Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Allergy Clin Immunol ; 2023 Jun 07.
Article in English | MEDLINE | ID: covidwho-20236241

ABSTRACT

This guidance updates 2021 GRADE recomendations regarding immediate allergic reactions following COVID-19 vaccines and addresses re-vaccinating individuals with 1st dose allergic reactions and allergy testing to determine re-vaccination outcomes. Recent meta-analyses assessed the incidence of severe allergic reactions to initial COVID-19 vaccination, risk of mRNA-COVID-19 re-vaccination after an initial reaction, and diagnostic accuracy of COVID-19 vaccine and vaccine excipient testing in predicting reactions. GRADE methods informed rating the certainty of evidence and strength of recommenations. A modified Delphi panel consisting of experts in allergy, anaphylaxis, vaccinology, infectious diseases, emergency medicine, and primary care from Australia, Canada, Europe, Japan, South Africa, the UK, and the US formed the recommendations. We recommend vaccination for persons without COVID-19 vaccine excipient allergy, and re-vaccination after a prior immediate allergic reaction. We suggest against >15-minute post-vaccination observation. We recommend against mRNA vaccine or excipient skin testing to predict outcomes. We suggest re-vaccination of persons with an immediate allergic reaction to the mRNA vaccine or excipients be performed by a person with vaccine allergy expertise, in a properly equipped setting. We suggest against pre-medication, split-dosing, or special precautions because of a comorbid allergic history.

2.
Pediatr Neonatol ; 2023 May 24.
Article in English | MEDLINE | ID: covidwho-2327237

ABSTRACT

BACKGROUND: To investigate Pfizer-BioNTech 162b2 mRNA COVID-19 vaccine (BNT162b2) immunization-related myocarditis and describe the risk factors for consequent hospitalization in the pediatric intensive care unit (PICU) in children between 12 and 18 years. METHODS: Children and adolescents 12 years of age and older who presented with discomfort after BNT162b2 immunization (BNTI) and visited pediatric emergency room (PER) at Chang Gung Memorial Hospital from September 22, 2021 to March 21, 2022, were included for analysis. RESULTS: 681 children presented with discomfort after BNTI and visited our PER. The mean age was 15.1 ± 1.7 years. Three hundred and ninety-four (57.9%) and 287 (42.1%) events were after 1st and 2nd dose, respectively. 58.4% (n = 398) were male. The most common complaints were chest pain (46.7%) and chest tightness (27.0%). The median (interquartile range [IQR]) interval of discomfort after BNTI was 3.0 (1.0-12.0) days. BNTI-related pericarditis, myocarditis and myopericarditis were diagnosed in 15 (2.2%), 12 (1.8%) and 2 (0.3%) patients, respectively. Eleven (1.6%) needed hospitalization in PICU. The median (IQR) hospital stay was 4.0 (3.0-6.0) days. There was no mortality. More patients were diagnosed myocarditis (p = 0.004) after 2nd dose BNTI. PICU admission occurred more commonly after 2nd dose BNTI (p = 0.007). Risk factors associated with hospitalization in PICU were abnormal EKG findings (p = 0.047) and abnormal serum troponin levels (p = 0.003) at PER. CONCLUSION: Myocarditis in children aged 12-18 years occurred more commonly following 2nd dose BNTI. Most cases were of mild or intermediate severity without death. Factors predicting BNTI-related myocarditis and consequent hospitalization in PICU were abnormal EKG findings and abnormal serum troponin levels at PER in this study.

3.
Cureus ; 14(10): e30896, 2022 Oct.
Article in English | MEDLINE | ID: covidwho-2311740

ABSTRACT

A case of autoimmune hepatitis (AIH) following COVID-19 vaccination in a very old patient is presented. An 85-year-old woman who had preexisting Sjögren's syndrome (SS) but had never shown evidence of liver disease was admitted to our hospital due to jaundice and liver dysfunction. Further laboratory tests, imaging studies, and liver histology proved this to be a case of definite AIH. Eight weeks before the disease onset, she had received the second dose of mRNA COVID-19 vaccination. To our knowledge, this is the first case of AIH following COVID-19 vaccination in a patient with a history of SS.

4.
BMC Nephrol ; 24(1): 108, 2023 04 24.
Article in English | MEDLINE | ID: covidwho-2300385

ABSTRACT

BACKGROUND: As messenger RNA (mRNA)-based vaccines for coronavirus disease 2019 (COVID-19) have been administered to millions of individuals worldwide, cases of de novo and relapsing glomerulonephritis after mRNA COVID-19 vaccination are increasing in the literature. While most previous publications reported glomerulonephritis after the first or second dose of an mRNA vaccine, few reports of glomerulonephritis occurring after the third dose of an mRNA vaccine currently exist. CASE PRESENTATION: We report a case of rapidly progressive glomerulonephritis in a patient following the third dose of an mRNA COVID-19 vaccine. A 77-year-old Japanese man with a history of hypertension and atrial fibrillation was referred to our hospital for evaluation of anorexia, pruritus, and lower extremity edema. One year before referral, he received two mRNA vaccines (BNT162b2) for COVID-19. Three months before the visit, he received a third mRNA vaccine (mRNA-1273) for COVID-19. On admission, the patient presented severe renal failure with a serum creatinine level of 16.29 mg/dL, which had increased from 1.67 mg/dL one month earlier, prompting us to initiate hemodialysis. Urinalysis showed nephrotic-range proteinuria and hematuria. Renal biopsy revealed mild mesangial proliferation and expansion, a lobular appearance, and double contours of the glomerular basement membrane. Renal tubules had severe atrophy. Immunofluorescence microscopy showed strong mesangial staining for IgA, IgM, and C3c. Electron microscopy exhibited mesangial and subendothelial electron-dense deposits, leading to a diagnosis of IgA nephropathy with membranoproliferative glomerulonephritis-like changes. The kidney function remained unchanged after steroid therapy. CONCLUSIONS: Although the link between renal lesions and mRNA vaccines remains unclear, a robust immune response induced by mRNA vaccines may play a role in the pathogenesis of glomerulonephritis. Further studies of the immunological effects of mRNA vaccines on the kidney are warranted.


Subject(s)
COVID-19 , Glomerulonephritis, IGA , Glomerulonephritis, Membranoproliferative , Glomerulonephritis , Male , Humans , Aged , Glomerulonephritis, IGA/diagnosis , Glomerulonephritis, Membranoproliferative/pathology , COVID-19 Vaccines , BNT162 Vaccine , COVID-19/complications , Glomerulonephritis/pathology
5.
JMIR Public Health Surveill ; 9: e45263, 2023 Jul 14.
Article in English | MEDLINE | ID: covidwho-2304706

ABSTRACT

BACKGROUND: The World Health Organization recently described sudden sensorineural hearing loss (SSNHL) as a possible adverse effect of COVID-19 vaccines. Recent discordant pharmacoepidemiologic studies invite robust clinical investigations of SSNHL after COVID-19 messenger RNA (mRNA) vaccines. This postmarketing surveillance study, overseen by French public health authorities, is the first to clinically document postvaccination SSNHL and examine the role of potential risk factors. OBJECTIVE: This nationwide study aimed to assess the relationship between SSNHL and exposure to mRNA COVID-19 vaccines and estimate the reporting rate (Rr) of SSNHL after mRNA vaccination per 1 million doses (primary outcome). METHODS: We performed a retrospective review of all suspected cases of SSNHL after mRNA COVID-19 vaccination spontaneously reported in France between January 2021 and February 2022 based on a comprehensive medical evaluation, including the evaluation of patient medical history, side and range of hearing loss, and hearing recovery outcomes after a minimum period of 3 months. The quantification of hearing loss and assessment of hearing recovery outcomes were performed according to a grading system modified from the Siegel criteria. A cutoff of 21 days was used for the delay onset of SSNHL. The primary outcome was estimated using the total number of doses of each vaccine administered during the study period in France as the denominator. RESULTS: From 400 extracted cases for tozinameran and elasomeran, 345 (86.3%) spontaneous reports were selected. After reviewing complementary data, 49.6% (171/345) of documented cases of SSNHL were identified. Of these, 83% (142/171) of SSNHL cases occurred after tozinameran vaccination: Rr=1.45/1,000,000 injections; no difference for the rank of injections; complete recovery in 22.5% (32/142) of cases; median delay onset before day 21=4 days (median age 51, IQR 13-83 years); and no effects of sex. A total of 16.9% (29/171) of SSNHL cases occurred after elasomeran vaccination: Rr=1.67/1,000,000 injections; rank effect in favor of the first injection (P=.03); complete recovery in 24% (7/29) of cases; median delay onset before day 21=8 days (median age 47, IQR 33-81 years); and no effects of sex. Autoimmune, cardiovascular, or audiovestibular risk factors were present in approximately 29.8% (51/171) of the cases. SSNHL was more often unilateral than bilateral for both mRNA vaccines (P<.001 for tozinameran; P<.003 for elasomeran). There were 13.5% (23/142) of cases of profound hearing loss, among which 74% (17/23) did not recover a serviceable ear. A positive rechallenge was documented for 8 cases. CONCLUSIONS: SSNHL after COVID-19 mRNA vaccines are very rare adverse events that do not call into question the benefits of mRNA vaccines but deserve to be known given the potentially disabling impact of sudden deafness. Therefore, it is essential to properly characterize postinjection SSNHL, especially in the case of a positive rechallenge, to provide appropriate individualized recommendations.


Subject(s)
COVID-19 , Hearing Loss, Sensorineural , Hearing Loss, Sudden , Humans , Middle Aged , Hearing Loss, Sudden/etiology , COVID-19 Vaccines/adverse effects , BNT162 Vaccine , 2019-nCoV Vaccine mRNA-1273 , Pharmacovigilance , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/complications , Hearing Loss, Sensorineural/complications , Vaccination/adverse effects
6.
Public Health ; 219: 18-21, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2301937

ABSTRACT

OBJECTIVES: The aim of this research was to investigate the possible association between smoking habits and the incidence of adverse effects (AEs) after mRNA COVID-19 vaccine. STUDY DESIGN: A longitudinal observational study was conducted on a sample of Italian healthcare workers. METHODS: Healthcare workers who were administered the mRNA COVID-19 vaccine (either BNT162b2 or mRNA-1273) were evaluated for the occurrence of AEs after three vaccine doses. Multivariate Poisson regression analyses were fitted to predict AE risk according to smoking characteristics - such as number of tobacco cigarettes smoked per day, smoking time, and use of electronic cigarette (e-cig). RESULTS: Of 320 total participants, 72 (22.5%) smoked cigarettes, and 50 (15.6%) used e-cig, 49 of which being dual users. Tobacco smoking significantly increased the risks of muscle and joint pain during the primary COVID-19 vaccination cycle and of chills during the whole vaccination series. The number of cigarettes smoked per day and vaping variously predicted AE onset during the whole cycle, with a tendency to respectively reduce and increase their risks. Duration of smoking did not affect any AE, except for headache after the booster dose. Most results remained significant after Bonferroni adjustment of significance level. CONCLUSION: Our pilot study indicated a possible effect of smoking habits on AE onset. Our research offers evidence that helps understanding possible predictors of the interindividual variability in COVID-19 vaccine response, serving as a reference for further studies on the effect of smoking on vaccine safety and effectiveness.


Subject(s)
COVID-19 , Drug-Related Side Effects and Adverse Reactions , Electronic Nicotine Delivery Systems , Smoking Cessation , Vaccines , Humans , Smoking/epidemiology , COVID-19 Vaccines/adverse effects , Pilot Projects , Smoking Cessation/methods , BNT162 Vaccine , COVID-19/epidemiology , COVID-19/prevention & control , RNA, Messenger
7.
Cureus ; 15(2): e34534, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2275697

ABSTRACT

Cancer antigen 125 (CA-125) is a transmembrane glycoprotein, and it is known to be an essential biomarker in detecting treatment response and recurrence of ovarian cancer. It may also be used in monitoring colorectal cancer. It tends to rise in states of inflammation. Recent studies have demonstrated a temporary rise in CA-125 levels and other cancer biomarkers in patients suffering from coronavirus disease 2019 (COVID-19) infection. However, in the following case report, we hope to shed light on a possible association between CA-125 levels and the COVID-19 mRNA vaccine. We present the case of a 79-year-old woman with moderately differentiated adenocarcinoma of the right adnexa, who had a transient increase in CA-125 levels after a period, during which she underwent treatment for COVID-19 infection and received the first dose of COVID-19 mRNA (Pfizer-BioNTech) vaccine with no evidence of disease progression on imaging.

8.
Vaccine ; 41(14): 2382-2386, 2023 03 31.
Article in English | MEDLINE | ID: covidwho-2270600

ABSTRACT

AIM: The present study aimed to estimate the anaphylaxis rates following mRNA COVID-19 vaccination in children and adolescents in Europe. METHODS: We retrieved data on 371 anaphylaxis cases following mRNA COVID-19 vaccination in children ≤ 17 years old notified to EudraVigilance as of October 8, 2022. Overall, 27,120,512 doses of BNT162b2 vaccine and 1,400,300 doses of mRNA-1273 vaccine have been delivered to children during the study period. RESULTS: The overall mean anaphylaxis rate was 12.81 [95% confidence interval (CI): 11.49-14.12] per 106 mRNA vaccine doses [12.14 (95% CI: 6.37-17.91) per 106 doses for mRNA-1273 and 12.84 (95% CI: 11.49-14.19) per 106 doses for BNT162b2]. Children 12-17 years old accounted for 317 anaphylaxis cases, followed by 48 cases in children 3-11 years old, and 6 cases in children 0-2 years old. Children 10-17 years old had a mean anaphylaxis rate of 13.52 (95% CI: 12.03-15.00) cases per 106 mRNA vaccine doses and children 5-9 years old had a mean anaphylaxis rate of 9.51 (95% CI: 6.82-12.20) cases per 106 mRNA vaccine doses. There were two fatalities, both in the 12-17 years age group. The fatal anaphylaxis rate was 0.07 cases per 106 mRNA vaccine doses. CONCLUSIONS: Anaphylaxis is a rare adverse event after receiving an mRNA COVID-19 vaccine in children. Continuous surveillance of serious adverse events is needed to guide vaccination policies as we move towards SARS-CoV-2 endemicity. Larger real-world studies on COVID-19 vaccination in children, using clinical case confirmation, are imperative.


Subject(s)
Anaphylaxis , COVID-19 , Humans , Adolescent , Child , Child, Preschool , Infant, Newborn , Infant , Anaphylaxis/chemically induced , Anaphylaxis/epidemiology , 2019-nCoV Vaccine mRNA-1273 , BNT162 Vaccine , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , Vaccination/adverse effects , RNA, Messenger
9.
Medicina (Kaunas) ; 59(3)2023 Mar 21.
Article in English | MEDLINE | ID: covidwho-2249626

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is a universal emergency public health issue. A large proportion of the world's population has had several spike antigen exposures to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and/or COVID-19 vaccinations in a relatively short-term period. Although sporadic hematopoietic adverse events after COVID-19 vaccine inoculation were reported, there is currently no sufficient evidence correlating anti-spike protein immune responses and hematopoietic adverse events of vaccinations. We reported the first case of Ph-positive B-cell acute lymphoblastic leukemia (ALL) occurring after a bivalent mRNA COVID-19 vaccine inoculation. The otherwise healthy 43-year-old female patient had a total of six spike antigen exposures in the past 1.5 years. Informative pre-vaccine tests and bone marrow study results were provided. Although the causal relationship between bivalent vaccinations and the subsequent development of Ph-positive B-cell ALL cannot be determined in the case report, we propose that anti-spike protein immune responses could be a trigger for leukemia. Clinicians must investigate the hematopoietic adverse events closely after COVID-19 vaccinations. Further pre-clinical studies to investigate the safety of bivalent mRNA COVID-19 vaccine are required.


Subject(s)
COVID-19 , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Female , Humans , Adult , COVID-19 Vaccines/adverse effects , SARS-CoV-2 , COVID-19/prevention & control , Antibodies, Viral
10.
Rinsho Ketsueki ; 64(1): 35-41, 2023.
Article in Japanese | MEDLINE | ID: covidwho-2280501

ABSTRACT

Acquired hemophilia A (AHA) is a rare disease characteized by bleeding symptoms caused by decreased factor VIII activity due to the appearance of inhibitors to factor VIII triggered by malignancy or collagen disease. An 86-year-old woman developed purpura on her extremities after the first dose of the BNT162b2 mRNA COVID-19 vaccine. This symptom subsided after a few days. After the second dose of the BNT162b2 mRNA COVID-19 vaccine, purpura appeared again, and the patient was referred to our hospital Her APTT was remarkably prolonged to 110 seconds, and a cross-mixing test revealed an inhibitor pattern. Since FVIII activity was <1% and FVIII inhibitor was 51.6 BU, she was diagnosed with AHA. Prednisolone therapy was started, and coagulative complete remission was achieved. Because acquired hemophilia can develop after mRNA COVID-19 vaccination, as in this case, it is critical to monitor the appearance of bleeding symptom.


Subject(s)
BNT162 Vaccine , COVID-19 , Hemophilia A , Aged, 80 and over , Female , Humans , BNT162 Vaccine/adverse effects , COVID-19/prevention & control , COVID-19/complications , Hemophilia A/chemically induced , Hemophilia A/therapy , Hemorrhage
11.
Cureus ; 14(11): e31554, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2244224

ABSTRACT

Perimyocarditis is the inflammation of the pericardium along with the myocardium. Presentation is similar to acute pericarditis, but it is associated with myocardial damage, leading to an elevation in serum troponin and a left ventricular dysfunction (manifested as an ejection fraction of less than 55 percent). Perimyocarditis is mostly managed like acute myocarditis. Etiology is generally idiopathic and likely secondary to viral infections. Cases of vaccine-associated myocarditis have been infrequently reported in past, most recently with the COVID-19 mRNA vaccine. We present a rare case of a young healthy adolescent male who developed perimyocarditis after the first booster dose of the COVID-19 mRNA vaccine.

12.
J Card Fail ; 2022 Jul 13.
Article in English | MEDLINE | ID: covidwho-2245090

ABSTRACT

BACKGROUND: Numerous studies have reported myocarditis resulting from messenger RNA (mRNA) coronavirus disease 2019 (COVID-19) vaccination. However, to date, there have been no reports highlighting the safety of mRNA COVID-19 vaccines in children and adults with a prior history of myocarditis, which was the intent of this study. METHODS AND RESULTS: Children and adults cared for at the Cleveland Clinic were identified through the electronic health records, who had a history of myocarditis before the COVID-19 pandemic and had subsequently received at least 2 doses of the mRNA COVID-19 vaccines (n = 34). Only 1 patient in this series had recurrence of myocarditis confirmed by cardiac magnetic resonance imaging after receiving the second dose. He was a White man who had his first episode of myocarditis at age 20 and was 27 years of age at the time of recurrence. He was hospitalized for 2 days with no need for cardiac support or reported arrhythmias and was stable at outpatient follow-up. CONCLUSIONS: In patients with an old history of non-COVID-19 myocarditis, the risk of recurrent myocarditis after receipt of mRNA COVID-19 vaccination is low, and when it occurs it seems to be self-limiting. Our study will be valuable to clinicians while discussing the risk-benefit ratio of vaccinations in patients with a prior history of myocarditis.

13.
Allergy ; 2022 Nov 02.
Article in English | MEDLINE | ID: covidwho-2241112

ABSTRACT

For persons with immediate allergic reactions to mRNA COVID-19 vaccines, skin testing (ST) to the vaccine/excipients (polyethylene glycol[PEG] and polysorbate 80 [PS]) has been recommended, but has unknown accuracy. To assess vaccine/excipient ST accuracy in predicting all-severity immediate allergic reactions upon re-vaccination, systematic review was performed searching Medline, EMBASE, Web of Science, and the WHO global coronavirus database (inception-Oct 4, 2021) for studies addressing immediate (≤4 h post-vaccination) all-severity allergic reactions to 2nd mRNA COVID-19 vaccination in persons with 1st dose immediate allergic reactions. Cases evaluating delayed reactions, change of vaccine platform, or revaccination without vaccine/excipient ST were excluded. Meta-analysis of diagnostic testing accuracy was performed using Bayesian methods. The GRADE approach evaluated certainty of the evidence, and QUADAS-2 assessed risk of bias. Among 20 studies of mRNA COVID-19 first dose vaccine reactions, 317 individuals underwent 578 ST to any one or combination of vaccine, PEG, or PS, and were re-vaccinated with the same vaccine. Test sensitivity for either mRNA vaccine was 0.2 (95%CrI 0.01-0.52) and specificity 0.97 (95%CrI 0.9-1). PEG test sensitivity was 0.02 (95%CrI 0.00-0.07) and specificity 0.99 (95%CrI 0.96-1). PS test sensitivity was 0.03 (95%CrI 0.00-0.0.11) and specificity 0.97 (95%CrI 0.91-1). Combined for use of any of the 3 testing agents, sensitivity was 0.03 (95%CrI 0.00-0.08) and specificity was 0.98 (95%CrI 0.95-1.00). Certainty of evidence was moderate. ST has low sensitivity but high specificity in predicting all-severity repeat immediate allergic reactions to the same agent, among persons with 1st dose immediate allergic reactions to mRNA COVID-19 vaccines. mRNA COVID-19 vaccine or excipient ST has limited risk assessment utility.

14.
Journal of Cutaneous Immunology and Allergy ; 6(1):18-23, 2023.
Article in English | ProQuest Central | ID: covidwho-2237202

ABSTRACT

BackgroundSince the campaign of vaccination against COVID-19 was started, a wide variety of cutaneous adverse effects after vaccination has been documented worldwide. Varicella zoster virus (VZV) reactivation was reportedly the most frequent cutaneous reaction in men after administration of mRNA COVID-19 vaccines, especially BNT162b2.AimsA patient, who had persistent skin lesions after BNT162b2 vaccination for such a long duration over 3 months, was investigated for VZV virus and any involvement of vaccine-derived spike protein.Materials & MethodsImmunohistochemistry for detection of VZV virus and the spike protein encoded by mRNA COVID-19 vaccine. PCR analysis for VZV virus.ResultsThe diagnosis of VZV infection was made for these lesions using PCR analyses and immunohistochemistry. Strikingly, the vaccine-encoded spike protein of the COVID-19 virus was expressed in the vesicular keratinocytes and endothelial cells in the dermis.DiscussionmRNA COVID-19 vaccination might induce persistent VZV reactivation through perturbing the immune system, although it remained elusive whether the expressed spike protein played a pathogenic role.ConclusionWe presented a case of persistent VZV infection following mRNA COVID-19 vaccination and the presence of spike protein in the affected skin. Further vigilance of the vaccine side effect and investigation for the role of SP is warranted.

15.
Front Med (Lausanne) ; 9: 811829, 2022.
Article in English | MEDLINE | ID: covidwho-2215309

ABSTRACT

Background: A few studies on vaccination in patients with rheumatic diseases, including arthritis, connective tissue diseases, vasculitis, and psoriatic arthropathy (PsA), demonstrated reduced production of neutralizing antibodies to SARS-CoV-2 Spike RBD (receptor-binding domain contained in the N-terminal of the S1 globular head region) when compared to the general population. Objective: The aim of our study was to observe whether different therapies for PsA [methotrexate, anti-TNF antibodies, soluble TNF receptor (etanercept) or IL-17 inhibitors] have a different impact on SARS-CoV-2 vaccination in a homogeneous population of patients. Methods: We enrolled 110 PsA patients in remission, assessed with Disease Activity in PSoriatic Arthritis (DAPSA). Of these: 63 were in treatment with anti-TNF-α therapy (26 etanercept, 15 certolizumab, 5 golimumab, 17 adalimumab); 37 with anti-IL17 secukinumab; 10 with methotrexate. All patients underwent vaccination for SARS-CoV-2 with mRNA BNT162b2 vaccine. Assessment of absolute and percentage lymphocyte subsets and anti-SARS-CoV-2 Spike RBD IgG antibody value 3 weeks after the second vaccine dose were performed. In addition, the serum antibody levels of 96 healthy healthcare workers (HCW) were analyzed. Results: The mean disease activity assessed with DAPSA score was 2.96 (SD = 0.60) with no significant differences between patients under different medications (p = 0.779). Median levels of neutralizing antibodies to SARS-CoV-2 Spike RBD were 928.00 binding antibody unit (BAU)/mL [IQR 329.25, 1632.0]; 1068.00 BAU/ml [IQR 475.00, 1632.00] in patients taking MTX, 846.00 BAU/ml [IQR 125.00, 1632.00] in patients taking etanercept, 908.00 BAU/mL [IQR 396.00, 1632.00] in patients taking anti-IL17 and 1148.00 BAU/ml [IQR 327.00, 1632.00] in patients taking TNF-α inhibitors, without statistically significant differences between these groups. Mean serum antibody level of HCW group was 1562.00 BAU/ml [IQR 975.00, 1632.00], being significantly higher than in the patient group (p = 0.000816). Absolute and percentage count of lymphocyte subsets were not statistically different between the subgroups under different treatments and when compared with HCW. Conclusions: As for other rheumatic diseases on immunomodulatory treatment, our data showed a reduced humoral response in PsA patients compared to the control group. However, antibody response did not significantly differ between groups treated with different medications.

16.
BMC Infect Dis ; 23(1): 5, 2023 Jan 05.
Article in English | MEDLINE | ID: covidwho-2196094

ABSTRACT

BACKGROUND: The Pfizer BioNTech COVID-19 vaccine was the first to receive emergency authorization and approval from the FDA. Therefore, it is preferred by most recipients; however, many people are concerned about the vaccine's side effects. At the time of the study, December 2021, Palestine lacked a national reporting system for monitoring adverse vaccine effects. Therefore, this study investigates the post-vaccine adverse events following the Pfizer/BioNTech COVID-19 Vaccine administration in Palestine and identifies the occurrence, extent, and severity among university staff, employees, and students at Birzeit University. METHOD: A questionnaire-based retrospective cross-sectional study was conducted using a university website (Ritaj), social media platforms (e.g., Facebook and Telegram), and in-person interviews. The Chi-square, Fisher's exact, and McNemar's tests were used to investigate significant relationships. Data were analyzed using SPSS version 22. RESULTS: In total, 1137 participants completed the questionnaire, 33.2% were males, and the mean age was 21.163 years. All participants received at least one dose of the Pfizer-BioNTech COVID-19 vaccine. Approximately one-third of participants reported no adverse effects after receiving the first, second, or third doses (34%, 33.6%, and 32.5%, respectively). The most commonly reported adverse events were fever, chills, headache, fatigue, pain and swelling at the injection site, muscle pain, and joint pain. Allergic reactions were reported by 12.7% of the participants; furthermore, participants with a history of allergy or anaphylaxis before vaccination had a significantly higher tendency for post-vaccination allergic reactions. Eight participants reported rare side effects, including 7 (0.6%) cases of thrombocytopenia and one (0.1%) case of myocarditis. Males aged less than 20 years and smokers were significantly less likely to complain of adverse events. The number of reported side effects was significantly higher after the second vaccine dose than after the first dose. Finally, participants infected with COVID-19 before vaccination was significantly associated with side effects such as fever, chills, shortness of breath, and persistent cough. CONCLUSION: In this study, the most common post- BNT162b2 Vaccination reported self-limiting side effects similar to those reported by Pfizer/BioNTech Company. However, higher rates of allergic reactions were reported in this sample. Rare side effects, such as thrombocytopenia and myocarditis, were reported by 8 participants. COVID vaccines have been developed at an accelerated pace, and vaccine safety is a top priority; therefore, standard monitoring through a national adverse event reporting system is necessary for safety assurance. Continuous monitoring and long-term studies are required to ensure vaccine safety.


Subject(s)
COVID-19 Vaccines , COVID-19 , Drug-Related Side Effects and Adverse Reactions , Hypersensitivity , Myocarditis , Adult , Female , Humans , Male , Young Adult , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Cross-Sectional Studies , Retrospective Studies , Universities
17.
World J Clin Cases ; 11(1): 177-186, 2023 Jan 06.
Article in English | MEDLINE | ID: covidwho-2203809

ABSTRACT

BACKGROUND: A 70-year-old man with hepatitis C virus-related recurrent hepatocellular carcinoma was admitted for further diagnosis of a 1 cm iso-hyperechoic nodule in segment (S) 5. CASE SUMMARY: Gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid-enhanced magnetic resonance imaging (EOB-MRI) revealed the nodule in S5 with a defect at the hepatobiliary phase, hyperintensity on diffusion weighted imaging (DWI) and hypointensity on apparent diffusion coefficient (ADC) map. Contrast-enhanced computed tomography revealed hypervascularity at the early phase, and delayed contrast-enhancement was observed at the late phase. Contrast-enhanced ultrasound (US) revealed incomplete defect at the late vascular phase. Inflammatory liver tumor, lymphoproliferative disease, intrahepatic cholangiocarcinoma (small duct type) and bile duct adenoma were suspected through the imaging studies. US guided biopsy, however, showed a noncaseating hepatic sarcoid-like epithelioid granuloma (HSEG), and histopathological analysis disclosed spindle shaped epithelioid cells harboring Langhans-type multinucleated giant cells. One month after admission, EOB-MRI signaled the disappearance of the defect at the hepatobiliary phase, of hyperintensity on DWI, of hypointensity on ADC map, and no stain at the early phase. CONCLUSION: That the patient had received BNT162b2 messenger RNA (mRNA) coronavirus disease 2019 vaccination 3 mo before the occurrence of HSEG, and that its disappearance was confirmed 4 mo after mRNA vaccination suggested that the drug-induced sarcoidosis-like reaction (DISR) might be induced by the mRNA vaccination. Fortunately, rechallenge of drug-induced DISR with the third mRNA vaccination was not confirmed.

18.
Vaccines (Basel) ; 11(2)2023 Jan 22.
Article in English | MEDLINE | ID: covidwho-2200991

ABSTRACT

The successful deployment of safe and effective vaccines against coronavirus disease 2019 (COVID-19) has been crucial in reducing the global disease burden. Owing to the need for vaccination series over time, continuous observational studies are needed to estimate the COVID-19 vaccine response in real-world conditions. In particular, the detection, assessment, and understanding of adverse effects following immunization (AEFI) with a COVID-19 vaccine are crucial to better address vaccination strategies. Therefore, this study aimed to investigate the risk of repeated AEFI post-administration of a booster dose of mRNA COVID-19 vaccine in a sample of healthcare workers (HCWs) in an Italian teaching hospital. The data on any local and systemic AEFI were studied in multivariate Poisson regression analyses to model the association between the incidence of each postvaccination symptom and its prior reporting after the administration of the previous doses. Overall, compared with the primary vaccination series, the majority of post-third dose AEFI were less reported. The results from multivariable models showed that the likelihood of reporting an AEFI after the third dose was higher in those who experienced the same postvaccination symptom after the second dose (all AEFI except for itch at injection site) and, although not significant for all AEFI, after the first dose. Any associations with age, gender, smoking habits, previous SARS-CoV-2 infection and other characteristics, as well as the health impact of AEFI were also assessed. Taken together, the results from this research support reframe AEFI symptoms as signals of a robust postvaccination reaction as well as of common vaccine response, and they add important data to inform booster vaccination strategies in HCWs and, extensively, in the adult population.

19.
Microorganisms ; 10(11)2022 Nov 16.
Article in English | MEDLINE | ID: covidwho-2116178

ABSTRACT

The effect of a third vaccine dose (3D) of homologous mRNA vaccine on blood levels of SARS-CoV-2-receptor binding domain (RBD)-total antibodies was assessed in 40 hemodialysis patients (HD) and 21 kidney transplant recipients (KTR) at a median of 46 days after 3D. Anti-RBD antibodies were detected in 39/40 HD and 19/21 KTR. Overall, 3D boosted anti-RBD antibody levels (median: 58-fold increase). Neutralizing antibodies (NtAb) against the Wuhan-Hu-1, Delta, and Omicron variants were detected in 14, 13, and 11 out of 14 HD patients, and in 5, 5, and 4 out of 8 KTR patients, respectively. The median fold increase in NtAb titers in HD patients was 77, 28, and 5 and 56, 37, and 9 in KTR patients for each respective variant. SARS-CoV-2-S S-IFN-γ-producing CD8+ and CD4+ T-cell responses were detected in the majority of HD (35 and 36/37, respectively) and all KTR (16/16) patients at 3D. Overall, the administration of 3D boosted T-cell levels in both population groups. In conclusion, a homologous mRNA COVID-19 vaccine 3D exerts a booster effect on anti-RBD antibodies, NtAb binding to Wuhan-Hu-1, Delta, and Omicron variants, and SARS-CoV-2-S-IFN-γ-producing T cells in both HD and KTR patients. The magnitude of the effect was more marked in HD than KTR patients.

20.
Rinsho Ketsueki ; 63(10): 1379-1385, 2022.
Article in Japanese | MEDLINE | ID: covidwho-2110946

ABSTRACT

There is currently no evidence that a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine might be associated with the development of autoimmune hemolytic anemia or disease progression in patients with mature B-cell neoplasm. Our patient was a 71-year-old man with indolent mature B-cell neoplasm who had been monitored for many years without treatment. After receiving the second dose of the BNT162b2 mRNA COVID-19 vaccine, he developed severe warm autoimmune hemolytic anemia. Although steroid therapy improved his anemia, he continued to develop IgM-monoclonal gammopathy, renal insufficiency, and splenomegaly. He was diagnosed with splenic marginal zone lymphoma after undergoing splenectomy. The splenectomy improved the patient's symptoms. We assessed his SARS-CoV-2 specific antibody response, but the patient's serologic response to the vaccine was impaired. In patients with mature B-cell neoplasm, a non-specific immune response after vaccination might be associated with paraneoplastic syndromes.


Subject(s)
Anemia, Hemolytic, Autoimmune , COVID-19 Vaccines , COVID-19 , Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, B-Cell , Paraproteinemias , Splenic Neoplasms , Aged , Humans , Male , Anemia, Hemolytic, Autoimmune/etiology , Anemia, Hemolytic, Autoimmune/complications , BNT162 Vaccine , COVID-19/complications , COVID-19 Vaccines/adverse effects , Immunoglobulin M , Leukemia, Lymphocytic, Chronic, B-Cell/complications , Lymphoma, B-Cell/drug therapy , Paraproteinemias/complications , SARS-CoV-2 , Splenic Neoplasms/complications
SELECTION OF CITATIONS
SEARCH DETAIL