissn = {2164-554X}, orcid-numbers = {Koller, Ákos/0000-0003-3256-8701} }