Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Rheumatol Int ; 43(9): 1621-1627, 2023 Sep.
Article in English | MEDLINE | ID: covidwho-20241087

ABSTRACT

OBJECTIVES: To characterize the antibody response to COVID-19 mRNA vaccination in patients with Systemic Lupus Erythematosus (SLE) and identify predictors of poor response. METHODS: SLE patients who are followed at the Beth Israel Deaconess Medical Center Lupus Cohort (BID-LC) were enrolled. SARS-CoV-2 IgG Spike antibody was measured in patients who received two doses of either the BNT162b2 (Pfizer-BioNTech) or the mRNA-1273 (Moderna) COVID-19 vaccine (n = 62). We defined non-responders as patients with an IgG Spike antibody titer less than two-fold (< 2) the index value of the test and responders as patients with antibody levels greater or equal to two-fold (≥ 2). A web-based survey was used to collect information regarding immunosuppressive medication use and SLE flares after vaccination. RESULTS: In our cohort of lupus patients, 76% were vaccine responders. The use of two or more immunosuppressive drugs was associated with being a non-responder (Odds Ratio 5.26; 95% CI 1.23-22.34, p = 0.02). Both Belimumab use and higher Prednisone dose were associated with vaccine non-response (p = 0.04 and p = 0.04). The non-responder group had higher mean levels of serum IL-18 than the responder group (p = 0.04) as well as lower C3 levels (p = 0.01). Lupus flares and breakthrough infections were uncommon post-vaccination. CONCLUSIONS: Immunosuppressive medications have a negative impact on vaccine humoral response in SLE individuals. We observed a trend towards vaccine no-response in BNT162b2 recipients and a relationship between IL-18 and impaired antibody response that merits further investigation.


Subject(s)
COVID-19 , Lupus Erythematosus, Systemic , Humans , COVID-19 Vaccines , BNT162 Vaccine , Interleukin-18 , Antibody Formation , COVID-19/prevention & control , SARS-CoV-2 , Antibodies, Viral , Immunoglobulin G , Vaccination
2.
Biomedicines ; 11(5)2023 Apr 28.
Article in English | MEDLINE | ID: covidwho-20233270

ABSTRACT

Coronavirus-disease-2019 (COVID-19) mRNA vaccination effectively reduces mortality and morbidity in cirrhotic patients, but the immunogenicity and safety of vaccination have been partially characterized. The study aimed to evaluate humoral response, predictive factors, and safety of mRNA-COVID-19 vaccination in cirrhotic patients compared to healthy subjects. A prospective, single-center, observational study enrolled consecutive cirrhotic patients who underwent mRNA-COVID-19 vaccination from April to May 2021. Anti-spike-protein (anti-S) and nucleocapsid-protein (anti-N) antibodies were evaluated before the first (T0) and the second (T1) doses and 15 days after completing the vaccination. An age and sex-matched healthy reference group was included. The incidence of adverse events (AEs) was assessed. In total, 162 cirrhotic patients were enrolled, 13 were excluded due to previous SARS-CoV-2 infection; therefore, 149 patients and 149 Health Care Workers (HCWs) were included in the analysis. The seroconversion rate was similar in cirrhotic patients and HCWs at T1 (92.5% vs. 95.3%, p = 0.44) and T2 (100% in both groups). At T2, anti-S-titres were significantly higher in cirrhotic patients compared to HCWs (2776.6 vs. 1756 BAU/mL, p < 0.001]. Male sex (ß = -0.32 [-0.64, -0.04], p = 0.027) and past-HCV-infection (ß = -0.31 [-0.59, -0.04], p = 0.029) were independent predictors of lower anti-S-titres on multiple-gamma-regression-analysis. No severe AEs occurred. The COVID-19-mRNA vaccination induces a high immunization rate and anti-S-titres in cirrhotic patients. Male sex and past-HCV infection are associated with lower anti-S-titres. The COVID-19-mRNA vaccination is safe.

3.
Clin Case Rep ; 11(5): e7354, 2023 May.
Article in English | MEDLINE | ID: covidwho-2324244

ABSTRACT

Venous thrombosis is a rare occurrence following the administration of the COVID-19 mRNA vaccine. The occurrence of superior mesenteric vein (SMV) is even more rare. SMV thrombosis should be considered as a differential diagnosis in patients who develop abdominal pain after COVID-19 mRNA vaccination.

4.
Mod Rheumatol Case Rep ; 2023 May 11.
Article in English | MEDLINE | ID: covidwho-2319715

ABSTRACT

BACKGROUND: According to previous reports, most cases of inflammatory myopathy following mRNA vaccination can be classified as idiopathic inflammatory myopathy (IIM), particularly dermatomyositis (DM), owing to their similar clinical features and courses. However, some patients have different clinical features and courses. We report a rare case of transient inflammatory myopathy involving the masseter muscle following the third dose of COVID-19 mRNA vaccination. CASE PRESENTATION: An 80-year-old woman presented with a history of fever and fatigue for 3 months soon after receiving the third COVID-19 mRNA vaccination. Her symptoms progressed to jaw pain and inability to open her mouth. She also experienced mild proximal muscle weakness in the lower limbs but no skin manifestations or daily difficulties. Fat-saturated T2-weighted magnetic resonance imaging showed bilateral high-intensity signals for the masseter and quadriceps muscles. The patient experienced spontaneous resolution of fever and improvement of symptoms 5 months after onset. The timing of the onset of symptoms, the lack of detectable autoantibodies, and the atypical presentation of myopathy in the masseter muscles, in addition to the spontaneous mild course of the disease, all indicate the substantial role of mRNA vaccination in this myopathy. Since then, the patient has been followed up for 4 months without any recurrence of symptoms or any additional treatment. CONCLUSION: It is important to recognize that the course of myopathy after COVID-19 mRNA vaccination could be different from that of typical IIMs.

5.
Front Immunol ; 13: 953949, 2022.
Article in English | MEDLINE | ID: covidwho-2316700

ABSTRACT

Two doses of BNT162b2 mRNA vaccine induces a strong systemic SARS-CoV-2 specific humoral response. However, SARS-CoV-2 airborne transmission makes mucosal immune response a crucial first line of defense. Therefore, we characterized SARS-CoV-2-specific IgG responses induced by BNT162b2 vaccine, as well as IgG responses to other pathogenic and seasonal human coronaviruses in oral fluid and plasma from 200 UK healthcare workers who were naïve (N=62) or previously infected with SARS-CoV-2 (N=138) using a pan-coronavirus multiplex binding immunoassay (Meso Scale Discovery®). Additionally, we investigated the impact of historical SARS-CoV-2 infection on vaccine-induced IgG, IgA and neutralizing responses in selected oral fluid samples before vaccination, after a first and second dose of BNT162b2, as well as following a third dose of mRNA vaccine or breakthrough infections using the same immunoassay and an ACE2 inhibition assay. Prior to vaccination, we found that spike-specific IgG levels in oral fluid positively correlated with IgG levels in plasma from previously-infected individuals (Spearman r=0.6858, p<0.0001) demonstrating that oral fluid could be used as a proxy for the presence of plasma SARS-CoV-2 IgG. However, the sensitivity was lower in oral fluid (0.85, 95% CI 0.77-0.91) than in plasma (0.94, 95% CI 0.88-0.97). Similar kinetics of mucosal and systemic spike-specific IgG levels were observed following vaccination in naïve and previously-infected individuals, respectively. In addition, a significant enhancement of OC43 and HKU1 spike-specific IgG levels was observed in previously-infected individuals following one vaccine dose in oral fluid (OC43 S: p<0.0001; HKU1 S: p=0.0423) suggesting cross-reactive IgG responses to seasonal beta coronaviruses. Mucosal spike-specific IgA responses were induced by mRNA vaccination particularly in previously-infected individuals (71%) but less frequently in naïve participants (23%). Neutralizing responses to SARS-CoV-2 ancestral and variants of concerns were detected following vaccination in naïve and previously-infected participants, with likely contribution from both IgG and IgA in previously-infected individuals (correlations between neutralizing responses and IgG: Spearman r=0.5642, p<0.0001; IgA: Spearman r=0.4545, p=0.0001). We also observed that breakthrough infections or a third vaccine dose enhanced mucosal antibody levels and neutralizing responses. These data contribute to show that a previous SARS-CoV-2 infection tailors the mucosal antibody profile induced by vaccination.


Subject(s)
COVID-19 , Viral Vaccines , Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , Humans , Immunoglobulin A , Immunoglobulin G , RNA, Messenger , SARS-CoV-2 , Vaccination , Vaccines, Synthetic , mRNA Vaccines
7.
Med ; 4(6): 353-360.e2, 2023 Jun 09.
Article in English | MEDLINE | ID: covidwho-2301231

ABSTRACT

BACKGROUND: Post-mRNA vaccination-associated cardiac complication is a rare but life-threatening adverse event. Its risk has been well balanced by the benefit of vaccination-induced protection against severe COVID-19. As the rate of severe COVID-19 has consequently declined, future booster vaccination to sustain immunity, especially against infection with new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, may encounter benefit-risk ratios that are less favorable than at the start of the COVID-19 vaccination campaign. Understanding the pathogenesis of rare but severe vaccine-associated adverse events to minimize its risk is thus urgent. METHODS: Here, we report a serendipitous finding of a case of cardiac complication following a third shot of COVID-19 mRNA vaccine. As this case was enrolled in a cohort study, pre-vaccination and pre-symptomatic blood samples were available for genomic and multiplex cytokine analyses. FINDINGS: These analyses revealed the presence of subclinical chronic inflammation, with an elevated expression of RNASE2 at pre-booster baseline as a possible trigger of an acute-on-chronic inflammation that resulted in the cardiac complication. RNASE2 encodes for the ribonuclease RNase2, which cleaves RNA at the 3' side of uridine, which may thus remove the only Toll-like receptor (TLR)-avoidance safety feature of current mRNA vaccines. CONCLUSIONS: These pre-booster and pre-symptomatic gene and cytokine expression data provide unique insights into the possible pathogenesis of vaccine-associated cardiac complication and suggest the incorporation of additional nucleoside modification for an added safety margin. FUNDING: This work was funded by the NMRC Open Fund-Large Collaborative Grant on Integrated Innovations on Infectious Diseases (OFLCG19May-0034).


Subject(s)
COVID-19 Vaccines , COVID-19 , Humans , COVID-19 Vaccines/adverse effects , Cohort Studies , COVID-19/prevention & control , SARS-CoV-2/genetics , mRNA Vaccines , Cytokines , Inflammation
8.
J Cancer Res Clin Oncol ; 2023 Apr 26.
Article in English | MEDLINE | ID: covidwho-2297200

ABSTRACT

Messenger RNA (mRNA) vaccination has proven to be highly successful in combating Coronavirus disease 2019 (COVID-19) and has recently sparked tremendous interest. This technology has been a popular topic of research over the past decade and is viewed as a promising treatment strategy for cancer immunotherapy. However, despite being the most prevalent malignant disease for women worldwide, breast cancer patients have limited access to immunotherapy benefits. mRNA vaccination has the potential to convert cold breast cancer into hot and expand the responders. Effective mRNA vaccine design for in vivo function requires consideration of vaccine targets, mRNA structures, transport vectors, and injection routes. This review provides an overview of pre-clinical and clinical data on various mRNA vaccination platforms used for breast cancer treatment and discusses potential approaches to combine appropriate vaccination platforms or other immunotherapies to improve mRNA vaccine therapy efficacy for breast cancer.

9.
Front Immunol ; 14: 1155728, 2023.
Article in English | MEDLINE | ID: covidwho-2305968

ABSTRACT

The advent of mRNA vaccines represents a significant advance in the field of vaccinology. While several vaccine approaches (mRNA, DNA, recombinant protein, and viral-vectored vaccines) had been investigated at the start of the COVID-19 pandemic, mRNA vaccines quickly gained popularity due to superior immunogenicity at a low dose, strong safety/tolerability profiles, and the possibility of rapid vaccine mass manufacturing and deployment to rural regions. In addition to inducing protective neutralizing antibody responses, mRNA vaccines can also elicit high-magnitude cytotoxic T-cell responses comparable to natural viral infections; thereby, drawing significant interest from cancer immunotherapy experts. This mini-review will highlight key developmental milestones and lessons we have learned from mRNA vaccines during the COVID-19 pandemic, with a specific emphasis on clinical trial data gathered so far for mRNA vaccines against melanoma and other forms of cancer.


Subject(s)
COVID-19 , Melanoma , Viral Vaccines , Humans , COVID-19/prevention & control , COVID-19 Vaccines , SARS-CoV-2 , RNA, Messenger/genetics , Pandemics/prevention & control , Antibodies, Neutralizing , Antibodies, Viral
10.
Cancer Med ; 12(12): 13135-13144, 2023 06.
Article in English | MEDLINE | ID: covidwho-2304425

ABSTRACT

BACKGROUND: The recently developed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine has a short history of use and further information is needed regarding its efficacy, especially in immunocompromised conditions, such as plasma cell dyscrasia (PCD). METHODS: We retrospectively measured serum SARS-CoV-2 antibodies against the spike protein (S-IgG) after the second and third mRNA vaccine doses (doses 2 and 3, respectively) in 109 patients with PCD. We evaluated the proportion of patients with an adequate humoral response (defined as S-IgG titers ≥300 antibody units/mL). RESULTS: Although active anti-myeloma treatments prior to vaccination had a significantly negative impact on adequate humoral response, specific drug subclasses including immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies were not negatively associated, except for B-cell maturation antigen-targeted therapy. Dose 3 (booster vaccination) led to significantly higher S-IgG titers and more patients acquired an adequate humoral response. Furthermore, evaluation of vaccine-induced cellular immune response in patients using T-spot Discovery SARS-CoV-2 kit, revealed an enhanced cellular immune response after Dose 3. CONCLUSIONS: This study highlighted the significance of booster SARS-CoV-2 mRNA vaccination in patients with PCD with respect to humoral and cellular immunity. Moreover, this study highlighted the potential impact of certain drug subclasses on vaccine-induced humoral immune response.


Subject(s)
COVID-19 , Paraproteinemias , Vaccines , Humans , SARS-CoV-2 , Retrospective Studies , COVID-19/prevention & control , Antibodies, Monoclonal , Antibodies, Viral , Immunity, Cellular , Immunoglobulin G
11.
Rev Med Virol ; 33(4): e2451, 2023 07.
Article in English | MEDLINE | ID: covidwho-2294883

ABSTRACT

People living with HIV (PLWH) are susceptible to severe COVID-19 infection and hence this fragile population has prioritised vaccination. This systematic review and meta-analysis aimed to assess the humoral immune response after receiving two doses schedule of COVID-19 mRNA vaccinations in this high-risk population. A systematic electronic search on the PubMed database and manual searches were performed for relevant articles until 30 Sep 2022. Two outcomes of interest were seroconversion rates and anti-spike receptor binding domain (anti-S-RBD) antibody titres at the median time of 14-35 days following two-dose vaccination among PLWH. Nineteen cohorts and one cross-sectional study were eligible for inclusion in this study. The pooled estimate of seroconversion rate after receiving two doses of mRNA vaccination schedule were 98.4% and 75.2% among PLWH with CD4>500 cells/mm3 and CD4<200 cells/mm3 , respectively. Compared with controls, PLWH with CD4>500 cells/mm3 had a 51% likelihood of having positive anti-Spike-RBD immunoglobulin G (IgG) (OR: 0.509, 95% CI: 0.228, 1.133, p = 0.098) post-vaccination and this value was only 1.4% (OR: 0.014, 95% CI: 0.002, 0.078, p = 0.000) for PLWH with CD4<200 cells/mm3 . There was no significant difference in titres of antibodies on 14-35 days post-vaccination between PLWH with CD4>500 cells/mm3 and healthy controls (p = 0.06). The pooled median of anti-S-RBD IgG values were 1461.93 binding antibody units (BAU)/ml and 457.41 BAU/ml in PLWH with CD4>500 cells/mm3 and CD4<200 cells/mm3 , respectively. According to these findings, vaccination with both Pfizer-BioNTech and Moderna vaccines induced a robust humoral response in ART-treated HIV patients with preserved CD4 cell count. A diminished humoral immune response to vaccination against COVID-19 in PLWH with unrestored CD4 count implied the need of specific vaccination schemes.


Subject(s)
COVID-19 , HIV Infections , Humans , Immunity, Humoral , COVID-19/prevention & control , Cross-Sectional Studies , Immunoglobulin G , Vaccination , Antibodies, Viral
12.
Case Rep Ophthalmol ; 14(1): 173-179, 2023.
Article in English | MEDLINE | ID: covidwho-2293254

ABSTRACT

Thromboembolic events as a result of COVID-19 mRNA vaccination are a rare, though life-threatening complication. In this case report, we describe a 40-year-old female patient who developed central retinal artery and ophthalmic artery occlusion progressing to intracranial thrombosis 3 weeks after vaccination with the Pfizer-BioNTech COVID-19 vaccine. Initially, she presented with progressive acute and painless unilateral vision loss in her left eye. Dilated fundoscopy of left eye showed macular whitening with sparing of the area of cilioretinal artery distribution. Labs revealed a normal erythrocyte sedimentation rate, C-reactive protein, and platelet count. Computerized tomography angiography of the head and neck showed an occlusion of the entire left cervical internal carotid artery and occlusion of the origin of the left external carotid artery. Despite treatment with heparin, her vision declined to no light perception. Ten days later, the patient presented with right peripheral vision loss and was found to have a new left posterior cerebral artery/posterior inferior cerebellar artery stroke. Seventeen days later, she presented to the hospital with nausea and vertigo and was found to have a subacute infarction in the left parietal lobe corresponding to left anterior communicating artery/middle cerebral artery watershed territory. Hypercoagulable disorders, vasculitis, cardiac arrhythmias, and intraventricular thrombi were excluded. Fundus fluorescein angiography confirmed central retinal artery occlusion and ophthalmic artery occlusion with impressive retina and choroid changes in fluorescein angiography patterns. This complication of mRNA COVID-19 vaccination has not been previously described in the literature and should be considered even weeks after initial presentation.

13.
JGH Open ; 7(4): 321-322, 2023 Apr.
Article in English | MEDLINE | ID: covidwho-2263855

ABSTRACT

A healthy 49-year-old female developed intractable watery diarrhea after the Pfizer SARS-Cov2 mRNA vaccination. She was subsequently diagnosed with collagenous colitis (CC). She had no prior history of medication use, suggesting of vaccination being the trigger. CC or lymphocytic colitis should be considered as differential diagnoses for persistent watery diarrhea after SARS-Cov2 mRNA vaccination.

14.
Cureus ; 15(1): e34452, 2023 Jan.
Article in English | MEDLINE | ID: covidwho-2258114

ABSTRACT

Myocarditis and pericarditis are inflammatory conditions affecting the myocardium and pericardium, respectively. They are caused by infectious and non-infectious conditions, including autoimmune disorders, drugs, and toxins. Vaccine-induced myocarditis has been reported with viral vaccines, including influenza and smallpox. The BNT162b2 mRNA vaccine (Pfizer-BioNTech) has shown great efficacy against symptomatic, severe coronavirus disease 2019 (COVID-19), hospital admissions, and deaths. The US FDA issued an emergency use authorization for the Pfizer-BioNTech COVID-19 mRNA vaccine for the prevention of COVID-19 in individuals ≥ five years. However, concerns were raised after reports of new cases of myocarditis following mRNA COVID-19 vaccines, especially among adolescents and young adults. Most cases developed symptoms after receiving the second dose. Here, we present a case of a previously healthy 34-year-old male who developed sudden and severe chest pain a week after the second dose of the Pfizer-BioNTech COVID-19 mRNA vaccine. Cardiac catheterization showed no angiographically obstructive coronary artery disease but it revealed intramyocardial bridging. This case report demonstrates that the mRNA COVID-19 vaccine can be associated with acute myopericarditis and the clinical presentation can mimic acute coronary syndrome. Despite that, acute myopericarditis associated with the mRNA COVID-19 vaccine is usually mild and can be managed conservatively. Incidental findings such as intramyocardial bridging should not exclude the diagnosis of myocarditis and should be carefully evaluated. COVID-19 infection has high mortality and morbidity even in young individuals, and all different COVID-19 vaccines were found effective in the prevention of severe COVID-19 infection and in decreasing COVID-19 mortality.

15.
Int J Infect Dis ; 131: 75-78, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2273810

ABSTRACT

OBJECTIVES: We aimed to estimate the rate of myocarditis after the messenger RNA (mRNA) COVID-19 booster vaccination by vaccine type, age, and sex. METHODS: We used data from the British Columbia COVID-19 Cohort, a population-based cohort surveillance platform. The exposure was a booster dose of an mRNA vaccine. The outcome was diagnosis of myocarditis during hospitalization or an emergency department visit within 7-21 days of booster vaccination. RESULTS: The overall rate of myocarditis was lower for the booster dose (6.41, 95% confidence interval [CI]: 3.50-10.75) than the second dose (17.97, 95% CI: 13.78-23.04); (Rate ratiobooster vs dose-2 = 0.34, 95% CI: 0.17-0.61). This difference was more apparent for the mRNA-1273 vaccine type. After the second dose, the myocarditis rate in males was significantly lower for BNT162b2 than mRNA-1273 overall and among those aged 18-39 years. In contrast, after the booster dose, no significant differences between myocarditis and vaccine type was observed overall or within the specific age groups among males or females. CONCLUSION: Myocarditis after mRNA COVID-19 vaccines is a rare event. A lower absolute risk of myocarditis was observed after a booster dose of mRNA vaccine than the primary series second dose.


Subject(s)
COVID-19 , Myocarditis , Female , Male , Adult , Humans , 2019-nCoV Vaccine mRNA-1273 , COVID-19 Vaccines/adverse effects , BNT162 Vaccine , Myocarditis/epidemiology , Myocarditis/etiology , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/prevention & control , Vaccination , RNA, Messenger , mRNA Vaccines
16.
Vaccines (Basel) ; 11(3)2023 Mar 16.
Article in English | MEDLINE | ID: covidwho-2270143

ABSTRACT

Elevated anti-apolipoprotein A-1 (AAA1) antibody levels associated with cardiovascular risk have been observed in previously SARS-CoV-2-infected or COVID-19-vaccinated individuals. Since patient safety is generally a priority in vaccination, we sought to investigate AAA1 antibody levels in healthy adults after mRNA vaccination. We conducted a prospective cohort study in healthy adult volunteers recruited from military workers of the Transport Air Base in Prague who had received two doses of mRNA vaccines. Anti-apolipoprotein A-1 antibody levels were determined using ELISA from serum samples obtained at three and four time points after the first and second vaccine doses, respectively, within almost 17 weeks of follow-up. The transient AAA1 positivity rate achieved 24.1% (95% confidence interval CI: 15.4-34.7%), i.e., 20 out of 83 participants had at least one positive post-vaccination sample, with a repeat positivity confirmed in only 5 of them. This rate was associated with a BMI > 26 kg/m2, as documented by an adjusted odds ratio of 6.79 (95% CI: 1.53-30.01). In addition, the highest positivity rate of 46.7% (21.3-73.4%) was observed in obese subjects with >30 kg/m2. Since the incidence rate of AAA1 positivity remained unchanged after the first and second vaccine doses, any relationship between AAA1 positivity and mRNA vaccination was inconclusive. The present study showed a transient AAA1 positivity rate associated with overweight or obesity without a proven association with mRNA vaccination.

17.
HLA ; 2023 Apr 03.
Article in English | MEDLINE | ID: covidwho-2251995

ABSTRACT

Host genetic variability contributes to susceptibility to SARS-CoV-2 infection and COVID-19 evolution and the role of HLA system has not clearly emerged, suggesting the involvement of other factors. Studying response to vaccination with Spyke protein mRNA represents an ideal model to highlight whether the humoral or cellular responses are influenced by HLA. Four hundred and sixteen workers, vaccinated with Comirnaty beginning 2021, were selected within the Azienda Ospedaliera Universitaria "Città della Salute e della Scienza di Torino." The humoral response was determined with the LIAISON® kit, while the analysis of the cellular response was performed with the Quantiferon SARS-CoV-2 assay, for the S1 (receptor-binding domain; Ag1) and S1 and S2 (Ag2) subunits of the Spyke protein. Six HLA loci were typed by next-generation sequencing. Associations between HLA and vaccine response were performed with univariate and multivariate analyses. An association was found between A*03:01, B*40:02 and DPB1*06:01 and high antibody concentration and between A*24:02, B*08:01 and C*07:01 and low humoral responses. The haplotype HLA-A*01:01 ~ B1*08:01 ~ C*07:01 ~ DRB1*03:01 ~ DQB1*02:01 conferred an increased risk of low humoral response. Considering cellular responses, 50% of the vaccinated subjects responded against Ag1 and 59% against Ag2. Carriers of DRB1*15:01 displayed a higher cellular response both to Ag1 and Ag2 compared to the rest of the cohort. Similarly, DRB1*13:02 predisposed to a robust cellular response to Ag1 and Ag2, while DRB1*11:04 showed an opposite trend. Cellular and humoral responses to Comirnaty are influenced by HLA. Humoral response is mainly associated to class I alleles, with A*03:01, previously associated to protection against severe COVID-19, and response to vaccination, standing out. Cellular response predominantly involves class II alleles, with DRB1*15:01 and DPB1*13:01 prevailing. Affinity analysis for Spyke peptides is generally in line with the association results.

18.
J Pers Med ; 13(3)2023 Feb 23.
Article in English | MEDLINE | ID: covidwho-2250474

ABSTRACT

Vaccination remains the best strategy against coronavirus disease 2019 (COVID-19) in terms of prevention. The efficacy and safety of COVID-19 vaccines is supported by well-designed clinical trials that recruited many participants. It is well-known that vaccination is associated with local side effects related to the injection site, and mild, systemic side effects. However, there has been an increase in the occurrence of what is known as infrequent adverse effects in the population of vaccinated individuals in real life. We present the case of a 46-year-old woman with no past medical history, who presented with a sharp chest pain with deep inspiration, a few days after receiving the third dose of the Pfizer-BioNTech COVID-19 mRNA vaccine (BNT162b2). There is an association between the BNT16b2 vaccination and myocarditis, pericarditis, and even bilateral pleural effusions. To the best of our knowledge, this is the first report featuring a unilateral pleural effusion in a patient with no known past medical history, who did not develop cardiac involvement nor have any viral infection. The aim of our report is to inform health professionals of the possibility of encountering this rare adverse event in their daily practice, as the population of individuals who are receiving additional vaccine doses is increasing steadily.

19.
Intern Med ; 62(10): 1547-1551, 2023 May 15.
Article in English | MEDLINE | ID: covidwho-2250021

ABSTRACT

A 78-year-old Japanese woman with no history of rheumatic disease received 2 doses of the BNT162b2 COVID-19 mRNA vaccine. Two weeks later, she noticed bilateral swelling in the submandibular region. Blood tests showed hyper-immunoglobulin (Ig)G4emia, and 18F-fluorodeoxyglucose (FDG)-positron emission tomography (PET) revealed the strong accumulation of FDG in the enlarged pancreas. She was diagnosed with IgG4-related disease (IgG4-RD) according to the American College of Rheumatology (ACR)/the European League Against Rheumatism (EULAR) classification criteria. Treatment was started with prednisolone at 30 mg/day, and the organ enlargement improved. We herein report a case of IgG4-RD that may have been associated with an mRNA vaccine.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunoglobulin G4-Related Disease , Rheumatic Diseases , Aged , Female , Humans , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Fluorodeoxyglucose F18 , Immunoglobulin G4-Related Disease/complications , Immunoglobulin G4-Related Disease/diagnosis , Immunoglobulin G4-Related Disease/drug therapy , United States , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL