756
Views
0
CrossRef citations to date
0
Altmetric
Case Report

New-onset of rheumatic diseases following COVID-19 vaccination: the report of three cases and a literature review

, , , , , , , , , , & show all
Received 19 Jan 2024, Accepted 26 Mar 2024, Published online: 16 Apr 2024

Abstract

Vaccines against coronavirus disease 2019 (COVID-19) have been distributed in most countries for the prevention of onset and aggravation of COVID-19. Recently, there have been increasing numbers of reports on new-onset autoimmune and autoinflammatory diseases following COVID-19 vaccination, however, only little information is available on the long-term safety of these vaccines. Here, we experienced three cases of new-onset rheumatic diseases following COVID-19 vaccination, one case each of rheumatoid arthritis (RA), anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) and systemic lupus erythematosus (SLE). The symptom onset ranged from one day to a few days following vaccination. The patients of AAV and SLE were treated successfully with glucocorticoid therapy, and the patient of RA died due to COVID-19. In the literature review of new-onset rheumatic diseases following COVID-19 vaccination, which including seven cases of RA, 37 cases of AAV and 18 cases of SLE, the mean time from vaccination to onset was approximately 11 to 12 days. Most cases improved with glucocorticoid, immunosuppressive drugs and biologic agents. Although such adverse effects are rare, and vaccines are useful in prevent onset and severity of infections, continued accumulation of similar cases is important in terms of examining the long-term safety and understanding pathogenic mechanism of rheumatic diseases.

1. Introduction

New onset cases of coronavirus disease 2019 (COVID-19) continue to occur worldwide, and the cumulative number of patients has also been increasing in Japan. COVID-19 vaccines have been distributed widely in most countries to prevent the onset and aggravation of COVID-19 [Citation1]. Vaccine effectiveness against SARS-CoV-2 infection, hospitalization and mortality generally declines over time. Receiving multiple doses of the vaccine improves booster effectiveness and immune recovery and therefore, improves immunity in a population [Citation2]. There is little information regarding rare adverse events or the long-term safety of the COVID-19 vaccines although recently, there have been increasing numbers of case reports on new-onset or relapsed autoimmune or autoinflammatory diseases following COVID-19 vaccination. We have already reported three cases of new-onset adult-onset Still’s disease (AOSD) following COVID-19 vaccination [Citation3]. However, the relationship between development of rare autoimmune or autoinflammatory diseases and COVID-19 vaccination remains controversial. Here we report three additional cases: one case each of rheumatoid arthritis (RA), anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), and systemic lupus erythematosus (SLE) with the critical literature review on this important topic.

2. Case reports

2.1. Case 1

A 79-year-old woman developed swelling and pain in finger joints and both ankle joints 2–3 days after the second dose of the BNT162b2 mRNA COVID-19 vaccine. She was admitted to our hospital with difficulty walking at 27 days after the vaccination. She had a history of hypertension and nontuberculous mycobacterial (NTM) infection. She and her family had no history of rheumatic diseases. She had no adverse events of note after the first vaccination. Physical examination revealed polyarthritis with swelling in 20/28 joints and tenderness in 23/28 joints, with high disease activity (disease activity score in 28 joints with ESR [DAS-28 ESR]: 8.48 and [DAS-28 CRP]: 7.94). Her health assessment questionnaire disability index (HAQ-DI) score was 1.875. Laboratory test results were as follows: white blood cell count 7680/µL (neutrophils 76.5%, lymphocytes 14.3%), Hb 8.2 g/dL, platelets 47.4 × 104/µL, erythrocite sedimentation rate (ESR) 95 mm/h, albumin 2.3 g/dL, creatine kinase 28 U/L, aspartate aminotransferase (AST) 18 U/L, alanine aminotransferase (ALT) 19 U/L, lactate dehydrogenase (LDH) 158 U/L, creatinine 0.81 mg/dL, C-reactive protein (CRP) 10.69 mg/dL, serum ferritin 621 ng/mL, matrix metalloproteinase-3 (MMP-3) 249.4 ng/mL, antinuclear antibody (ANA) negative, rheumatoid factor (RF) 59 IU/mL and anti-cyclic citrullinated peptide (CCP) antibody 184.6 U/mL. The interferon gamma release assay was negative. polymerase chain reaction (PCR) test for SARS-CoV-2 from nasopharyngeal swabs was negative. Anti-mycobacterium avium complex antibody was positive (1.0 U/mL). Ultrasonography of wrists and finger joints revealed synovitis. X-rays of the joints found no evidence of erosions. RA was diagnosed according to the 2010 American College of Rheumatology (ACR)/European League Against Rheumatism classification criteria for RA [Citation4]. The patient was treated with subcutaneous abatacept (125 mg, weekly) without methotrexate because of concomitant NTM. Later, her arthritis was refractory to abatacept, followed by etanercept, so she was switched to baricitinib. Baricitinib was effective but was discontinued after she developed a urinary tract infection, and was switched to sarilumab but then discontinued due to herpes zoster. She had been treated with tacrolimus, but then, developed COVID-19 and died ().

