Next Article in Journal
Efficacy and Safety of Native and Recombinant Zona Pellucida Immunocontraceptive Vaccines Formulated with Non-Freund’s Adjuvants in Donkeys
Previous Article in Journal
Immunogenicity and Efficacy of Monovalent and Bivalent Formulations of a Virus-Like Particle Vaccine against SARS-CoV-2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

A Possibility of Vasospastic Angina after mRNA COVID-19 Vaccination

Department of Cardiovascular Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi Meguro-ku, Tokyo 153-8515, Japan
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(12), 1998; https://doi.org/10.3390/vaccines10121998
Submission received: 24 October 2022 / Revised: 13 November 2022 / Accepted: 23 November 2022 / Published: 24 November 2022
(This article belongs to the Section COVID-19 Vaccines and Vaccination)

Abstract

:
We report a case of vasospastic angina (VSA) following COVID-19 mRNA vaccination. Despite the widespread occurrence of myocarditis, there have been few reports of post-vaccinal VSA. A 41-year-old male patient was referred for chest pain at rest following mRNA vaccination; he had never experienced chest pain prior to vaccination. He was diagnosed by an acetylcholine (Ach) provocation test that showed multivessel vasospasm. After the initiation of treatment with a calcium channel blocker and nitrate, no further exacerbation of chest pain was observed. To our knowledge, this constitutes the first reported case of VSA proven by Ach provocation test after COVID-19 vaccination. The vaccination may increase coronary artery spasticity. VSA should be ruled out in post-vaccine new onset resting chest pain.

1. Introduction

Messenger RNA (mRNA)-based vaccination against COVID-19 is used to combat the spread of the SARS-CoV-2 virus. Cardiovascular diseases, including acute coronary syndrome (ACS) and myocarditis, are the most serious side effects of COVID-19 mRNA vaccination [1,2]. Plausible causes of ACS following vaccination are assumed to be angiotensin-converting enzyme 2 (ACE2) downregulation [3,4,5], inflammatory cytokine [6,7,8,9,10,11], and allergic response [12,13]. Although ACS has been reported many times, vasospastic angina (VSA) is rarely reported after the COVID-19 mRNA vaccine.

2. Case Presentation

A 41-year-old Japanese male smoker with no medical history received the first dose of the mRNA-BNT162b2 COVID-19 vaccine (Pfizer-BioNTech, Cambridge, MA, USA). One week after vaccination, chest pain and palpitations appeared after drinking alcohol; he had no previous documented chest pain prior to his COVID-19 vaccination. His chest pain repeated exacerbations and remissions at rest and he was admitted to our hospital on the 11th day after vaccination.
On admission, his body temperature was 37.2 °C, blood pressure was 126/80 mmHg, heart rate was 98/min, and oxygen saturation was 98% on room air. When he arrived at our hospital, his chest pain was resolved, and his electrocardiogram (ECG) and transthoracic echocardiogram were normal. His blood test results were within normal limits, including creatine kinase (124 IU/L), high-sensitivity cardiac troponin T (0.003 ng/mL), potassium (3.7 mEq/L), creatinine (0.83 mg/dL), and C-reactive protein (0.04 mg/dL) on admission. After he developed chest pain at rest and after alcohol consumption, we suspected VSA. Emergency coronary angiography revealed no coronary artery stenosis. However, the acetylcholine (Ach) provocation test showed multivessel vasospasm with chest pain (left coronary artery (LCA), segments #7 99% stenosis and #12 total occlusion at 100 µg dose into LCA) with ST elevation in ECG leads I, aVL and large T wave in leads V1–V4 (Figure 1A–E). He was diagnosed with VSA because his findings fulfilled all criteria (i) reproduction of the usual chest pain, (ii) ischemic ECG changes, and (iii) 90% vasoconstriction on angiography.
Therefore, we initiated a calcium channel blocker (CCB) with nitrate and he was discharged the next day. Antibody titers of various viruses, including SARS-CoV-2, were negative. After the initiation of treatment with a CCB and nitrate, no further exacerbation of chest pain was observed.

3. Discussion

To our knowledge, this constitutes the first reported case of VSA proven by Ach provocation test after COVID-19 vaccination. He was diagnosed with VSA because his Ach provocation test findings fulfilled all criteria: (i) reproduction of the usual chest pain, (ii) ischemic ECG changes, and (iii) 90% vasoconstriction on angiography [14]. Additionally, after the initiation of treatment with a CCB and nitrate, no further exacerbation of chest pain was observed.
Chest discomfort is present in 3% of cases after receiving the mRNA vaccine [15]. Myocarditis, acute coronary syndrome, and VSA have been reported as potential causes of chest pain associated with mRNA vaccination against COVID-19 [1,2]. However, reported cases of VSA following the COVID-19 mRNA vaccine are scarce and the true incidence remains unknown. Acute coronary syndromes (ACS) and angina pectoris in the elderly are reported to often present as atypical chest pain and more cardiac failure [16,17]. Therefore, it is important not to rule out ACS or angina even in the presence of atypical chest pain after vaccination in the elderly. Notably, a flowchart of the differential diagnoses of chest discomfort and palpitation after COVID-19 vaccination has been reported [18].
Regarding the relationship between vaccination and VSA, several mechanisms may induce VSA. The spike protein of the SARS-CoV-2 virus or the mRNA component of the vaccine binds to the angiotensin-converting enzyme 2 (ACE2) receptor, which leads to the downregulation of ACE2 (Figure 2) [3,4]. ACE2 converts the potent vasoconstrictor angiotensin II (AngII) to the vasodilator angiotensin 1–7. Hence, the downregulation of ACE2 activates AngII, which leads to vasoconstriction and also produces interleukin (IL)-6 [3,5]. Various inflammatory stimuli, including AngII and IL-6, upregulate the activity of Rho-kinase. The activation of Rho-kinase plays a central role in coronary artery spasms caused by the hypercontraction of vascular smooth muscle cells (Figure 2) [19].
Other plausible mechanisms are the involvement of inflammatory cytokines and allergic reactions. While lipid nanoparticles (LNPs) play an important role as a delivery system for mRNA vaccines, cationic lipids in LNPs themselves reportedly cause inflammatory cytokines such as interleukin (IL)-6 and IL-1β (Figure 2) [7,8,9]. The IL6 genotype has been reported to affect the prevalence of VSA [10]. Four post-mRNA vaccine autopsy cases have recently been reported to express genes that upregulate inflammatory cytokine signaling compared with controls [6]. Inflammatory cytokines inhibit endothelial nitric oxide and increase Rho-kinase activity, causing coronary artery spasms (Figure 2) [11,19].
East Asians are genetically predisposed to VSA since 40% are aldehyde dehydrogenase 2 (ALDH2)*2 carriers with deficient ALDH2 activity [20]. In ALDH2*2 carriers, alcohol intake and tobacco smoking increase toxic aldehydes, which cause endothelial dysfunction (Figure 2). Although we did not examine the ALDH2 genotype of the patient, we speculated that he was an ALDH2*2 carrier since he had alcohol flushing syndrome (AFS). AFS is associated with ALDH2*2 (odds ratio 51.6) [20]. He had never experienced chest pain prior to receiving the COVID-19 mRNA vaccine; therefore, post-vaccinal alcohol consumption may be a factor that triggered VSA.
COVID-19 mRNA vaccine components, including polyethylene glycol, LNPs, etc., may cause an allergic reaction, resulting in a coronary vasospasm called Kounis syndrome [2,12]. Mast cells activated by the vaccine antigen induce histamine with or without immunoglobulin E [12], and histamine reportedly induces coronary vasospasm [13]. β1-adrenoceptors (ARs) are predominantly found in the coronary epithelium, while β2-ARs are found in the microvascular [21]. It has recently been reported that Kounis syndrome is more likely to occur when β2-ARs, but not β1, are blocked [22]. Therefore, patients taking nonselective β-ARs antagonists such as propranolol should be aware of Kounis syndrome following vaccination.
Although this case highly suggested the relationship between COVID-19 mRNA vaccine and VSA, VSA and microvascular dysfunction, not epicardial coronary artery stenosis has also been reported in COVID-19 patients [23,24]. Therefore, the benefit–risk balance should always be considered when deciding whether or not to vaccinate.

4. Conclusions

This case demonstrates a possibility of VSA after COVID-19 mRNA vaccination. VSA should be ruled out in post-vaccine new onset resting chest discomfort and palpitation, especially in cases with repeated exacerbations and remissions.

Author Contributions

Conceptualization, T.A.; investigation, F.N. and S.T.; writing—original draft preparation, T.A.; writing—review and editing, M.M., Y.E. and M.N; visualization, T.A., M.M., S.T., M.W. and T.K.; supervision, M.M. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Written informed consent was obtained from the subject for the publication of this case report.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

VSAvasospastic angina
COVID-19coronavirus disease 2019
mRNAmessenger RNA
ACSacute coronary syndrome
ACE2angiotensin-converting enzyme 2
ECGelectrocardiogram
Achacetylcholine
LCAleft coronary artery
CCBcalcium channel blocker
AngIIangiotensin II
ILinterleukin
LNPslipid nanoparticles
ALDH2aldehyde dehydrogenase 2
AFSalcohol flushing syndrome
ARsadrenoceptors

References

  1. Rosenblum, H.G.; Gee, J.; Liu, R.; Marquez, P.L.; Zhang, B.; Strid, P.; Abara, W.E.; McNeil, M.M.; Myers, T.R.; Hause, A.M.; et al. Safety of mRNA vaccines administered during the initial 6 months of the US COVID-19 vaccination programme: An observational study of reports to the Vaccine Adverse Event Reporting System and v-safe. Lancet Infect. Dis. 2022, 22, 802–812. [Google Scholar] [CrossRef] [PubMed]
  2. Aye, Y.N.; Mai, A.S.; Zhang, A.; Lim, O.Z.H.; Lin, N.; Ng, C.H.; Chan, M.Y.; Yip, J.; Loh, P.H.; Chew, N.W. Acute Myocardial Infarction and Myocarditis following COVID-19 Vaccination. QJM 2021, hcab252. [Google Scholar] [CrossRef] [PubMed]
  3. Kuba, K.; Imai, Y.; Penninger, J.M. Multiple functions of angiotensin-converting enzyme 2 and its relevance in cardiovascular diseases. Circ. J. 2013, 77, 301–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Verdecchia, P.; Cavallini, C.; Spanevello, A.; Angeli, F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur. J. Intern. Med. 2020, 76, 14–20. [Google Scholar] [CrossRef]
  5. Recinos, A., 3rd; LeJeune, W.S.; Sun, H.; Lee, C.Y.; Tieu, B.C.; Lu, M.; Hou, T.; Boldogh, I.; Tilton, R.G.; Brasier, A.R. Angiotensin II induces IL-6 expression and the Jak-STAT3 pathway in aortic adventitia of LDL receptor-deficient mice. Atherosclerosis 2007, 194, 125–133. [Google Scholar] [CrossRef] [Green Version]
  6. Murata, K.; Nakao, N.; Ishiuchi, N.; Fukui, T.; Katsuya, N.; Fukumoto, W.; Oka, H.; Yoshikawa, N.; Nagao, T.; Namera, A.; et al. Four cases of cytokine storm after COVID-19 vaccination: Case report. Front. Immunol. 2022, 13, 967226. [Google Scholar] [CrossRef]
  7. Lonez, C.; Bessodes, M.; Scherman, D.; Vandenbranden, M.; Escriou, V.; Ruysschaert, J.M. Cationic lipid nanocarriers activate Toll-like receptor 2 and NLRP3 inflammasome pathways. Nanomedicine 2014, 10, 775–782. [Google Scholar] [CrossRef]
  8. Ndeupen, S.; Qin, Z.; Jacobsen, S.; Bouteau, A.; Estanbouli, H.; Igyártó, B. The mRNA-LNP platform’s lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. iScience 2021, 24, 103479. [Google Scholar] [CrossRef]
  9. Alameh, M.G.; Tombácz, I.; Bettini, E.; Lederer, K.; Sittplangkoon, C.; Wilmore, J.R.; Gaudette, B.T.; Soliman, O.Y.; Pine, M.; Hicks, P.; et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity 2021, 54, 2877–2892.e7. [Google Scholar] [CrossRef]
  10. Murase, Y.; Yamada, Y.; Hirashiki, A.; Ichihara, S.; Kanda, H.; Watarai, M.; Takatsu, F.; Murohara, T.; Yokota, M. Genetic risk and gene-environment interaction in coronary artery spasm in Japanese men and women. Eur. Heart J. 2004, 25, 970–977. [Google Scholar] [CrossRef]
  11. Kidde, J.; Gorabi, A.M.; Jamialahmadi, T.; Sahebkar, A. COVID-19 Is an Endothelial Disease: Implications of Nitric Oxide. Adv. Exp. Med. Biol. 2021, 1321, 109–113. [Google Scholar] [CrossRef] [PubMed]
  12. Kounis, N.G.; Koniari, I.; de Gregorio, C.; Velissaris, D.; Petalas, K.; Brinia, A.; Assimakopoulos, S.F.; Gogos, C.; Kouni, S.N.; Kounis, G.N.; et al. Allergic Reactions to Current Available COVID-19 Vaccinations: Pathophysiology, Causality, and Therapeutic Considerations. Vaccines 2021, 9, 221. [Google Scholar] [CrossRef] [PubMed]
  13. Ginsburg, R.; Bristow, M.R.; Kantrowitz, N.; Baim, D.S.; Harrison, D.C. Histamine provocation of clinical coronary artery spasm: Implications concerning pathogenesis of variant angina pectoris. Am. Heart J. 1981, 102, 819–822. [Google Scholar] [CrossRef] [PubMed]
  14. Beltrame, J.F.; Crea, F.; Kaski, J.C.; Ogawa, H.; Ong, P.; Sechtem, U.; Shimokawa, H.; Bairey Merz, C.N.; Coronary Vasomotion Disorders International Study Group (COVADIS). International standardization of diagnostic criteria for vasospastic angina. Eur. Heart J. 2017, 38, 2565–2568. [Google Scholar] [CrossRef] [Green Version]
  15. Lee, Y.W.; Lim, S.Y.; Lee, J.H.; Lim, J.S.; Kim, M.; Kwon, S.; Joo, J.; Kwak, S.H.; Kim, E.O.; Jung, J.; et al. Adverse Reactions of the Second Dose of the BNT162b2 mRNA COVID-19 Vaccine in Healthcare Workers in Korea. J. Korean Med. Sci. 2021, 36, e153. [Google Scholar] [CrossRef]
  16. Corsini, F.; Scaglione, A.; Iacomino, M.; Mascia, G.; Melorio, S.; Riccio, C.; Romano, S.; Vetrano, A.; Celardo, S.; Corsini, G.; et al. L’infarto miocardico acuto nell’ultrasettantacinquenne. Studio caso-controllo con una popolazione più giovane e rassegna della letteratura [Acute myocardial infarction in the elderly. A case-control study with a younger population and review of literature]. Monaldi Arch. Chest Dis. 2006, 66, 13–19. (In Italian) [Google Scholar] [CrossRef] [Green Version]
  17. Marchionni, N.; Orso, F. Stable angina in the elderly: Diagnostic and therapeutic approach. J. Cardiovasc. Med. 2018, 19 (Suppl. 1), e84–e87. [Google Scholar] [CrossRef]
  18. Awaya, T.; Moroi, M.; Enomoto, Y.; Kunimasa, T.; Nakamura, M. What Should We Do after the COVID-19 Vaccination? Vaccine-Associated Diseases and Precautionary Measures against Adverse Reactions. Vaccines 2022, 28, 866. [Google Scholar] [CrossRef]
  19. Takahashi, J.; Suda, A.; Nishimiya, K.; Godo, S.; Yasuda, S.; Shimokawa, H. Pathophysiology and Diagnosis of Coronary Functional Abnormalities. Eur. Cardiol. 2021, 16, e30. [Google Scholar] [CrossRef]
  20. Mizuno, Y.; Hokimoto, S.; Harada, E.; Kinoshita, K.; Yoshimura, M.; Yasue, H. Variant Aldehyde Dehydrogenase 2 (ALDH2*2) in East Asians Interactively Exacerbates Tobacco Smoking Risk for Coronary Spasm—Possible Role of Reactive Aldehydes. Circ. J. 2016, 81, 96–102. [Google Scholar] [CrossRef]
  21. Barbato, E.; Wijns, W.; De Bruyne, B.; Mascia, G.; Piscione, F. Attuali concetti sulla fisiopatologia dei recettori beta-adrenergici nelle coronarie e sulle sue applicazioni cliniche [Current concepts of the physiopathology of coronary beta-adrenergic receptors, and their clinical applications]. Recenti Prog. Med. 2005, 96, 411–415. [Google Scholar] [PubMed]
  22. Kuda, Y.; Shibamoto, T.; Yang, W.; Zhang, T.; Tanida, M.; Kurata, Y. Blockade of β2-adrenoceptor, rather than β1-adrenoceptor, deteriorates cardiac anaphylaxis in isolated blood-perfused rat hearts. Cardiol. J. 2017, 24, 403–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Aikawa, T.; Ogino, J.; Oyama-Manabe, N.; Funayama, N. Vasospastic Angina: A Cause of Post-acute COVID-19 Syndrome. Intern. Med. 2022, 61, 2693–2695. [Google Scholar] [CrossRef] [PubMed]
  24. Vallejo Camazón, N.; Teis, A.; Martínez Membrive, M.J.; Llibre, C.; Bayés-Genís, A.; Mateu, L. Long COVID-19 and microvascular disease-related angina. Rev. Esp. Cardiol. Engl. Ed. 2022, 75, 444–446. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A,C): Coronary angiography (CAG) revealed a nonobstructive coronary artery on admission. (B,D): An acetylcholine (Ach) provocation test revealed multivessel vasospasm (left coronary artery (LCA) segments, #7 99% stenosis and #12 total occlusion (yellow arrows) at 100 µg dose into LCA). (E): An electrocardiogram showed ST elevation in leads I, aVL, and large T wave in leads V1–V4 (black arrows) during the Ach provocation test.
Figure 1. (A,C): Coronary angiography (CAG) revealed a nonobstructive coronary artery on admission. (B,D): An acetylcholine (Ach) provocation test revealed multivessel vasospasm (left coronary artery (LCA) segments, #7 99% stenosis and #12 total occlusion (yellow arrows) at 100 µg dose into LCA). (E): An electrocardiogram showed ST elevation in leads I, aVL, and large T wave in leads V1–V4 (black arrows) during the Ach provocation test.
Vaccines 10 01998 g001
Figure 2. The potential mechanisms of vasospastic angina following mRNA vaccination in this case. mRNA, messenger RNA; IL, interleukin; ACE2, angiotensin-converting enzyme 2.
Figure 2. The potential mechanisms of vasospastic angina following mRNA vaccination in this case. mRNA, messenger RNA; IL, interleukin; ACE2, angiotensin-converting enzyme 2.
Vaccines 10 01998 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Awaya, T.; Moroi, M.; Nakamura, F.; Toi, S.; Wakiya, M.; Enomoto, Y.; Kunimasa, T.; Nakamura, M. A Possibility of Vasospastic Angina after mRNA COVID-19 Vaccination. Vaccines 2022, 10, 1998. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10121998

AMA Style

Awaya T, Moroi M, Nakamura F, Toi S, Wakiya M, Enomoto Y, Kunimasa T, Nakamura M. A Possibility of Vasospastic Angina after mRNA COVID-19 Vaccination. Vaccines. 2022; 10(12):1998. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10121998

Chicago/Turabian Style

Awaya, Toru, Masao Moroi, Fuminori Nakamura, Satoru Toi, Momoko Wakiya, Yoshinari Enomoto, Taeko Kunimasa, and Masato Nakamura. 2022. "A Possibility of Vasospastic Angina after mRNA COVID-19 Vaccination" Vaccines 10, no. 12: 1998. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10121998

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop