4,289
Views
0
CrossRef citations to date
0
Altmetric
Coronavirus

Polymyalgia rheumatica and giant cell arteritis following COVID-19 vaccination: Results from a nationwide survey

ORCID Icon, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , & show all
Article: 2334084 | Received 17 Jan 2024, Accepted 19 Mar 2024, Published online: 02 Apr 2024

ABSTRACT

We conducted a national in-depth analysis including pharmacovigilance reports and clinical study to assess the reporting rate (RR) and to determine the clinical profile of polymyalgia rheumatica (PMR) and giant cell arteritis (GCA) in COVID-19-vaccinated individuals. First, based on the French pharmacovigilance database, we estimated the RR of PMR and GCA cases in individuals aged over 50 who developed their initial symptoms within one month of receiving the BNT162b2 mRNA, mRNA-1273, ChAdOx1 nCoV-19, and Ad26.COV2.S vaccines. We then conducted a nationwide survey to gather clinical profiles, therapeutic management, and follow-up data from individuals registered in the pharmacovigilance study. A total of 70 854 684 COVID-19 vaccine doses were administered to 25 260 485 adults, among which, 179 cases of PMR (RR 7. 1 cases/1 000 000 persons) and 54 cases of GCA (RR 2. 1 cases/1 000 000 persons) have been reported. The nationwide survey allowed the characterization of 60 PMR and 35 GCA cases. Median time to the onset of first symptoms was 10 (range 2–30) and 7 (range 2–25) days for PMR and GCA, respectively. Phenotype, GCA-related ischemic complications and -large vessel vasculitis as well as therapeutic management and follow-up seemed similar according to the number of vaccine shots received and when compared to the literature data of unvaccinated population. Although rare, the short time between immunization and the onset of first symptoms of PMR and GCA suggests a temporal association. Physician should be aware of this potential vaccine-related phenomenon.

Introduction

Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are two closely related inflammatory diseases that primarily affect individuals aged 50 years and older. GCA is a systemic large vessel vasculitis (LVV), while PMR is characterized by pain in the shoulders and pelvic girdles.Citation1 There is a 50% overlap in features between GCA and PMR cases and a 20% overlap within PMR cases. The prevalence of GCA varies among ethnic groups, ranging from 1.5 per 100,000 in Japan to 250 per 100,000 in the United Kingdom.Citation2,Citation3 PMR follows a similar distribution pattern as GCA but is 3 times more frequent overall. While the exact triggers of both diseases remain unknown, it is likely that genetic and environmental factors both play a role in their development. The introduction of coronavirus disease 2019 (COVID-19) vaccines represents the most promising opportunity to control the global pandemic. When the vaccination campaign started in Europe, 4 types of vaccines utilizing 2 different immunological processes (mRNA and viral vector vaccines) became available: BNT162b2 mRNA (Pfizer-BioNTech), mRNA-1273 (Moderna), ChAdOx1 nCoV-19 (Oxford-AstraZeneca), and Ad26.COV2.S (Johnson & Johnson/Janssen). Each COVID-19 vaccine employs a harmless version of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein as an antigen to stimulate the production of antibodies targeted against the spike protein, thereby preventing the virus from entering host cells.Citation4 There have been reports of some serious adverse events, like myocarditis following mRNA vaccinations, or thrombosis following non-replicating viral vector vaccinations.Citation5 However, there is limited knowledge regarding cases of PMR and GCA occurring after COVID-19 vaccination.

Some individuals experiencing the onset of PMR and GCA symptoms shortly after receiving COVID-19 vaccination have raised concerns about potential adverse events associated with immunization.Citation6,Citation7 Intriguingly, a recent PMR/GCA-focused pharmacovigilance study utilizing an international database and conducted after the initiation of COVID-19 vaccination campaigns indicated a possible safety signal.Citation8 However, both types of studies faced limitations regarding access to clinical data and follow-up information. To address this context, we initiated a national in-depth analysis including pharmacovigilance reports and clinical study among medical experts of PMR and GCA. First, based on the French pharmacovigilance database, we aimed to estimate the reporting rate (RR) of PMR and GCA cases in individuals aged over 50 who developed their initial symptoms within one month of receiving the BNT162b2 mRNA (Pfizer-BioNTech), mRNA-1273 (Moderna), ChAdOx1 nCoV-19 (Oxford-AstraZeneca), and Ad26.COV2.S (Johnson & Johnson/Janssen) vaccines. In the second part of the study, we conducted a nationwide survey to gather comprehensive clinical profiles, therapeutic management, and follow-up data from individuals registered in the pharmacovigilance database and an overview by worldwide literature.

Patients and methods

Pharmacovigilance study

We conducted a retrospective national population-based pharmacovigilance study of PMR and GCA cases using the well-established French pharmacovigilance system coordinated by the French drug Agency.Citation9 Only French reports of PMR and GCA, who developed their symptoms within one month of receiving COVID-19 vaccination (BNT162b2 mRNA (Pfizer- BioNTech), mRNA-1273 (Moderna),ChAdOx1 nCoV-19 Oxford-AstraZeneca), and Ad26.COV2.S (Johnson & Johnson/Janssen), between December 27th, 2020, and August 30th, 2022 were included in the study. All reports underwent a comprehensive pharmacological, clinical, and biological assessment conducted by a pharmacologist at the Regional Center. The main outcomes were the national RR of PMR and GCA cases per million vaccinated individuals and per million doses administered in adults over 50 years old. To calculate the national RR, we used the total number of COVID-19 vaccinated individuals or the total number of administered doses over the study period as the denominator (available at https://solidarites-sante.gouv.fr/grands-dossiers/vaccin-covid-19/article/le-tableau-de-bord-de-la-vaccination). Secondary outcomes included the RR of PMR and GCA following the first (1st), second (2nd), and third (3rd) injections of COVID-19 vaccines, as well as variations based on the type of COVID-19 vaccine administered.