Figure 1. Clinical course of case 3, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the second vaccination date, set as day 0. ABT, abatacept; ETN, etanercept; BAR, baricitinib; SASP, salazosulfapyridine; SAR, sarilumab; TAC, tacrolimus.

Figure 1. Clinical course of case 3, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the second vaccination date, set as day 0. ABT, abatacept; ETN, etanercept; BAR, baricitinib; SASP, salazosulfapyridine; SAR, sarilumab; TAC, tacrolimus.

2.2. Case 2

A 71-year-old woman developed a fever and cough the day after her second dose of the BNT162b2 mRNA COVID-19 vaccine. She received medication from her previous physician but did not improve. Seven days after the vaccination, her laboratory data were abnormal with white blood cell count 12,230/µL, CRP 10.9 mg/dL and no computed tomography (CT) findings in the chest and abdomen. Her cough improved with antibiotic treatment, but she then developed pain in her shoulders, lower back and lower legs. She was admitted to our hospital 18 days after her second COVID-19 vaccination. She and her family had no history of rheumatic diseases. She had no adverse events of note after the first vaccination. She developed swelling and pain in the finger joints and grasping pain in the bilateral lower legs. Laboratory test results were as follows: white blood cell count 12,290/µL (neutrophils 75.1%, lymphocytes 14.6%), Hb 10.5 g/dL, platelets 50.2 × 104/µL, ESR 120 mm/h, albumin 2.5 g/dL, creatine kinase 11 U/L, AST 15 U/L, ALT 16 U/L, LDH 128 U/L, creatinine 0.78 mg/dL, CRP 14.53 mg/dL, serum ferritin 248 ng/mL, ANA negative, RF 64 IU/mL and Myeloperoxidase Anti-neutrophil Cytoplasmic Antibody (MPO-ANCA) 12.0 U/mL. PCR test for SARS-CoV-2 from nasopharyngeal swabs was negative. Other infectious diseases were excluded by blood, sputum and urine culture tests. Magnetic resonance imaging demonstrated high signal intensity in the bilateral lower leg muscles on short-tau inversion recovery (STIR) images (). A biopsy of the left gastrocnemius muscle revealed fragmented elastic fibers with Elastica van Gieson staining, signifying the presence of blood vessels, and AAV was diagnosed (). Oral glucocorticoid therapy (prednisolone 35 mg/day; 0.7 mg/kg/day) was administered initially. Her symptoms improved. The inflammatory findings disappeared, MPO-ANCA decreased to negative and the patient was discharged. During her treatment, she developed pyelonephritis, idiopathic osteonecrosis of the femoral head and urinary tract infection. The dose of prednisolone was gradually reduced to 3 mg, and no relapse was observed ().

Figure 2. Magnetic resonance imaging finding in the lower legs. STIR images show heterogeneous high-signal areas in the bilateral tibialis anterior, extensor digitorum longus, peroneus longus, popliteus, soleus, plantaris and gastrocnemius muscles, suggesting myositis or vasculitis spreading to these areas.

Figure 2. Magnetic resonance imaging finding in the lower legs. STIR images show heterogeneous high-signal areas in the bilateral tibialis anterior, extensor digitorum longus, peroneus longus, popliteus, soleus, plantaris and gastrocnemius muscles, suggesting myositis or vasculitis spreading to these areas.

Figure 3. Histopathological findings of the left gastrocnemius muscle. Hematoxylin-Eosin staining shows an inflammatory cell infiltrate, including lymphocytes and neutrophils (A, B: arrowheads), and in some cases, neutrophils are clustered. Elastica Van Gieson staining reveals scattered fragmented elastic fibres (C: arrows).

Figure 3. Histopathological findings of the left gastrocnemius muscle. Hematoxylin-Eosin staining shows an inflammatory cell infiltrate, including lymphocytes and neutrophils (A, B: arrowheads), and in some cases, neutrophils are clustered. Elastica Van Gieson staining reveals scattered fragmented elastic fibres (C: arrows).

Figure 4. Clinical course of case 4, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the second vaccination date, set as day 0. MPO-ANCA, myeloperoxidase-anti-neutrophil cytoplasmic antibody.

Figure 4. Clinical course of case 4, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the second vaccination date, set as day 0. MPO-ANCA, myeloperoxidase-anti-neutrophil cytoplasmic antibody.

