Research article Topical Sections

Etiology model for many vaccination adverse reactions, including SARS-CoV-2 spike vaccines

  • Received: 26 May 2022 Revised: 16 September 2022 Accepted: 29 September 2022 Published: 12 October 2022
  • Objective

    Vaccinated individuals (vaccinees) experience no adverse events, mild adverse events, multiple adverse events, or serious adverse events post vaccination. Many of these vaccine adverse events occur with different vaccines with different occurrence frequencies. Many of these adverse events are generally considered as associated with immune responses to the active vaccine components (antigens) and/or to possibly one or more of the vaccine excipients. Most of these vaccine adverse events are self-limiting and resolve within days. The number of vaccine adverse reactions is higher for SARS-CoV-2 spike vaccines than all other vaccines. Can data analysis of vaccine adverse reactions responses provide etiology insights for high reactogenicity vaccines?

    Methods

    The Vaccine Adverse Event Reporting System (VAERS) database was data mined for all vaccine adverse events data by vaccine, age, gender, dose, and day of onset post vaccination. Results for vaccines with the highest number of adverse events were compared.

    Results

    For vaccines and adverse events with the highest numbers of reports, the day of onset approximates a power of two decay pattern for the first three days. The consistency of this pattern for multiple unrelated vaccines narrows possible etiology mechanisms. Many of these adverse event symptoms overlap symptoms associated with elevated histamine levels. Herein, innate immune responses and specifically elevated histamine levels are proposed to be causative for the majority of these adverse events. This hypothesis is based on a model of innate immune responses releasing a surge of inflammatory molecules, including histamine; this surge is hypothesized to exceed the normal histamine tolerance level for vaccinees causing reactogenicity adverse events. Further, these symptoms resolve as histamine levels fall below the vaccinee's tolerance threshold. This model can be evaluated by the detection of elevated histamine levels in vaccinees corresponding to timing of symptoms onset. If confirmed, a direct consequence of this model predicts that specific antihistamine treatments, mast cell stabilizers, and possibly diamine oxidase enzyme may reduce the incidence or severity of adverse events experienced by vaccinees post vaccinations for most or all high reactogenicity vaccines including coronavirus disease 2019 (COVID-19) spike vaccines.

    Conclusions

    The reported onset occurrences of the majority of reported adverse events are consistent with the likely etiology of innate immune responses to vaccinations for vaccines with higher reactogenicity levels. Herein, the hypothesis is proposed that the majority of these adverse events result from a histamine surge that temporarily exceeds the vaccinee's tolerance level.

    Citation: Darrell O. Ricke. Etiology model for many vaccination adverse reactions, including SARS-CoV-2 spike vaccines[J]. AIMS Allergy and Immunology, 2022, 6(4): 200-215. doi: 10.3934/Allergy.2022015

    Related Papers:

  • Objective

    Vaccinated individuals (vaccinees) experience no adverse events, mild adverse events, multiple adverse events, or serious adverse events post vaccination. Many of these vaccine adverse events occur with different vaccines with different occurrence frequencies. Many of these adverse events are generally considered as associated with immune responses to the active vaccine components (antigens) and/or to possibly one or more of the vaccine excipients. Most of these vaccine adverse events are self-limiting and resolve within days. The number of vaccine adverse reactions is higher for SARS-CoV-2 spike vaccines than all other vaccines. Can data analysis of vaccine adverse reactions responses provide etiology insights for high reactogenicity vaccines?

    Methods

    The Vaccine Adverse Event Reporting System (VAERS) database was data mined for all vaccine adverse events data by vaccine, age, gender, dose, and day of onset post vaccination. Results for vaccines with the highest number of adverse events were compared.

    Results

    For vaccines and adverse events with the highest numbers of reports, the day of onset approximates a power of two decay pattern for the first three days. The consistency of this pattern for multiple unrelated vaccines narrows possible etiology mechanisms. Many of these adverse event symptoms overlap symptoms associated with elevated histamine levels. Herein, innate immune responses and specifically elevated histamine levels are proposed to be causative for the majority of these adverse events. This hypothesis is based on a model of innate immune responses releasing a surge of inflammatory molecules, including histamine; this surge is hypothesized to exceed the normal histamine tolerance level for vaccinees causing reactogenicity adverse events. Further, these symptoms resolve as histamine levels fall below the vaccinee's tolerance threshold. This model can be evaluated by the detection of elevated histamine levels in vaccinees corresponding to timing of symptoms onset. If confirmed, a direct consequence of this model predicts that specific antihistamine treatments, mast cell stabilizers, and possibly diamine oxidase enzyme may reduce the incidence or severity of adverse events experienced by vaccinees post vaccinations for most or all high reactogenicity vaccines including coronavirus disease 2019 (COVID-19) spike vaccines.