Nationwide survey

A retrospective nationwide survey was conducted to gather de novo cases of PMR and GCA registered in the pharmacovigilance database. These cases were obtained from the French National Society of Internal Medicine (SNFMI) and the French Study Group for Large Vessel Vasculitis (GEFA). The diagnosis of PMR and GCA was confirmed when patients met the ACR/EULAR classification criteria for PMR and GCA, respectively.Citation10,Citation11 Patients with positive anti- citrullinated protein antibodies (ACPA) were excluded. This study received approval from the Institutional Review Board of the Assistance Publique – Hôpitaux de Marseille (Health Data Hub number F20211207134309) and was conducted in accordance with the Declaration of Helsinki. A standardized electronic form was sent to each physician who declared a patient. Anonymized data were thus collected in a centralized database. We retrieved the following for all of the included patients: demographics, cardiovascular risk factors, type of vaccination and immediate clinical reaction, time to first symptoms, clinical manifestations at diagnosis, laboratory tests (including C-reactive protein (CRP), erythrocyte sedimentation rate, hemoglobin and platelet levels), HLA DRB typing by sequence-specific primers (PCR-SSP), temporal artery biopsy (TAB) status and the results from imaging (including articular echography, temporal arteries echography, computed tomography angiography (CTA) scan and fluorodeoxyglucose positron emission tomography with computed tomography (PET-CT) scan) were reported when available. The outcomes and follow-up data for all the patients were retrieved: clinical status, dose of corticosteroids (CS) per kilogram of body weight and requirement for immunosuppressive drugs (mainly methotrexate (MTX) or tocilizumab (TCZ)) at treatment initiation and at months 3, 6 and 12 (for PMR), at months 3, 6, 12 and 18 (for GCA). We defined relapse as a reoccurrence of symptoms and/or inflammatory parameters on laboratory findings without any other cause identified than PMR or GCA and that required a sustained increase in the CS dose and/or the addition of a CS-sparing agent.

Literature review

We conducted a review of the MEDLINE database (National Library of Medicine, Bethesda, MD) from 2021 to 2023, utilizing and combining the following keywords: PMR, GCA, Horton’s disease, COVID-19 vaccination, BNT162b2 mRNA(Pfizer-BioNTech), mRNA-1273 (Moderna), ChAdOx1 nCoV-19 (Oxford-AstraZeneca), and Ad26.COV2.S (Johnson & Johnson/Janssen) vaccines. Only confirmed cases of PMR and GCA with pertinent clinical data available were included in the analysis.

Statistical analysis

Categorical variables were presented as numbers (%), and quantitative variables were expressed as medians [range]. The three clinical groups, categorized by the number of vaccine shots received (1, 2, or 3), were compared with respect to patient characteristics, biological and imaging parameters, administered treatment, and outcomes using the Kruskal-Wallis test with False Discovery Rate (FDR) correction for multiple comparisons. Statistical analyses were performed using GraphPad Prism 5.0, and p-values <.05 were considered statistically significant.

Results

Pharmacovigilance study

During the study period, a total of 70,854,684 COVID-19 vaccine doses were administered, comprising 50,587,266 doses of BNT162b2 mRNA (Pfizer-BioNTech), 11924,026 doses of mRNA-1273 (Moderna), 7,390,842 doses of ChadOx1 nCoV-19 (Oxford-AstraZeneca), and 952,550 doses of Ad26.COV2.S (Johnson & Johnson/Janssen) to 25,260,485 adults over the age of 50. Pharmacovigilance centers received reports of 179 cases of PMR with a RR of 7.1 cases per 1,000,000 persons and 54 cases of GCA with a RR of 2.1 cases per 1,000,000 persons. The details of these cases are summarized in .

Table 1. Reporting rate of PMR and GCA in patients following COVID-19 vaccination.

Considering all cases of PMR and GCA, regardless of the type of vaccine administered, a national RR of 2.53 (range 0–3.76) and 0.76 (range 0–1.5) per 1,000,000 vaccine doses was observed, respectively. The highest RR for PMR (3.76) and GCA (1.5) occurred after the 1st dose of the ChadOx1 nCOV-19 (Oxford-AstraZeneca) vaccine. For mRNA vaccines, the highest RR was consistently observed after the 2nd shot for PMR cases (BNT162b2 mRNA (Pfizer-BioNTech): 3.64; mRNA-1273 (Moderna): 2.98) and after the 1st dose for GCA cases (BNT162b2 mRNA (Pfizer-BioNTech): 1.03; mRNA-1273 (Moderna): 0.73). Regarding non-mRNA vaccines, the highest RR occurred after the 1st shot for PMR (ChadOx1 nCoV-19 (Oxford-AstraZeneca): 3.76; Ad26.COV2.S (Johnson & Johnson/Janssen): 2.11) and for GCA cases (ChadOx1 nCoV-19 (Oxford-AstraZeneca): 0.88). No GCA was reported after Ad26.COV2.S (Johnson & Johnson/Janssen) vaccine.

Nationwide survey

PMR

The nationwide survey facilitated the identification of 60 PMR cases (22 (36.6%) occurring after the 1st shot, 28 (46.6%) after the 2nd shot, and 10 (16.8%) after the 3rd shot), median age of 71 years (range 52–94), 26 women (43.3%), whose characteristics are summarized in . The primary cardiovascular risk factor identified was hypertension, mentioned in 30 (50%) cases. Among the patients, 54 (90.1%) were vaccinated with mRNA vaccines, while 6 (9.9%) received viral vector vaccines. The median time to the onset of the first symptoms was 10 (range 2–30) days. All patients experienced girdle pain. Distal arthralgia was reported by 24 (40%) patients. Systemic signs were reported by 49 (81.6%) patients. Unusual symptoms were reported by 6 (10%) patients, including neck pain (n = 3), distal edema (n = 1), hemicrania (negative TAB)) (n = 1), and exudative pleuritis (n = 1). All patients exhibited a biological inflammatory syndrome, with a median CRP level of 55 mg/L (range 14–224). Among the 32 (53.3%) patients who underwent a PET-CT scan, 29 (90.6%) exhibited the typical hypermetabolism associated with PMR. No significant differences in terms of phenotype, biological markers, or PET-CT imaging were detected between the subgroups identified based on the number of vaccine shots received.

Table 2. Characteristics of the whole cohort of PMR patients following COVID-19 vaccination.

GCA

The nationwide survey led to the identification of 35 GCA cases (14 (40%) occurring after the 1st shot, 13 (37.1%) after the 2nd shot, and 8 (22.9%) after the 3rd shot), median age of 72 years (range 50–99), 22 women (62.8%) whose characteristics are summarized in . The primary cardiovascular risk factors identified included hypertension and overweight, both mentioned in 13 (37.1%) patients. Among the patients, 31 (88.5%) were vaccinated with mRNA vaccines, while 4 (11.5%) received viral vector vaccines. The median time to the onset of the first symptoms was 7 (range 2–25) days.

Table 3. Characteristics of the whole cohort of GCA patients following COVID-19 vaccination.

Cephalic signs were reported by 30 (85.7%) patients, among which 25 (71.4%) had headache, 18 (51.4%) had scalp tenderness, 13 (37.1%) had jaw claudication, and 21 (60%) had a sensitive temporal artery. Ophthalmologic signs were mentioned by 7 (20%) patients, including binocular diplopia (n = 3) and unilateral arteritic anterior ischemic optic neuropathy (AAION) (n = 4), with one case associated with central artery occlusion.

Associated PMR was mentioned in 17 (48.5%) patients. Systemic signs were reported by 30 (85.7%) patients. Other symptoms like pleuro-pericarditis and cough were reported in one and 3 patients, respectively. All patients, except one, exhibited a biological inflammatory syndrome, with a median CRP level of 84 mg/L (range 5–286). TAB was performed in 30 (85.7%) patients, and 19 (63.3%) of them confirmed the diagnosis of GCA. LVV was detected in 18 out of 32 (56.2%) patients, with involvement of the aorta and carotid arteries being predominant (aorta: 11 (34.3%), carotid arteries: 5 (15.6%)). No significant differences in terms of phenotype, biological markers, or PET-CT imaging were detected among the subgroups identified based on the number of vaccine shots received.

HLA study

HLADRB1 typing data were available for 15 patients with PMR and 11 patients with GCA, and these findings are summarized in . HLADRB1*04, a genetic marker associated with an increased susceptibility to PMR and GCA, was reported in 3 (20%) of PMR cases and 2 (18.1%) of GCA cases.Citation1

Table 4. Results of HLA DRB1 typing of 26 patients with PMR or GCA following COVID-19 vaccination.

Treatments and outcomes

PMR

The median follow-up period was 16 (range 12–20) months. Details of treatment and follow-up are provided in . At the initiation of treatment, the median oral corticosteroids (CS) dose was 0.31 mg/kg/day [range 0.14–1.09]. Over time, the CS doses were tapered to a median of 0.17 (range 0.05–0.33), 0.09 (range 0.02–0.25), and 0.08 (range 0.05–0.13) mg/kg/day at months 3, 6, and 12, respectively. Discontinuation of CS treatment was achieved in 15 (25.4%) patients at month 6 and 44 (74.5%) patients at month 12, with a median treatment duration of 8 (range 4–17) months. Six (10%) patients experienced a relapse with a median time after CS initiation of 6 (range 6) months. Among these, 5 (8.3%) patients received steroid-sparing treatment, including methotrexate (MTX) in 3 cases and tocilizumab (TCZ) in 2 cases.

Table 5. Treatment and outcomes in the whole cohort of patients with PMR following COVID-19 vaccination.

No significant differences in terms of therapeutic management or relapse rates were observed among the subgroups identified based on the number of vaccine shots received. Overall, 24 (40%) patients were re-challenged with mRNA vaccines after their initial diagnosis. Among them, one (1.6%) experienced a relapse 10 days after re-challenge, exhibiting a GCA phenotype with pathological confirmation via TAB, and was subsequently included in the GCA survey.

GCA

The median follow-up duration was 18 (range 18–24) months. An overview of treatment and follow-up is presented in . Treatment was initiated with a median oral CS dose of 0.7 mg/kg/day (range 0.3–1.1). Subsequently, CS doses were tapered to a median of 0.3 (range 0.08–1), 0.2 (range 0.07–0.75), 0.1 (range 0.05–0.21), and 0.1 (range 0.01–0.25) mg/kg/day at months 3, 6, 12, and 18, respectively. Discontinuation of CS treatment was achieved in 5 (14.3%) patients at month 12 and 20 (57.1%) patients at month 18, with a median treatment duration of 16 (range 6–23) months.

Table 6. Treatment and outcomes in the whole cohort of patients with GCA following COVID-19 vaccination.

The number of patients who were free of CS at month 18 was significantly higher in the group that developed the disease after receiving the 3rd shot compared to the other two groups (3rd: 8/8 (100%) vs. 2nd: 7/13 (53.8%), 1st: 5/14 (35.7%), p < .05). A relapse occurred in 14 (40%) patients, with a median time after CS initiation of 6 (range 2–12) months. Sixteen (45.7%) patients received steroid-sparing agents, including MTX in 5 cases and TCZ in 12 cases. One patient sequentially received MTX (at month 6), TCZ (at month 9), and infliximab (at month 12) due to uncontrolled disease. In total, 10 (28.6%) patients were re-vaccinated with mRNA vaccines after their diagnosis, and none of them experienced a relapse.

Literature review

A systematic literature review allowed us to identify an additional 43 cases of PMR and 7 cases of GCA, the characteristics of which are described in Supplemental Tables 1 (PMR) and 2 (GCA).Citation6,Citation7,Citation12–30 MRNA vaccines were received by 31 (72%) PMR patients and 6 (85.7%) GCA patients. The median time to the onset of the first symptoms was 8 (range 1–20) days for PMR and 2 days (range 2–10) for GCA, respectively. For PMR cases: all had hip/girdle pain associated with a biological inflammatory syndrome (median CRP level: 85 mg/L (range 29–180)). CS were initiated in 36 (83.7%) patients, with 3 (6.3%) cases also receiving MTX. Regarding GCA cases, cephalic and systemic signs were reported in 6 (85.7%) and 5 (71.4%) patients, respectively. All patients, except one, exhibited a biological inflammatory syndrome (median CRP level: 63.2 mg/L (range 8.2–272)). Four (57.1%) patients had a positive TAB, while the remaining 3 (50%) showed LVV on imaging. All patients received CS treatment, with 2 cases (40%) also receiving steroid-sparing agents (MTX in one case and TCZ in another). No data on the outcomes and follow-up were available for these patients, regardless of their pathology.

Discussion

While PMR and GCA are two well-known inflammatory diseases affecting adults over the age of 50, there have been reports of de novo cases where individuals developed symptoms shortly after receiving the COVID-19 vaccination. In 2021, a case/non-case study on the international pharmacovigilance database VigiBase, conducted after a couple of months of COVID-19 vaccination, identified an elevated odds ratio for PMR (2.3) and GCA (2.7) when compared to other drugs suggesting a potential safety signal.Citation8

We initially conducted a national pharmacovigilance study encompassing all cases of PMR and GCA in individuals whose symptoms began within one month after receiving the COVID-19 vaccination. Over the study period, more than 70 million doses were administered to 25 million adults aged over 50. Regardless of the type and number of vaccine shots received, we identified 179 cases of PMR and 54 cases of GCA, resulting in a RR of 7.1 and 2.1 per 1,000,000 adults, and 2.53 and 0.73 per 1,000,000 injected doses for PMR and GCA, respectively. The elevated RR for both PMR and GCA was observed after the first dose of the ChadOx1 nCoV-19 vaccine (Oxford – AstraZeneca); however, the number of patients and injected doses was significantly lower compared to mRNA vaccines, primarily due to suspected coagulation side effects.Citation31 When combining the RR of both PMR and GCA per adult and per injected dose, it raises suspicion of a lower incidence of these diseases after COVID-19 vaccination compared to the unvaccinated population, making the interpretation of pharmacovigilance studies in this context more challenging.Citation1,Citation32

We provided a more comprehensive characterization of these patients through a nationwide survey. This survey identified 60 cases of PMR and 35 cases of GCA. Additionally, our literature review, which was based on one retrospective series and several case reports, enabled us to identify an additional 43 cases of PMR and 7 cases of GCA. However, the available data for these cases were limited, and the follow-up was relatively short. In our survey, it was observed that PMR and GCA were more likely to occur after the 1st or 2nd vaccine shot, with a lesser occurrence after the 3rd shot. This trend was expected, given the predominant use of mRNA vaccines, which resulted in a significantly higher number of patients receiving mRNA vaccines. Similar to previous large case series of unvaccinated PMR/GCA, patients in our study were primarily from the eighth decade of life, with 70 to 75% having cardiovascular risk factors. Notably, in contrast to the usual 2–3:1 female-to-male ratio reported in unvaccinated cohorts, patients with PMR after vaccination were predominantly male.Citation32–34

Aside from systemic manifestations, the phenotype of PMR and GCA cases following COVID-19 vaccination did not appear to differ from the unvaccinated PMR/GCA population and was not influenced by the number of vaccine shots received. Systemic manifestations were reported in 80% of COVID-19-vaccinated PMR/GCA cases, compared to 40 to 50% in the unvaccinated PMR/GCA population.Citation35 In addition, systemic manifestations are the most commonly reported COVID-19 vaccine-related adverse reaction suggesting a confounding factor, since we included patients who started their symptoms quickly after the immunizations.Citation36

In terms of ischemic complications, 20% of vaccinated GCA cases exhibited ophthalmologic manifestations, particularly AAION, which resembled those seen in unvaccinated GCA cases.Citation37 Importantly, although we had a limited number of vaccinated GCA patients in our study, no cerebrovascular or myocardial events were reported, that was in line with the low prevalence of 4% and 2.5% of cerebrovascular and myocardial ischemic complications reported in unvaccinated GCA, respectively.Citation38,Citation39 Imaging for LVV was conducted in over 90% of vaccinated GCA cases, predominantly using PET-CT since a 2015-published meta-analysis has reported a sensitivity and specificity of 90% in detecting LVV with PET-CT.Citation40 Combining both PET- CT and CTA imaging, LVV was identified in more than 50% of vaccinated GCA cases, with 35% exhibiting aortitis – a proportion slightly lower than the 45 to 65% of cases detected in the unvaccinated population.Citation41 Collectively, these data suggest a largely similar phenotype of PMR/GCA cases occurring after COVID-19 vaccination, particularly GCA-related ischemic complications and -LVV, when compared to the unvaccinated population. CS serve as the fundamental medical treatment for both PMR and GCA, although the initial dose and tapering schedule differ between these 2 diseases. In our study, the starting dose for PMR and GCA aligned with previous recommendations.Citation42–44 However, in PMR patients, the tapering schedule appeared to be somewhat shorter than recommended, as 75% were able to discontinue CS after 12 months, with a median duration of CS use being 8 months. During the follow-up period, 10% of vaccinated PMR cases and 40% of vaccinated GCA cases experienced relapses, which seemed slightly lower compared to unvaccinated cases. Some studies have reported relapse rates of 20–55% in PMR patients during the first year and 50% in GCA cases during the first 2 years.Citation45,Citation46

In line with society recommendations, steroid-sparing agents were initiated to manage most relapses, with MTX and TCZ being the primary choices, especially TCZ in the case of GCA.Citation39,Citation40 These findings suggest that vaccinated PMR and GCA cases did not exhibit a higher risk of relapse, prolonged CS use, or increased requirements for steroid-sparing treatments, indicating a medical management approach similar to that of unvaccinated cases.

The median time between the first reported symptoms of PMR/GCA and immunization was short, from 7 to 10 days suggesting a temporal association that is considered a primary contributing factor in identifying and defining environmentally associated disorders.Citation47 Furthermore, among 34 patients who underwent re-challenge with vaccination, one PMR patient exhibited a GCA-like phenotype 10 days after receiving a new immunization, which was subsequently confirmed pathologically, adding another noteworthy element to consider. A descriptive analysis conducted across three international databases revealed that PMR accounted for 9.2% of reported cases following immunization, with a higher likelihood of occurrence following influenza vaccination, whereas GCA constituted 3% of all reported cases.Citation48 However, detailed clinical data were not available. Interestingly, another study reported ten cases of GCA that occurred shortly after influenza vaccination, indicating a prevalence of 2.8% for these cases within a hospital-based recruitment context.Citation49

Thus far, while no specific components of COVID-19 vaccines have been strictly identified as responsible for these rare inflammatory-mediated adverse events, one hypothesis that could be considered is consistent with the concept of autoimmune/inflammatory syndrome induced by adjuvant (ASIA).Citation50 Although the association of HLA DRB1 haplotypes has been described as a minor criterion for diagnosing ASIA, our study found HLA DRB1*04 in 20% of PMR/GCA patients, suggesting a genetic predisposition previously documented in PMR and GCA, which may contribute to the development of both diseases.Citation1

Interestingly, the lipid nanoparticles used in COVID-19 mRNA vaccines have been found to induce significant inflammatory interleukin (IL)-1β/IL-6 secretion in a murine model, potentially enhancing the adjuvant immunogenicity of the vaccine.Citation51 COVID-19 vaccines encode the SARS-CoV- 2 spike protein, which plays a crucial role in binding the virus to host cells expressing its receptor, angiotensin-converting enzyme 2 (ACE2). A recent study elegantly demonstrated that the spike protein promotes an ACE2 type 1 receptor-mediated signaling cascade, induces transcriptional regulation involving nuclear factor-kappa B (NF-κB) and AP- 1/c-FOS via Mitogen-activated protein kinases (MAPK) activation, and increases the release of IL-6, a key cytokine in the pathogenesis of PMR and GCA.Citation52 Considering the well-established involvement of cellular immunity in GCA pathogenesis, a Th1 profile was also detected by day 8 after COVID-19 vaccine immunization, similar in magnitude to those observed after natural infection, which could contribute to the disease process.Citation53

This study has several limitations. Some relevant cases may have been missed in the national in-depth analysis since reports have been declarative. In addition, some clinical data were not available owing to the retrospective design and the heterogeneity of management of these patients. However, we used stringent inclusion criteria regarding the diagnosis as well as the timeline from the immunization to reduce selection and confusion bias. Finally, we were not able to perform statistical comparisons regarding the phenotype presentation, the treatment strategy and the follow-up between vaccinated and unvaccinated patients, since we did not have control-cohort group of unvaccinated PMR/GCA patients.

In conclusion, although cases of PMR or GCA following COVID-19 vaccination appear to be rare, the short time between immunization and the onset of initial symptoms suggests a temporal association. The phenotype, ischemic and vascular complications, as well as the therapeutic management of these patients, resemble those of PMR or GCA in the unvaccinated population, strongly supporting the beneficial effects of COVID-19 vaccination in the context of widespread circulation of SARS-CoV-2. While a clear causative link between immunization and the onset of the disease remains unclear, various hypotheses have been proposed.

Physicians should be aware of this vaccine-related phenomenon, and re-challenging may be considered while maintaining vigilance.

Author contributors

PAJ collected data, conducted statistical analysis and wrote the manuscript. PAJ, AM, JM, DS and GK designed the study, and wrote the manuscript. SO, SC, KHS, EL, SP, MM, BT, PEG, LT, VL, JK, DR, BB, TW, DR, AP, CC, VG, MH, MP, CW, LK, PG, TR, NB, VP, EB, AL, JH, NS, NS, JMD, BC, AP, RA, BG, PG, HL, PJW collected data. PG, SG, VG, JM conducted the pharmacovigilance study. All authors provided care for study patients. PAJ and GK are guarantors. They accept full responsibility for the work and of the conduct of the study, had access to the data, and controlled the decision to publish.

Patient consent for publication

Consent obtained directly from patient(s).

Supplemental material

Supplemental tables 1 and 2.docx

Download MS Word (20.8 KB)

Acknowledgments

We thank the French Network of Regional Center of Pharmacovigilance for their help in the study.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data are available upon reasonable request.

Supplementary material

Supplemental data for this article can be accessed on the publisher’s website at https://doi.org/10.1080/21645515.2024.2334084

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Salvarani C, Pipitone N, Versari A, Hunder GG. Clinical features of polymyalgia rheumatica and giant cell arteritis. Nat Rev Rheumatol. 2012;8(9):509–11. doi:10.1038/nrrheum.2012.97.
  • Kobayashi S, Yano T, Matsumoto Y, Numano F, Nakajima N, Yasuda K, Yutani C, Nakayama T, Tamakoshi A, Kawamura T. et al. Clinical and epidemiologic analysis of giant cell (temporal) arteritis from a nationwide survey in 1998 in Japan: the first government-supported nationwide survey. Arthritis & Rheumatism. 2003;49(4):594–8. doi:10.1002/art.11195.
  • Yates M, Graham K, Watts RA, MacGregor AJ. The prevalence of giant cell arteritis and polymyalgia rheumatica in a UK primary care population. BMC Musculoskelet Disord. 2016;17(1):285. doi:10.1186/s12891-016-1127-3.
  • Zhang Z, Shen Q, Chang H. Vaccines for COVID-19: a systematic review of immunogenicity, current development, and future prospects. Front Immunol. 2022;13:843928. doi:10.3389/fimmu.2022.843928.
  • Lamprinou M, Sachinidis A, Stamoula E, Vavilis T, Papazisis G. COVID-19 vaccines adverse events: potential molecular mechanisms. Immunol Res. 2023;71(3):356–72. doi:10.1007/s12026-023-09357-5.
  • Mejren A, Sørensen CM, Gormsen LC, Tougaard RS, Nielsen BD. Large-vessel giant cell arteritis after COVID-19 vaccine. Scand J Rheumatol. 2022;51(2):154–5. doi:10.1080/03009742.2021.1961401.
  • Haruna K, Shirota S, Nishioka H. Polymyalgia Rheumatica (PMR) lacking shoulder pain following COVID-19 vaccination. Cureus. 2023;15:e34714. doi:10.7759/cureus.34714.
  • Mettler C, Jonville-Bera A-P, Grandvuillemin A, Treluyer J-M, Terrier B, Chouchana L. Risk of giant cell arteritis and polymyalgia rheumatica following COVID-19 vaccination: a global pharmacovigilance study. Rheumatology (Oxford). 2022;61(2):865–7. doi:10.1093/rheumatology/keab756.
  • Jonville-Bera AP, Gautier S, Micallef J, Massy N, Atzenhoffer M, Grandvuillemin A, Drici M-D. Monitoring the safety of drugs and COVID-19 vaccines by the French Pharmacovigilance Centers during the pandemic: a win-win bet with Health Authorities! Therapies. 2023;78(5):467–75. doi:10.1016/j.therap.2023.03.002.
  • Dasgupta B, Cimmino MA, Maradit-Kremers H, Schmidt WA, Schirmer M, Salvarani C, Bachta A, Dejaco C, Duftner C, Jensen HS. et al. 2012 provisional classification criteria for polymyalgia rheumatica: a European League Against Rheumatism/American College of Rheumatology collaborative initiative. Ann Rheum Dis. 2012;71(4):484–92. doi:10.1136/annrheumdis-2011-200329.
  • Ponte C, Grayson PC, Robson JC, Suppiah R, Gribbons KB, Judge A, Craven A, Khalid S, Hutchings A, Watts RA. et al. 2022 American College of Rheumatology/EULAR classification criteria for giant cell arteritis. Ann Rheum Dis. 2022;81(12):1647–53. doi:10.1136/ard-2022-223480.
  • Greb CS, Aouhab Z, Sisbarro D, Panah E. A case of giant cell arteritis presenting after COVID-19 vaccination: is it just a coincidence? Cureus. 2022;14:e21608. doi:10.7759/cureus.21608.
  • Xia C, Edwards R, Omidvar B. A case of giant cell arteritis with a normal Erythrocyte Sedimentation Rate (ESR) post ChAdOx1 nCoV-19 vaccination. Cureus. 2022;14:e25388. doi:10.7759/cureus.25388.
  • Yamada S, Yamada K, Nishida H. A case of sequential development of polymyalgia rheumatica and guillain-barré syndrome following administration of the Pfizer-BioNTech COVID-19 vaccine. Intern Med. 2022;61(19):2995. doi:10.2169/internalmedicine.0319-22.
  • Ahmad A, Baker DL. An unusual side effect of the COVID-19 vaccine: a possible trigger of Polymyalgia Rheumatica. Cureus. 2022;14:e26617. doi:10.7759/cureus.26617.
  • Gambichler T, Krogias C, Tischoff I, Tannapfel A, Gold R, Susok L. Bilateral giant cell arteritis with skin necrosis following SARS-CoV-2 vaccination. Br J Dermatol. 2022;186(2):e83. doi:10.1111/bjd.20824.
  • Che S-A, Lee KY, Yoo Y-J. Bilateral ischemic optic neuropathy from giant cell arteritis following COVID-19 vaccination. J Neuroophthalmol. Published Online First: 14 June 2022. doi:10.1097/WNO.0000000000001570.
  • Hussain W, Moorthy A. Covid 19 vaccination -trigger for new onset polymyalgia rheumatica. Int J Rheum Dis. 2021;24:158–158.
  • Ishizuka DK, Katayama K, Ohira Y. Giant cell arteritis presenting with chronic cough and headache after BNT162b2 mRNA COVID-19 vaccination. QJM. 2022;115(9):621–2. doi:10.1093/qjmed/hcac171.
  • Watad A, De Marco G, Mahajna H, Druyan A, Eltity M, Hijazi N, Haddad A, Elias M, Zisman D, Naffaa ME. et al. Immune-mediated disease flares or new-onset disease in 27 subjects following mRNA/DNA SARS-CoV-2 vaccination. Vaccines. 2021;9(5):435. doi:10.3390/vaccines9050435.
  • Ursini F, Ruscitti P, Addimanda O, Foti R, Raimondo V, Murdaca G, Caira V, Pigatto E, Cuomo G, Lo Gullo A. et al. Inflammatory rheumatic diseases with onset after SARS-CoV-2 infection or COVID-19 vaccination: a report of 267 cases from the COVID-19 and ASD group. RMD Open. 2023;9(2):e003022. doi:10.1136/rmdopen-2023-003022.
  • Anzola AM, Trives L, Martínez-Barrio J, Pinilla B, Álvaro-Gracia JM, Molina-Collada J. New-onset giant cell arteritis following COVID-19 mRNA (BioNtech/Pfizer) vaccine: a double-edged sword? Clin Rheumatol. 2022;41(5):1623–5. doi:10.1007/s10067-021-06041-7.
  • Osada A, Sakuragi C, Toya C, Mitsuo A. New-onset polymyalgia rheumatica following the administration of the Pfizer-BioNTech COVID-19 vaccine. Intern Med. 2022;61(5):749–53. doi:10.2169/internalmedicine.8651-21.
  • Mohareb M, Bharadwaj A, Nandagudi A. P061 polymyalgia rheumatica following COVID-19 vaccination: presentation of four patients. Rheumatology (Oxford). 2022;61(Supplement_1):keac133.060. doi:10.1093/rheumatology/keac133.060.
  • Lourenço C, Pascoal A, Paiva A, Campos, I, Pagaimo, J. 2022. Polymyalgia rheumatica after ChAdOx1 nCov-19 vaccine: a case report. Cureus. 14:e25346.
  • Manzo C, Castagna A, Isetta M. Polymyalgia rheumatica and polymyalgia-like syndromes as adverse events following COVID-19 vaccines: working notes from a narrative review of published literature. Reumatologia. 2022;60(2):142–7. doi:10.5114/reum.2022.115665.
  • Yokote A, Fujioka S, Takahashi N, Mishima T, Tsuboi Y. Polymyalgia rheumatica following COVID-19 vaccination. Intern Med. 2022;61(11):1775–7. doi:10.2169/internalmedicine.8934-21.
  • Furr T, Garg M. Rare cases of polymyalgia rheumatica after receiving COVID-19 vaccinations. Cureus. 2023;15(4):e37782. doi:10.7759/cureus.37782.
  • Izuka S, Komai T, Natsumoto B, Shoda H, Fujio K. Self-limited polymyalgia rheumatica-like syndrome following mRNA-1273 SARS-CoV-2 vaccination. Intern Med. 2022;61(6):903–6. doi:10.2169/internalmedicine.8829-21.
  • Nielsen AW, Hansen IT, Gormsen LC, Hauge E-M, Keller KK. Self-limiting reactive disease mimicking polymyalgia rheumatica following moderna COVID-19 vaccine. Scand J Rheumatol. 2022;51(5):411–3. doi:10.1080/03009742.2022.2070959.
  • Thiele T, Weisser K, Schönborn L, Funk MB, Weber G, Greinacher A, Keller-Stanislawski B. Laboratory confirmed vaccine-induced immune thrombotic thrombocytopenia: retrospective analysis of reported cases after vaccination with ChAdOx-1 nCoV-19 in Germany. Lancet Reg Health Eur. 2022;12:100270. doi:10.1016/j.lanepe.2021.100270.
  • Gonzalez-Gay MA, Vazquez-Rodriguez TR, Lopez-Diaz MJ, Miranda‐Filloy JA, Gonzalez‐Juanatey C, Martin J, Llorca J. Epidemiology of giant cell arteritis and polymyalgia rheumatica. Arthritis & Rheumatism. 2009;61(10):1454–61. doi:10.1002/art.24459.
  • de Boysson H, Daumas A, Vautier M, Parienti J-J, Liozon E, Lambert M, Samson M, Ebbo M, Dumont A, Sultan A. et al. Large-vessel involvement and aortic dilation in giant-cell arteritis. A multicenter study of 549 patients. Autoimmun Rev. 2018;17(4):391–8. doi:10.1016/j.autrev.2017.11.029.
  • Toro-Gutiérrez CE, CañCañAs CA, Mantilla RD, Beltrán S, Pastrana-Gonzalez V, Vecino MJ, Rodriguez-Jimenez M, Rojas M. Polymyalgia rheumatica: a case series from Colombia and analysis of Latin America. J Transl Autoimmun. 2021;4:100115. doi:10.1016/j.jtauto.2021.100115.
  • Salvarani C, Cantini F, Boiardi L, Hunder GG. Polymyalgia rheumatica and giant-cell arteritis. N Engl J Med. 2002;347(4):261–71. doi:10.1056/NEJMra011913.
  • Dhamanti I, Suwantika AA, Adlia A, Yamani LN, Yakub F. Adverse reactions of COVID-19 vaccines: a scoping review of observational studies. Int J Gen Med. 2023;16:609–18. doi:10.2147/IJGM.S400458.
  • Soriano A, Muratore F, Pipitone N, Boiardi L, Cimino L, Salvarani C. Visual loss and other cranial ischaemic complications in giant cell arteritis. Nat Rev Rheumatol. 2017;13(8):476–84. doi:10.1038/nrrheum.2017.98.
  • Penet T, Lambert M, Baillet C, Outteryck O, Hénon H, Morell-Dubois S, Hachulla E, Launay D, Pokeerbux MR. Giant cell arteritis-related cerebrovascular ischemic events: a French retrospective study of 271 patients, systematic review of the literature and meta-analysis. Arthritis Res Ther. 2023;25(1):116. doi:10.1186/s13075-023-03091-x.
  • Greigert H, Zeller M, Putot A, Steinmetz E, Terriat B, Maza M, Falvo N, Muller G, Arnould L, Creuzot-Garcher C. et al. Myocardial infarction during giant cell arteritis: a cohort study. Eur J Intern Med. 2021;89:30–8. doi:10.1016/j.ejim.2021.02.001.
  • Soussan M, Nicolas P, Schramm C, Katsahian S, Pop G, Fain O, Mekinian A. Management of large-vessel vasculitis with FDG-PET: a systematic literature review and meta-analysis. Medicine (Baltimore). 2015;94(14):e622. doi:10.1097/MD.0000000000000622.
  • Prieto-González S, Arguis P, García-Martínez A, Espígol-Frigolé G, Tavera-Bahillo I, Butjosa M, Sánchez M, Hernández-Rodríguez J, Grau JM, Cid MC. et al. Large vessel involvement in biopsy-proven giant cell arteritis: prospective study in 40 newly diagnosed patients using CT angiography. Ann Rheum Dis. 2012;71(7):1170–6. doi:10.1136/annrheumdis-2011-200865.
  • Hellmich B, Agueda A, Monti S, Buttgereit F, de Boysson H, Brouwer E, Cassie R, Cid MC, Dasgupta B, Dejaco C. et al. 2018 update of the EULAR recommendations for the management of large vessel vasculitis. Ann Rheum Dis. 2020;79(1):19–30. doi:10.1136/annrheumdis-2019-215672.
  • Dejaco C, Singh YP, Perel P, Hutchings A, Camellino D, Mackie S, Abril A, Bachta A, Balint P, Barraclough K. et al. 2015 recommendations for the management of polymyalgia rheumatica: a European League Against Rheumatism/American College of Rheumatology collaborative initiative. Ann Rheum Dis. 2015;74(10):1799–807. doi:10.1136/annrheumdis-2015-207492.
  • Mackie SL, Dejaco C, Appenzeller S, Camellino D, Duftner C, Gonzalez-Chiappe S, Mahr A, Mukhtyar C, Reynolds G, de Souza AWS. et al. British society for rheumatology guideline on diagnosis and treatment of giant cell arteritis. Rheumatology (Oxford). 2020;59(3):e1–23. doi:10.1093/rheumatology/kez672.
  • González-Gay MA, Matteson EL, Castañeda S. Polymyalgia rheumatica. Lancet. 2017;390(10103):1700–12. doi:10.1016/S0140-6736(17)31825-1.
  • Moreel L, Betrains A, Molenberghs G, Vanderschueren S, Blockmans D. Epidemiology and predictors of relapse in giant cell arteritis: a systematic review and meta-analysis. Joint Bone Spine. 2023;90(1):105494. doi:10.1016/j.jbspin.2022.105494.
  • Miller FW, Hess EV, Clauw DJ, Hertzman PA, Pincus T, Silver RM, Mayes MD, Varga J, Medsger TA, Love LA. et al. Approaches for identifying and defining environmentally associated rheumatic disorders. Arthritis & Rheumatism. 2000;43(2):243–9. doi:10.1002/1529-0131(200002)43:2<243:AID-ANR2>3.0.CO;2-K.
  • Felicetti P, Trotta F, Bonetto C, Santuccio C, Brauchli Pernus Y, Burgner D, Chandler R, Girolomoni G, Hadden RDM, Kochhar S. et al. Spontaneous reports of vasculitis as an adverse event following immunization: a descriptive analysis across three international databases. Vaccine. 2016;34(51):6634–40. doi:10.1016/j.vaccine.2015.09.027.
  • Liozon E, Parreau S, Filloux M, Dumonteil S, Gondran G, Bezanahary H, Ly KH, Fauchais AL. Giant cell arteritis or polymyalgia rheumatica after influenza vaccination: a study of 12 patients and a literature review. Autoimmun Rev. 2021;20(2):102732. doi:10.1016/j.autrev.2020.102732.
  • Cohen Tervaert JW, Martinez-Lavin M, Jara LJ, Halpert G, Watad A, Amital H, Shoenfeld Y. Autoimmune/Inflammatory syndrome induced by adjuvants (ASIA) in 2023. Autoimmun Rev. 2023;22(5):103287. doi:10.1016/j.autrev.2023.103287.
  • Ndeupen S, Qin Z, Jacobsen S. et al. The mRNA-LNP platform’s lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. iScience. 2021;24:103479. doi:10.1016/j.isci.2021.103479.
  • Patra T, Meyer K, Geerling L, Isbell TS, Hoft DF, Brien J, Pinto AK, Ray RB, Ray R. SARS-CoV-2 spike protein promotes IL-6 trans-signaling by activation of angiotensin II receptor signaling in epithelial cells. PloS Pathog. 2020;16(12):e1009128. doi:10.1371/journal.ppat.1009128.
  • Moss P. The T cell immune response against SARS-CoV-2. Nat Immunol. 2022;23(2):186–93. doi:10.1038/s41590-021-01122-w.