2.3. Case 3

A 32-year-old woman developed chest pain, left axillary lymphadenopathy and erythema on her cheeks 2–3 days after her third dose of the COVID-19 vaccine (first and second vaccines, BNT162b2; third vaccine, mRNA-1273). The erythema spread and lip swelling appeared. She developed a low-grade fever around 37 °C. Despite treatment by a dermatologist, the erythema on her cheeks spread and appeared also on her upper extremities and body. After treatment with methylprednisolone (drug dosage unknown) by her previous physician, the fever and lip swelling improved. The patient was admitted to our hospital with suspected collagen disease approximately four months after the vaccination. She had no history of rheumatic diseases and her grandmother had Takayasu arteritis. She had no adverse events of note after the first and second vaccinations. On physical examination, there was discoid lupus around her nasal wings, erythema chilblains with desquamation of her auricle, erythema chilblains on her fingers and left axillary lymphadenopathy. Laboratory test results were as follows: white blood cell count 2500/µL (neutrophils 56.4%, lymphocytes 30.8%), Hb 12.8 g/dL, platelets 16.2 × 104/µL, ESR 64 mm/h, albumin 3.8 g/dL, creatine kinase 24 U/L, AST 40 U/L, ALT 39 U/L, LDH 346 U/L, creatinine 0.46 mg/dL, CRP 0.19 mg/dL, 50% hemolytic complement (CH 50) activity 19.9 U/mL (normal range: 31.6–57.6), the third component of complement (C3) 36 mg/dL (normal range: 73–138), the fourth component of complement (C4) 7 mg/dL (normal range: 11–31), serum ferritin 87 ng/mL, IgG 1735 mg/dL, IgA 299 mg/dL, IgM 92 mg/dL and activated partial thromboplastin time 33.4 s. ANA was positive (1:160, speckled patterns). Anti-ribonucleoprotein antibody (119.0 U/mL), anti-Sm antibody (481.0 U/mL) and anti-SS-A antibody (1200.0 U/mL) were positive. Anti-dsDNA antibody, lupus anticoagulant test and anticardiolipin IgG were negative. PCR test for SARS-CoV-2 from nasopharyngeal swabs was negative. Real-time PCR test for Epstein-Barr virus was negative. Other infectious diseases were excluded by blood, sputum and urine culture tests. CT of the neck, chest, abdomen and pelvis revealed multiple lymphadenopathy at the bilateral axillas and splenomegaly. SLE was diagnosed according to the 2019 European League Against Rheumatism/ACR criteria for SLE [Citation5], based on discoid lupus, leukopenia, low complement, ANA-positivity and anti-Sm antibody–positivity. Oral glucocorticoid therapy (prednisolone 30 mg/day; 0.6 mg/kg/day) was administered initially. Hydroxychloroquine was then used concomitantly. Her symptoms improved and the inflammatory findings disappeared, and the patient was discharged. The dose of prednisolone was then gradually reduced to 5 mg, and no relapse was observed ().

Figure 5. Clinical course of case 5, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the third vaccination date, set as day 0. HCQ, Hydroxychlorquine; CH50, 50% hemolytic complement activity; dsDNA, double-stranded deoxyribonucleic acid; WBC, white blood cell.

Figure 5. Clinical course of case 5, including symptoms, laboratory findings and treatment. ‘Day’ means the number of days after the third vaccination date, set as day 0. HCQ, Hydroxychlorquine; CH50, 50% hemolytic complement activity; dsDNA, double-stranded deoxyribonucleic acid; WBC, white blood cell.

3. Discussion

We have previously reported three cases of AOSD following COVID-19 vaccination [Citation3]. We herein report additional three cases, including one case each of RA, AAV and SLE that developed following vaccination with an mRNA COVID-19 vaccine. The symptom onset ranged from one day to a few days following vaccination. The patients of AAV and SLE were treated successfully with glucocorticoid therapy, and the patients of RA, experienced with several infections under disease-modifying antirheumatic drugs, died due to COVID-19.

summarize the details of the previously reported and present cases of new-onset autoimmune diseases following COVID-19 vaccination.

Table 1. Previous and present cases of rheumatoid arthritis following COVID-19 vaccination.

Table 2. Previous and present cases of ANCA-associated vasculitis following COVID-19 vaccination.

Table 3. Previous and present cases of SLE following COVID-19 vaccination.

The previously reported cases of RA were only six cases and the mean time from vaccination to onset of RA was 12.8 days ( [Citation6–11]:), in contrast, our case had a short time, 2-3 days, between onset and vaccination. RF and ACPA were strongly positive in five cases including our case. The previously reported cases improved with methotrexate, iguratimod, leflunomide or biologics concomitant with glucocorticoid. Although the disease activity of our patient also improved with biologics and JAK inhibitors, she died from COVID-19 since our case was an advanced age, as a risk factor.

The previously reported cases of AAV are summarized in [Citation12–42]. The mean time from vaccination to onset was 11.8 days, and most cases occurred within 7 days. Of the 37cases, 22 were MPO-ANCA positive, 4 PR3-ANCA positive, 4 both positive and 1 both negative. One was positive for both p-ANCA and C-ANCA. Thirty patients were diagnosed with glomerulonephritis on renal biopsy. The organ lesions of other cases included auricular cartilage, pulmonary hemorrhage or brain hemorrhage. All previously reported cases improved with glucocorticoid, immunosuppressive drugs, biologic agents, and plasma exchange. Our case diagnosed vasculitis on muscle biopsy with MPO positive and responded well to glucocorticoid therapy.

The previously reported cases of SLE are summarized in [Citation43–57]. The mean time from vaccination to onset was 12.8 days, and most cases occurred within 14 days. ANA was positive in all 18 cases, anti-dsDNA antibody was positive in 12 cases, and anti-Sm antibody was positive in six cases. Lupus anticoagulant test was positive in two cases and beta-2 glycoprotein IgG was positive in two cases. Affected organs included Evans syndrome in one patient, leukopenia in three patients, pancytopenia in three patients, lupus nephritis in three patients, myocarditis in one patient, autoimmune pancreatitis in one patient and one patient had complication of antiphospholipid syndrome. There was no specific trend in the clinical features of patients. All cases improved or partially improved with glucocorticoid and immunosuppressive drugs.

Previous studies have reported autoimmune diseases that developed after vaccines other than COVID-19 vaccine, such as vaccines for influenza virus, human papilloma virus and hepatitis B virus [Citation58–60]. These vaccines have been suspected to trigger autoimmunity through molecular mimicry and immune crossreactivity. The possibility that adjuvants in vaccines may cause autoimmune diseases has also been considered.

There are two main types of COVID-19 vaccine formulations; mRNA encoding the SARS-CoV-2 spike protein (S) encapsulated in lipid nanoparticles and adenovirus (AdV) vectors encoding the S protein [Citation61]. COVID-19 vaccine mRNA and AdV particles invade dendritic cells, resulting in the production of S-proteins. The adjuvant activity of the vaccine activates innate immune sensors, resulting in the production of type I interferon and multiple pro-inflammatory mediators, which activate T cells and induce immunity against SARS-CoV-2 [Citation62]. Increased production of type I interferon and multiple pro-inflammatory mediators following vaccination may trigger the onset or exacerbation of existing pre-autoimmune conditions and develop autoimmune diseases such as RA, AAV or SLE. In most of reported cases in , the duration of vaccination and onset of the diseases is relatively short, that is, within a month, suggesting that the stimulation of existing pre-autoimmune conditions considered to be the main pathogenic factor. Padjen et al. [Citation63] analyzed the onset of arthralgia and arthritis following COVID-19 vaccination and reported that the mean time from vaccination to arthralgia onset ranged from 1.54 to 5.31 days and the mean time to arthritis onset ranged from 7.74 to 13.60 days. These results were similar to those of the cases we summarized here.

The clinical features of autoimmune disease following COVID-19 vaccination are reported to be short-lived and responsive to glucocorticoid and other immunosuppressive drugs, with a good prognosis [Citation64, Citation65]. Kim et al. investigated autoimmune disorders reported following COVID-19 vaccination using the World Health Organization (WHO) database [Citation66]. They identified 25,219 adverse events following immunizations (AEFIs) associated with autoimmune disorders following COVID-19 vaccination, including mRNA (n = 21,470) and viral vector (n = 3366) platforms in the WHO database. The number of AEFIs appeared to be two times higher in females than in males. The tendency for AEFI related to autoimmune disorders to be more prevalent in females was consistently observed in all other vaccine groups as well. They also reported that the age of patients who developed autoimmune disorders following COVID-19 vaccination was predominantly between 45 and 64 years old, while the age of patients who developed them after other vaccinations was mostly under 18 years old. Approximately half of the cases of autoimmune disorders following COVID-19 vaccination occurred within 7 days, whereas after other vaccinations, occurrences within 7 days and beyond 28 days were approximately 30% each. Among autoimmune diseases, ankylosing spondylitis or psoriatic arthritis, inflammatory bowel disease, polymyalgia rheumatica and thyroiditis were significantly more frequently reported to occur following COVID-19 vaccines than following all other vaccines. Meanwhile, the reported rates of RA, vasculitis and SLE were similar or lower after COVID-19 vaccines than after all other vaccines.

With regard to the number of vaccinations, in our cases and in the previously reported cases summarized in the Tables, RA and AAV tended to develop more after the second dose of mRNA vaccine and more after the first dose of Vector vaccine. In contrast, the incidence of SLE was similar after the first and second doses of mRNA vaccine. The most common type of vaccine was mRNA: 71.4% of RA cases, 79.5% of AAV cases and 84.2% of SLE cases. Further case accumulation is considered necessary to analyze trends according to the number of vaccinations and types of vaccine.

Risk factors for developing autoimmune disease after vaccination include a history of symptoms after vaccination in the past, history of allergy to vaccines, immunosuppressed state, family history of autoimmune disease or known autoantibodies, and genetic predisposition are presumed [Citation67, Citation68]. Therefore, individuals who develop symptoms of autoimmune disease after receiving the vaccine should consider the possibility that similar symptoms may occur when they receive the vaccine again.

We report three cases of new-onset rheumatic disease following COVID-19 vaccination. Although such adverse effects are rare, and vaccines are useful in prevent onset and severity of infections, continued accumulation of similar cases is important in terms of examining long-term safety. It may provide an opportunity to elucidate the pathogenic mechanism of autoimmune diseases and to clarify the risk factors and effective treatments in the future.

Ethical approval statement

Ethics approval for this study was obtained from the Institutional Review Board of Saitama Medical University Hospital (Approval Number: 2021-142).

Acknowledgment

We are grateful to Mei Hamada and Atsushi Sasaki (Pathology, Saitama Medical University, Saitama, Japan) for providing technical assistance.

Disclosure statement

The authors have no conflicts of interest.

References

  • Hoxha I, Agahi R, Bimbashi A, et al. Higher COVID-19 vaccination rates are associated with lower COVID-19 mortality: a global analysis. Vaccines (Basel). 2023;11(1):74. doi: 10.3390/vaccines11010074.
  • Wu N, Joyal-Desmarais K, Ribeiro PAB, et al. Long-term effectiveness of COVID-19 vaccines against infections, hospitalisations, and mortality in adults: findings from a rapid living systematic evidence synthesis and meta-analysis up to December 2022. Lancet Respir Med. 2023;11(5):439–452. doi: 10.1016/S2213-2600(23)00015-2.
  • Matsuda M, Asanuma YF, Yokota K, et al. New-onset adult-onset still’s disease following COVID-19 vaccination: three case reports and a literature review. Intern Med. 2022;62(2):299–305. doi: 10.2169/internalmedicine.0590-22.
  • Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–2581. doi: 10.1002/art.27584.
  • Aringer M, Costenbader K, Daikh D, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis. 2019;78(9):1151–1159. doi: 10.1136/annrheumdis-2018-214819.
  • Baimukhamedov C, Makhmudov S, Botabekova A. Seropositive rheumatoid arthritis after vaccination against SARS-CoV-2 infection. Int J Rheum Dis. 2021;24(11):1440–1441. doi: 10.1111/1756-185X.14220.
  • Yonezawa H, Ohmura S, Ohkubo Y, et al. New-onset seropositive rheumatoid arthritis following COVID-19 vaccination in a patient with seronegative status. Intern Med. 2022;61(22):3449–3452. doi: 10.2169/internalmedicine.0257-22.
  • Watanabe T, Minaga K, Hara A, et al. Case report: new-onset rheumatoid arthritis following COVID-19 vaccination. Front Immunol. 2022;13:859926. doi: 10.3389/fimmu.2022.859926.
  • Emran J, Menakuru S, Khan I, et al. Unilateral erosive arthritis following moderna COVID-19 vaccination. Cureus. 2022;14(5):e25020. doi: 10.7759/cureus.25020.
  • Singh R, Kaur U, Singh A, et al. Refractory hypereosinophilia associated with newly diagnosed rheumatoid arthritis following inactivated BBV152 COVID-19 vaccine. J Med Virol. 2022;94(8):3482–3487. doi: 10.1002/jmv.27742.
  • Nahra V, Makandura M, Mattar M, et al. A case series on the COVID-19 vaccines and possible immune-related adverse events: a new challenge for the rheumatologists. Cureus. 2022;14(9):e12492. doi: 10.7759/cureus.29660.
  • Villa M, Díaz-Crespo F, Pérez de José A, et al. A case of ANCA-associated vasculitis after AZD1222 (Oxford-AstraZeneca) SARS-CoV-2 vaccination: casualty or causality? Kidney Int. 2021;100(4):937–938. doi: 10.1016/j.kint.2021.07.026.
  • Shakoor MT, Birkenbach MP, Lynch M. ANCA-associated vasculitis following Pfizer-BioNTech COVID-19 vaccine. Am J Kidney Dis. 2021;78(4):611–613. doi: 10.1053/j.ajkd.2021.06.016.
  • Okuda S, Hirooka Y, Sugiyama M. Propylthiouracil-induced antineutrophil cytoplasmic antibody-associated vasculitis after COVID-19 vaccination. Vaccines (Basel). 2021;9(8):842. doi: 10.3390/vaccines9080842.
  • Hakroush S, Tampe B. Case report: ANCA-associated vasculitis presenting with rhabdomyolysis and pauci-immune crescentic glomerulonephritis after Pfizer-BioNTech COVID-19 mRNA vaccination. Front Immunol. 2021;12:762006. doi: 10.3389/fimmu.2021.762006.
  • Chen CC, Chen HY, Lu CC, et al. Case report: anti-neutrophil cytoplasmic antibody-associated vasculitis with acute renal failure and pulmonary hemorrhage may occur after COVID-19 vaccination. Front Med (Lausanne). 2021;8:765447. doi: 10.3389/fmed.2021.765447.
  • Prabhahar A, Naidu G, Chauhan P, et al. ANCA-associated vasculitis following ChAdOx1 nCoV19 vaccination: case-based review. Rheumatol Int. 2022;42(4):749–758. doi: 10.1007/s00296-021-05069-x.
  • Al-Yafeai Z, Horn BJM, Terraccaine W, et al. A case of antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis post COVID-19 vaccination. Cureus. 2022;14(3):e23162. doi: 10.7759/cureus.23162.
  • Baier E, Olgemöller U, Biggemann L, et al. Dual-positive MPO-and PR3-ANCA-associated vasculitis following SARS-CoV-2 mRNA booster vaccination: a case report and systematic review. Vaccines (Basel). 2022;10(5):653. doi: 10.3390/vaccines10050653.
  • Nappi E, De Santis M, Paoletti G, et al. New onset of eosinophilic granulomatosis with polyangiitis following mRNA-based COVID-19 vaccine. Vaccines (Basel). 2022;10(5):716. doi: 10.3390/vaccines10050716.
  • So D, Min KW, Jung WY, et al. Microscopic polyangiitis following mRNA COVID-19 vaccination: a case report. J Korean Med Sci. 2022;37(19):e154. doi: 10.3346/jkms.2022.37.e154.
  • Kim BC, Kim HS, Han KH, et al. A case report of MPO-ANCA-associated vasculitis following heterologous mRNA1273 COVID-19 booster vaccination. J Korean Med Sci. 2022;37(26):e204. doi: 10.3346/jkms.2022.37.e204.
  • Hasbani GE, Uthman I. ANCA-associated vasculitis following the first dose of Pfizer-BioNTech COVID-19 vaccine. Nephron. 2023;147(2):103–107. doi: 10.1159/000525562.
  • Noel E, Rashid U, Rabbani R, et al. Antineutrophil cytoplasmic autoantibody-associated glomerulonephritis as a possible side effect of COVID-19 vaccination. Cureus. 2022;14(10):e30565. doi: 10.7759/cureus.30565.
  • Christodoulou M, Iatridi F, Chalkidis G, et al. ANCA-associated vasculitis may result as a complication to both SARS-CoV-2 infection and vaccination. Life. 2022;12(7):1072. doi: 10.3390/life12071072.
  • Yadav R, Shah S, Chhetri S. ANCA-associated vasculitis following Johnson and Johnson COVID-19 vaccine. Ann Med Surg (Lond). 2022;79:104123. doi: 10.1016/j.amsu.2022.104123.
  • Suzuki M, Sekiguchi Y, Sasaki M, et al. Antineutrophil cytoplasmic antibody-associated vasculitis after COVID-19 vaccination with Pfizer-BioNTech. Intern Med. 2022;61(19):2925–2929. doi: 10.2169/internalmedicine.9807-22.
  • Ma Y, Huang T, Xu G. ANCA-associated vasculitis following the CoronaVac vaccination. Ther Adv Chronic Dis. 2022;13:1–5.
  • Obata S, Hidaka S, Yamano M, et al. MPO-ANCA-associated vasculitis after the Pfizer/BioNTech SARS-CoV-2 vaccination. Clin Kidney J. 2022;15(2):357–359. doi: 10.1093/ckj/sfab181.
  • Zamoner W, Scardini JB, Dio BJD, et al. ANCA-associated vasculitis following Oxford-AstraZeneca COVID-19 vaccine in Brazil: is there a causal relationship? A case report. Front Med (Lausanne). 2022;9:1003332. doi: 10.3389/fmed.2022.1003332.
  • Uddin K, Mohamed KH, Agboola AA, et al. Antineutrophil cytoplasmic antibody (ANCA)-associated renal vasculitis following COVID-19 vaccination: a case report and literature review. Cureus. 2022;14(10):e30206.
  • Anderegg MA, Liu M, Saganas C, et al. De novo vasculitis after mRNA-1273 (moderna) vaccination. Kidney Int. 2021;100(2):474–476. doi: 10.1016/j.kint.2021.05.016.
  • Dube GK, Benvenuto LJ, Batal I. Antineutrophil cytoplas-mic autoantibody-associated glomerulonephritis following the Pfzer-BioNTech COVID-19 vaccine. Kidney Int Rep. 2021;6(12):3087–3089. doi: 10.1016/j.ekir.2021.08.012.
  • Takenaka T, Matsuzaki M, Fujiwara S, et al. Myeloperoxidase anti-neutrophil cytoplasmic antibody positive optic perineuritis after mrna coronavirus disease-19 vaccine: a case report. QJM. 2021;114(10):737–738. doi: 10.1093/qjmed/hcab227.
  • Gupta RK, Ellis BK. Concurrent anti-GBM nephritis and ANCA-mediated glomerulonephritis after second dose of SARS-CoV-2 mRNA vaccination. Kidney Int Rep. 2022;7(1):127–128. doi: 10.1016/j.ekir.2021.10.020.
  • Klomjit N, Alexander MP, Fervenza FC, et al. COVID-19 vaccination and glomerulonephritis. Kidney Int Rep. 2021;6(12):2969–2978. doi: 10.1016/j.ekir.2021.09.008.
  • Prema J, Muthukumaran A, Haridas N, et al. Two cases of double-positive antineutrophil cytoplasmic autoantibody and antiglomerular basement membrane disease after BBV152/covaxin vaccination. Kidney Int Rep. 2021;6(12):3090–3091. doi: 10.1016/j.ekir.2021.10.004.
  • Caza TN, Cassol CA, Messias N, et al. Glomerular disease in temporal association to SARS-CoV-2 vaccination – a series of 29 cases. Kidney360. 2021;2(11):1770–1780. doi: 10.34067/KID.0005372021.
  • Davidovic T, Schimpf J, Sprenger-Mähr H, et al. De novo and relapsing glomerulonephritis following SARS-CoV-2 mRNA vaccination in microscopic polyangiitis. Case Rep Nephrol. 2021;2021:1–5. doi: 10.1155/2021/8400842.
  • Seif N, Ellis CL, Wadhwani S. ANCA-associated glomerulonephritis and vasculitis following COVID-19 vaccination in a patient with giant cell arteritis. J Am Soc Nephrol. 2021;32:87.
  • Feghali EJ, Zafar M, Abid S, et al. De-novo antineutrophil cytoplasmic antibody-associated vasculitis following the mRNA-1273 (Moderna) vaccine for COVID-19. Cureus. 2021;13(11):e19616. doi: 10.7759/cureus.19616.
  • Gen S, Iwai T, Ohnari S, et al. ANCA-associated vasculitis after moderna COVID-19 vaccination. Case Rep Nephrol. 2023;2023:4906876. doi: 10.1155/2023/4906876.
  • Hidaka D, Ogasawara R, Sugimura S, et al. New-onset evans syndrome associated with systemic lupus erythematosus after BNT162b2 mRNA COVID-19 vaccination. Int J Hematol. 2022;115(3):424–427. doi: 10.1007/s12185-021-03243-2.
  • Nune A, Iyengar KP, Ish P, et al. The emergence of new-onset SLE following SARS-CoV-2 vaccination. QJM. 2021;114(10):739–740. doi: 10.1093/qjmed/hcab229.
  • Báez-Negrón L, Vila LM. New-onset systemic lupus erythematosus after mRNA SARS-CoV-2 vaccination. Case Rep Rheumatol. 2022;2022:6436839. doi: 10.1155/2022/6436839.
  • Zavala-Miranda MF, Gonzalez-Ibarra SG, Pérez-Arias AA, et al. New-onset systemic lupus erythematosus beginning as class Ⅴ lupus nephritis after COVID-19 vaccination. Kidney Int. 2021;100:1334–1345.
  • Patil S, Patil A. Systemic lupus erythematosus after COVID-19 vaccination: a case report. J Cosmet Dermatol. 2021;20(10):3103–3104. doi: 10.1111/jocd.14386.
  • Kaur I, Zafar S, Capitle E, et al. COVID-19 vaccination as a potential trigger for new-onset systemic lupus erythematosus. Cureus. 2022;14(2):e21917.
  • Molina-Rios S, Rojas-Martinez R, Estévez-Ramirez GM, et al. Systemic lupus erythematosus and antiphospholipid syndrome after COVID-19 vaccination. A case report. Mod Rheumatol Case Rep. 2023;7(1):43–46. doi: 10.1093/mrcr/rxac018.
  • Sogbe M, Matteo ABD, Frisco IMD, et al. Systemic lupus erythematosus myocarditis after COVID-19 vaccination. Reumatol Clin. 2023;19(2):114–116.
  • Sagy I, Zeller L, Raviv Y, et al. New-onset systemic lupus erythematosus following BNT16b2 mRNA COVID-19 vaccine: a case series and literature review. Rheumatol Int. 2022;42(12):2261–2266. doi: 10.1007/s00296-022-05203-3.
  • Alrashdi Mousa A, Saleh AM, Khalid A, et al. Systemic lupus erythematosus with acute pancreatitis and vasculitic rash following COVID-19 vaccine: a case report and literature review. Clin Rheumatol. 2022;41:1577–1582.
  • Lemoine C, Padilla C, Krampe N, et al. Systemic lupus erythematosus after Pfizer COVID-19 vaccine: a case report. Clin Rheumatol. 2022;41(5):1597–1601. doi: 10.1007/s10067-022-06126-x.
  • Khanna U, Oprea Y, Mir A, et al. New diagnosis of systemic lupus erythematosus after COVID-19 vaccination: a case report and review of literature. JAAD Case Rep. 2022;30:30–34. doi: 10.1016/j.jdcr.2022.09.026.
  • Raviv Y, Betesh-Abay B, Valdman-Grinshpoun Y, et al. First presentation of systemic lupus erythematosus in a 24-year-old male following mRNA COVID-19 vaccine. Case Rep Rheumatol. 2022;2022:9698138. doi: 10.1155/2022/9698138.
  • Zengarini C, Pileri A, Salamone FP, et al. Subacute cutaneous lupus erythematosus induction after SARS-CoV-2 vaccine in a patient with primary biliary cholangitis. J Eur Acad Dermatol Venereol. 2022;36(3):e179–e180. doi: 10.1111/jdv.17827.
  • Kim HJ, Jung M, Lim BJ, et al. New-onset class III lupus nephritis with multi-organ involvement after COVID-19 vaccination. Kidney Int. 2022;101(4):826–828. doi: 10.1016/j.kint.2022.01.013.
  • Pellegrino P, Carnovale C, Pozzi M, et al. On the relationship between human papilloma virus vaccine and autoimmune diseases. Autoimmun Rev. 2014;13(7):736–741. doi: 10.1016/j.autrev.2014.01.054.
  • Segal Y, Shoenfeld Y. Vaccine-induced autoimmunity: the role of molecular mimicry and immune crossreaction. Cell Mol Immunol. 2018;15(6):586–594. doi: 10.1038/cmi.2017.151.
  • Olivieri B, Betterle C, Zanoni G. Vaccinations and autoimmune diseases. Vaccines (Basel). 2021;9(8):815. doi: 10.3390/vaccines9080815.
  • Teijaro JR, Farber DL. COVID-19 vaccines: modes of immune activation and future challenges. Nat Rev Immunol. 2021;21(4):195–197. doi: 10.1038/s41577-021-00526-x.
  • Pardi N, Hogan MJ, Porter FW, et al. mRNA vaccines—a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261–279. doi: 10.1038/nrd.2017.243.
  • Padjen EP, Marcec R, Zidar M, et al. Comparison of reporting rates of arthritis and arthralgia following AstraZeneca, Pfizer-BioNTech, Moderna, and Janssen vaccine administration against SARS-CoV-2 in 2021: analysis of European pharmacovigilance large-scale data. Rheumatol Int. 2023;44(2):273–281. doi: 10.1007/s00296-023-05512-1.
  • Jara LJ, Vera‑Lastra O, Mahroum N, et al. Autoimmune post-COVID vaccine syndromes: does the spectrum of autoimmune/infammatory syndrome expand? Clin Rheumatol. 2022;41(5):1603–1609. doi: 10.1007/s10067-022-06149-4.
  • Chen Y, Xu Z, Wang P, et al. New-onset autoimmune phenomena post-COVID-19 vaccination. Immunology. 2022;165(4):386–401. doi: 10.1111/imm.13443.
  • Kim S, Bea S, Choe S-A, et al. Autoimmune disorders reported following COVID-19 vaccination: a disproportionality analysis using the WHO database. Eur J Clin Pharmacol. 2024;80(3):445–453. doi: 10.1007/s00228-023-03618-w.
  • Soriano A, Nesher G, Shoenfeld Y. Predicting post-vaccination autoimmunity: who might be at risk? Pharmacol Res. 2015;92:18–22. doi: 10.1016/j.phrs.2014.08.002.
  • Vadalà M, Poddighe D, Laurino C, et al. Vaccination and autoimmune disease: is prevention of adverse health effects on the horizon? Epma J. 2017;8(3):295–311. doi: 10.1007/s13167-017-0101-y.