    Conclusions

    The reported onset occurrences of the majority of reported adverse events are consistent with the likely etiology of innate immune responses to vaccinations for vaccines with higher reactogenicity levels. Herein, the hypothesis is proposed that the majority of these adverse events result from a histamine surge that temporarily exceeds the vaccinee's tolerance level.


    Abbreviations

    COVID-19

    coronavirus disease 2019

    DTAP

    diphtheria, tetanus, and pertussis vaccine

    FLU3

    influenza vaccine

    HEP

    hepatitis vaccine

    HIT

    histamine intolerance

    MMR

    measles, mumps, and rubella vaccine

    PPV

    pneumococcal polysaccharide vaccine

    SARS-CoV-2

    severe acute respiratory syndrome coronavirus 2

    VAERS

    Vaccine Adverse Event Reporting System

    VARCEL

    chickenpox varicella vaccine

    VARZOS

    shingles varicella-zoster virus vaccine

    加载中


    Conflict of interest



    The author declares that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

    [1] Hervé C, Laupèze B, Del Giudice G, et al. (2019) The how's and what's of vaccine reactogenicity. NPJ Vaccines 4: 39. https://doi.org/10.1038/s41541-019-0132-6
    [2] Forsyth JR (1967) An assessment of oil adjuvant and aqueous influenza vaccines. I. Reactions to the vaccines. J Hyg (Lond) 65: 485-495. https://doi.org/10.1017/S0022172400046027
    [3] Bruusgaard-Mouritsen MA, Johansen JD, Garvey LH (2021) Clinical manifestations and impact on daily life of allergy to polyethylene glycol (PEG) in ten patients. Clin Exp Allergy 51: 463-470. https://doi.org/10.1111/cea.13822
    [4] Rubing SS (1946) An allergic reaction following typhus-fever vaccine and yellow-fever vaccine due to egg yolk sensitivity. J Allergy 17: 21-23. https://doi.org/10.1016/0021-8707(46)90017-2
    [5] Kyriakidis NC, López-Cortés A, González EV, et al. (2021) SARS-CoV-2 vaccines strategies: a comprehensive review of phase 3 candidates. NPJ Vaccines 6: 28. https://doi.org/10.1038/s41541-021-00292-w
    [6] VAERSVaccine Adverse Event Reporting System, U.S. Department of Health & Human Services (2021). Available from: https:/vaers.hhs.gov/data/datasets.html.
    [7] Ricke DO VAERS-Tools (2022). Available from: https://github.com/doricke/VAERS-Tools/.
    [8] Comas-Basté O, Sánchez-Pérez S, Veciana-Nogués MT, et al. (2020) Histamine intolerance: The current state of the art. Biomolecules 10: 1181. https://doi.org/10.3390/biom10081181
    [9] Kovacova-Hanuskova E, Buday T, Gavliakova S, et al. (2015) Histamine, histamine intoxication and intolerance. Allergol Immunopathol 43: 498-506. https://doi.org/10.1016/j.aller.2015.05.001
    [10] Mali S, Jambure R (2012) Anaphyllaxis management: Current concepts. Anesth Essays Res 6: 115-123. https://doi.org/10.4103/0259-1162.108284
    [11] Schnedl WJ, Lackner S, Enko D, et al. (2019) Evaluation of symptoms and symptom combinations in histamine intolerance. Intest Res 17: 427-433. https://doi.org/10.5217/ir.2018.00152
    [12] Shim WS, Oh U (2008) Histamine-induced itch and its relationship with pain. Mol Pain 4: 1-6. https://doi.org/10.1186/1744-8069-4-29
    [13] Lundius EG, Sanchez-Alavez M, Ghochani Y, et al. (2010) Histamine influences body temperature by acting at H1 and H3 receptors on distinct populations of preoptic neurons. J Neurosci 30: 4369. https://doi.org/10.1523/JNEUROSCI.0378-10.2010
    [14] Krantz MS, Kwah JH, Stone CA, et al. (2021) Safety evaluation of the second dose of messenger RNA COVID-19 vaccines in patients with immediate reactions to the first dose. JAMA Intern Med 181: 1530-1533. https://doi.org/10.1001/jamainternmed.2021.3779
    [15] Maintz L, Novak N (2007) Histamine and histamine intolerance. Am J Clin Nutr 85: 1185-1196. https://doi.org/10.1093/ajcn/85.5.1185
    [16] Hrubisko M, Danis R, Huorka M, et al. (2021) Histamine intolerance—The more we know the less we know. A review. Nutrients 13: 2228. https://doi.org/10.3390/nu13072228
    [17] Riley JF (1963) Functional significance of histamine and heparin in tissue mast cells. Ann N Y Acad Sci 103: 151-163. https://doi.org/10.1111/j.1749-6632.1963.tb53695.x
    [18] Graham HT, Lowry OH, Wahl N, et al. (1955) Mast cells as sources of tissue histamine. J Exp Med 102: 307-318. https://doi.org/10.1084/jem.102.3.307
    [19] Parsons ME, Ganellin CR (2006) Histamine and its receptors. Br J Pharmacol 147: S127-S135. https://doi.org/10.1038/sj.bjp.0706440
    [20] Rosa AC, Fantozzi R (2013) The role of histamine in neurogenic inflammation. Br J Pharmacol 170: 38-45. https://doi.org/10.1111/bph.12266
    [21] Izquierdo-Casas J, Comas-Basté O, Latorre-Moratalla ML, et al. (2018) Low serum diamine oxidase (DAO) activity levels in patients with migraine. J Physiol Biochem 74: 93-99. https://doi.org/10.1007/s13105-017-0571-3
    [22] Shavit R, Maoz-Segal R, Iancovici-Kidon M, et al. (2021) Prevalence of allergic reactions after Pfizer-BioNTech COVID-19 vaccination among adults with high allergy risk. JAMA Netw Open 4: e2122255-e2122255. https://doi.org/10.1001/jamanetworkopen.2021.22255
    [23] Sen DK, Arora S, Gupta S, et al. (1974) Studies of adrenergic mechanisms inrelation to histamine sensitivity in children immunized with Bordetella pertussis vaccine. J Allergy Clin Immunol 54: 25-31. https://doi.org/10.1016/S0091-6749(74)80005-9
    [24] Bilò MB, Martini M, Tontini C, et al. (2019) Idiopathic anaphylaxis. Clin Exp Allergy 49: 942-952. https://doi.org/10.1111/cea.13402
    [25] Malone RW, Tisdall P, Fremont-Smith P, et al. (2021) COVID-19: famotidine, histamine, mast cells, and mechanisms. Front Pharmacol 12: 216. https://doi.org/10.3389/fphar.2021.633680
    [26] Tomera KM, Malone, Robert W, Kittah JK (2020) Brief report: Rapid clinical recovery from severe COVID-19 with high dose famotidine and high dose celecoxib adjuvant therapy. Enliven: Pharmacovigil Drug Saf 6: 1-5. https://doi.org/10.20944/preprints202008.0519.v1
    [27] Mather JF, Seip RL, McKay RG (2020) Impact of famotidine use on clinical outcomes of hospitalized patients with COVID-19. Am J Gastroenterol 115: 1617-1623. https://doi.org/10.14309/ajg.0000000000000832
    [28] Sethia R, Prasad M, Mahapatra SJ, et al. (2020) Efficacy of famotidine for COVID-19: A systematic review and meta-analysis. medRxiv . https://doi.org/10.1101/2020.09.28.20203463
    [29] Blanco JIM, Bonilla JAA, Homma S, et al. (2021) Antihistamines and azithromycin as a treatment for COVID-19 on primary health care—A retrospective observational study in elderly patients. Pulm Pharmacol Ther 67: 101989-101989. https://doi.org/10.1016/j.pupt.2021.101989
    [30] Hogan II RB, Hogan III RB, Cannon T, et al. (2020) Dual-histamine receptor blockade with cetirizine-famotidine reduces pulmonary symptoms in COVID-19 patients. Pulm Pharmacol Ther 63: 101942. https://doi.org/10.1016/j.pupt.2020.101942
  • allergy-06-04-015-s001.xlsx
  • Reader Comments
  • © 2022 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1111) PDF downloads(139) Cited by(0)

Article outline

Figures and Tables

Figures(2)  /  Tables(3)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog