Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
JHEP Reports ; 5(1), 2023.
Article in English | Scopus | ID: covidwho-2239870

ABSTRACT

Background & Aims: Liver injury with autoimmune features after vaccination against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is increasingly reported. We investigated a large international cohort of individuals with acute hepatitis arising after SARS-CoV-2 vaccination, focusing on histological and serological features. Methods: Individuals without known pre-existing liver diseases and transaminase levels ≥5x the upper limit of normal within 3 months after any anti-SARS-CoV-2 vaccine, and available liver biopsy were included. Fifty-nine patients were recruited;35 females;median age 54 years. They were exposed to various combinations of mRNA, vectorial, inactivated and protein-based vaccines. Results: Liver histology showed predominantly lobular hepatitis in 45 (76%), predominantly portal hepatitis in 10 (17%), and other patterns in four (7%) cases;seven had fibrosis Ishak stage ≥3, associated with more severe interface hepatitis. Autoimmune serology, centrally tested in 31 cases, showed anti-antinuclear antibody in 23 (74%), anti-smooth muscle antibody in 19 (61%), anti-gastric parietal cells in eight (26%), anti-liver kidney microsomal antibody in four (13%), and anti-mitochondrial antibody in four (13%) cases. Ninety-one percent were treated with steroids ± azathioprine. Serum transaminase levels improved in all cases and were normal in 24/58 (41%) after 3 months, and in 30/46 (65%) after 6 months. One patient required liver transplantation. Of 15 patients re-exposed to SARS-CoV-2 vaccines, three relapsed. Conclusion: Acute liver injury arising after SARS-CoV-2 vaccination is frequently associated with lobular hepatitis and positive autoantibodies. Whether there is a causal relationship between liver damage and SARS-CoV-2 vaccines remains to be established. A close follow-up is warranted to assess the long-term outcomes of this condition. Impact and implications: Cases of liver injury after vaccination against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) have been published. We investigated a large international cohort of individuals with acute hepatitis after SARS-CoV-2 vaccination, focusing on liver biopsy findings and autoantibodies: liver biopsy frequently shows inflammation of the lobule, which is typical of recent injury, and autoantibodies are frequently positive. Whether there is a causal relationship between liver damage and SARS-CoV-2 vaccines remains to be established. Close follow-up is warranted to assess the long-term outcome of this condition. © 2022 The Author(s)

2.
Clin Toxicol (Phila) ; : 1-7, 2022 Dec 02.
Article in English | MEDLINE | ID: covidwho-2236056

ABSTRACT

INTRODUCTION: Post-marketing data on coronavirus vaccines are limited. This study evaluated adverse reactions reported to a statewide hotline after the administration of a coronavirus disease-2019 (COVID-19) vaccine. METHODS: We collected reports between 1 December 2020 through 30 August 2021 of any individual 12 years of age and older who received an FDA EUA-approved vaccine and experienced an adverse reaction. For each case, we collected vaccine brand, demographics, adverse reaction type, severity, onset of reaction, duration, and outcome. Relative risk analyses were conducted to investigate factors associated with vaccine adverse reactions. RESULTS: 638 adverse drug reaction cases were recorded. The majority identified as female (70.8%) and the median age was 56. Implicated brands were Pfizer BNT162b2 (46.6%), Moderna mRNA-1273 (43.41%), and Janssen Ad26.COV2.S (8.78%). Although the lowest number of cases was with Janssen, this vaccine had the highest incident rate based on reactions per 100,000 doses. Adverse reactions with the highest incidence were systemic reactions (92.7%), injection-site reactions (8.5%), and local non-injection-site reactions (10.4%), with most judged as minor severity. Relative risk was higher for Moderna compared to Pfizer for injection-site non-severe (RR 2.01) and injection-site severe (RR 1.94) reactions. Janssen had a higher risk of headache, dyspnea, and vision changes compared to Pfizer, and a higher risk of headache compared to Moderna. The relative risk for fever, chills, and lymphadenopathy was higher for the second dose than the first dose for all patients. CONCLUSION: This observational study analyzing adverse drug reactions of the COVID-19 vaccine found that most complaints concerned systemic reactions. We found reaction differences among vaccine brands, between first and second doses for some effects, and selected recurrent events. Poison control centers are uniquely positioned to conduct post-marketing surveillance for the new vaccines as they are available 24/7 to the public and are healthcare providers. Further post-marketing studies are essential to provide a holistic safety profile of COVID-19 vaccines.

3.
Hematology, Transfusion and Cell Therapy ; 44(Supplement 2):S669, 2022.
Article in English | EMBASE | ID: covidwho-2179229

ABSTRACT

Objetivos: Avaliar as taxas de seroconversao apos duas doses da vacina contra SARS-CoV-2 em pacientes com Leucemia Mieloide Cronica (LMC). Material e Metodos: Foram coletadas amostras para teste sorologico de triagem para avaliacao da presenca de anticorpos IgG (CMIA, SARS-CoV-2 IgM, IgG - Alinity System, Abbott Laboratories, Ireland) no periodo de 1-3 meses apos duas doses de vacina para COVID-19. Nas amostras positivas, foi realizada analise quantitativa de IgG (anti-S1 - SARS-CoV-2 IgG II Quant, Alinity System, Abbott Laboratories, Ireland) e os Titulos de Anticorpos Neutralizantes (TAN), que detectam o efeito citopatico do virus em cultura celular induzidos pelas vacinas (Vero CCL-81 cells). Resultados: Entre agosto e novembro de 2021, foram avaliados 102 pacientes com LMC com media de idade de 56,2 anos (33-85), sendo 58,8% do sexo masculino, 98,5% em Fase Cronica (FC), 80% apresentavam ao menos Resposta Molecular Maior (RMM). 87% dos pacientes estavam em uso de ITQ e 13% estavam em descontinuacao da medicacao. 66,7% receberam a vacina ChAdOx1 nCoV-19 (AZD1222)-Covishield (Oxford/AstraZeneca/Fiocruz), 29,41% CoronaVac (Sinovac/Butatan), 1,96% BNT162b2 (Pfeizer/BioNTech/Fosun Pharma) e 1,96% Ad26.COV2.S (Janssen-Cilag). 15% dos pacientes apresentaram COVID-19 antes da vacinacao. O teste sorologico de triagem (CMIA) foi positivo em 25% dos pacientes. COVID-19 previa foi associada a presenca de anticorpos IgG (p<=0,001). Os TAN foram maiores que 1:320 em 13/26 casos, dentre os quais 5 haviam apresentado COVID-19 antes de completar o esquema vacinal. Neste grupo, 76% receberam a vacina ChAdOx1 nCoV-19, 19% Coronavac e 1% BNT162b2. Dentre os casos com infeccao previa pelo SARS-CoV-2, 7 apresentaram confirmacao laboratorial e 2 tinham quadro clinico sugestivo. Oito casos ocorreram antes da vacinacao e um paciente apresentou quadro leve, apos ter recebido a primeira dose da vacina ChAdOx1 nCoV-19. Onze pacientes estavam em tratamento com Imatinibe, 6 com Dasatinibe e um com Nilotinibe (6 com RMM), 5 em terceira ou quarta linha (sem RMM) e 3 pacientes em descontinuacao de ITQ. A proporcao de pacientes com TAN >1:320 foi superior no grupo que recebia terceira ou quarta linha (p=0,022). Entretanto, neste grupo havia mais pacientes com infeccao previa por COVID-19. Nao houve diferenca estatistica entre as taxas de seroconversao (CMIA, IgM and IgG) entre pacientes que receberam Coronavac ou ChAdOx1 nCoV-19. Nao houve diferencas nas taxas de seroconversao entre pacientes que receberam ITQ e naqueles em descontinuacao (p=0,77). Discussao: Estudos observacionais demonstraram que pacientes com LMC-FC produzem anticorpos em niveis semelhantes a populacao geral, apos receberem as vacinas ChAdOx1 nCoV-19 ou BNT162b2. No estudo conduzido por Rotterdam et al., 100/101 pacientes apresentaram seroconversao apos duas doses. Outro estudo avaliou as taxas de conversao em pacientes que receberam Coronavac, demonstrando menores taxas de conversao se comparados a vacina BNT162b2. Nosso estudo demonstrou que a resposta sorologica apos duas doses de vacina contra SARS-CoV-2 foi menor do que a descrita previamente na literatura. Conclusoes: Nao foram demonstradas diferencas entre os tipos de vacina (Coronavac vs. ChAdOx1 nCoV-19) ou em relacao a fase da doenca na taxa de seroconversao. Duas doses de vacinas para COVID-19 foram insuficientes para imunizacao adequada em pacientes com LMC. Agradecimentos: Brazilian National Council for Scientific and Technological Development (CNPq), grant ndegree 401977/2020-0. Copyright © 2022

4.
Cell Rep Med ; 4(1): 100898, 2023 01 17.
Article in English | MEDLINE | ID: covidwho-2165956

ABSTRACT

Multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposures, from infection or vaccination, can potently boost spike antibody responses. Less is known about the impact of repeated exposures on T cell responses. Here, we compare the prevalence and frequency of peripheral SARS-CoV-2-specific T cell and immunoglobulin G (IgG) responses in 190 individuals with complex SARS-CoV-2 exposure histories. As expected, an increasing number of SARS-CoV-2 spike exposures significantly enhances the magnitude of IgG responses, while repeated exposures improve the number of T cell responders but have less impact on SARS-CoV-2 spike-specific T cell frequencies in the circulation. Moreover, we find that the number and nature of exposures (rather than the order of infection and vaccination) shape the spike immune response, with spike-specific CD4 T cells displaying a greater polyfunctional potential following hybrid immunity compared with vaccination only. Characterizing adaptive immunity from an evolving viral and immunological landscape may inform vaccine strategies to elicit optimal immunity as the pandemic progress.


Subject(s)
COVID-19 , Immunoglobulin G , T-Lymphocytes , Humans , Antibody Formation , CD4-Positive T-Lymphocytes , COVID-19/epidemiology , SARS-CoV-2
5.
Pharmaceutical Journal ; 306(7949), 2022.
Article in English | EMBASE | ID: covidwho-2064952
6.
American Journal of Transplantation ; 22(Supplement 3):643, 2022.
Article in English | EMBASE | ID: covidwho-2063436

ABSTRACT

Purpose: Understanding the dynamics of antibody response to a third dose (D3) of anti-SARS-CoV-2 vaccine in solid organ transplant recipients (SOTRs) is important to inform booster strategies. Method(s): We studied the the dynamics of anti-RBD (Roche, <0.8 to >2500 U/dL) and anti-S (Euroimmun, 0.1 to >8.9 AU) antibody levels in a cohort of SOTRs at 2 weeks, 1 month and 3 months after D3. We compared the proportion of seroconversion at 1 month or 3 months after D3 between mRNA and Ad.26.COV2.S D3 recipients, using Poisson regression with robust standard error, adjusting for age and numbers of immunosuppressants. Result(s): Among 928 SOTRs with 2-week (n=655), 1-month (n=651) or 3-month (n=404) post-D3 titer, 78%, 82% and 86% tested positive for antibodies. The median (IQR) anti-RBD at the three timepoints were >2500 (73, >2500), 2494 (49, >2500) and 1234 (59, >2500) U/mL (Figure 1A, blue), and there were 61% (n=436), 60% (n=491) and 53% (n=313) with anti-RBD> 1000 u/mL, respectively. The median (IQR) anti-S at the three timepoints were 3.2 (0.3, 8.4), 8 (2, >8.9) and 7.4 (2, >8.9) AU (Figure 1B, blue), and there were 47% (n=218), 61% (n=161) and 64% (n=91) who developed anti-S>4 AU. Among patients with no or minimal immune response at 2 weeks post-D3 (n=102), 3/41 (7%) had increased anti-RBD by 1 month while 11/18 (61%) had increased anti-S (Fisher exact p<0.001). 6/29 (21%) had increased anti-RBD by 3 months while 12/20 (60%) had increased anti-S (p<0.01) (Figure 1A&B, yellow). 27/102 (27%) of them seroconverted at 1 or 3 months after D3. Having received Ad.26.COV2.S as D3 is associated with 3.9X increased proportion of seroconversion at 1 month or 3 months among patients with no or minimal immune response at 2 weeks after D3 (aIRR=2.223.926.92, p<0.001). Conclusion(s): Among SOTRs who received a booster anti-SARS-CoV-2 vaccination, dynamics of Anti-RBD and Anti-S antibodies differed substantially. Anti-RBD titers on average declined only slightly after 14 days post-D3, while anti-S increased up through 30-60 days post-D3. After the peak, average titer values for both antibodies declined slightly through three months post-D3.

7.
American Journal of Transplantation ; 22(Supplement 3):1064-1065, 2022.
Article in English | EMBASE | ID: covidwho-2063403

ABSTRACT

Purpose: Some solid-organ transplant recipients (SOTRs) with low or negative antibody levels after a 2-dose mRNA vaccine series against SARS-CoV-2 experience boosting after a third dose (D3), but long-term antibody durability after D3 is unknown. We describe six-month SARS-CoV-2 antibody kinetics and durability in 31 SOTRs who received D3. Method(s): 31 SOTRs without prior COVID-19 were identified within our national observational study. Serologic testing was performed a median of 30 (IQR 27-40) days after D3 and repeated at a median of 166 (148-184) days after D3. Semiquantitative anti-spike serologic testing using the Roche Elecsys anti-S enzyme immunoassay (EIA) or EUROIMMUN anti-S1 EIA was performed. Result(s): Over 6 months of follow-up, antibody levels increased in 16/27(59%), remained stable in 6/27(22%) (one negative, the others above the assay limit), and decreased in 5/27(19%). One-month post-D3, 24/31(77%) were seropositive and 7/31(23%) were seronegative. Six-months post-D3, 29/31(94%) were seropositive and 2/31(6%) remained seronegative. Both nonresponders received the BNT-162b2 primary series;one received Ad.26.CoV2.S and the other mRNA-1273 for D3. This difference in seroconversion after D3 was not statistically significant (Fisher exact = 0.49, between primary series). There were no reported cases of COVID-19 during the study period. Conclusion(s): We observed a very high rate of seroconversion after D3 in SOTRs, with marked heterogeneity in timing and strength of response depending on baseline antibody level and vaccine platform received. These results are encouraging evidence for the durable immunogenicity of additional vaccine doses in most SOTRs, and demonstrate the need for dedicated analysis of heterologous boosting strategies.

8.
American Journal of Transplantation ; 22(Supplement 3):595, 2022.
Article in English | EMBASE | ID: covidwho-2063375

ABSTRACT

Purpose: Even though a high efficacy and immunogenicity of COVID-19 vaccines have been reported in the general population, vaccine immunogenicity is suboptimal in SOT recipients and breakthrough SARS-CoV-2 infection has already been reported in this immunocompromised population. Thus, several approaches including booster dose administration was investigated and showed better outcome. However, as a booster, mix and match method has not been investigated enough yet. The aim and objectives of this study is to check the immunogenicity after third dose of the SARSCoV- 2 vaccines either with adenovirus vector vs. mRNA vaccine. Method(s): This is a single center, single blinded (patient blinded), randomized controlled trial comparing BNT 162b2 and JNJ-78436735 as a third dose after completion of two doses of BNTT 162b2 vaccine in SOT recipients. We included adult SOT recipients with functional graft on at least one immunosuppressive medication. Also, the participants should have completed two doses of BNT162b2 vaccination at least 28 days prior to the third dose. As a primary end point, we are going to check the anti-spike protein of SARS-CoV-2 IgG positive rate in one month after vaccination. Result(s): We have finished the enrollment and there were 60 SOT recipients including 39 kidney, 11 liver, 4 lung, 2 heart and 4 combined organ transplant recipients. Already, 58 recipients completed the follow up blood test visit. Median age of the enrolled recipients was 56.7 (IQR 51 - 63) and 22 (37.9%) were female. As a maintenance immunosuppression, 52 (89.7%), 45 (77.5%) and 26 (44.8%) recipients received tacrolimus, mycophenolate and prednisone, respectively. Conclusion(s): Immunogenicity data will be analyzed once the follow up blood test result are finished. Adverse events and the rate of COVID-19 also will be reported.

9.
American Journal of Transplantation ; 22(Supplement 3):599, 2022.
Article in English | EMBASE | ID: covidwho-2063343

ABSTRACT

Purpose: Solid organ transplant recipients are at high risk of morbidity and mortality from coronavirus disease 2019 (COVID-19) with mortality rates as high as 30% reported in the early pandemic period. COVID-19 vaccine efficacy in the immunosuppressed population is lower than the general population. Early studies suggest that monoclonal antibody (MAB) treatment against the SARS-CoV-S spike protein may decrease hospitalizations and emergency department (ED) visits. Herein, we report our single center experience with use of MAB for COVID-19 treatment in kidney transplant recipients. Method(s): We performed a retrospective chart review of all kidney transplant recipients who developed COVID-19 from March 17, 2020 to January 26, 2022 at our transplant center. Date of diagnosis, vaccine status, MAB treatment, hospitalization and patient outcome was reviewed. Result(s): Two hundred ninety-one kidney transplant recipients had positive testing for SARS-CoV-2 in the period reviewed. 120 (41%) patients received MAB treatment. One patient death, not COVID related, was excluded from analysis. Of those who received MAB treatment, 99.2% survived compared to 82.4% of those who did not (p=0.00), Figure 1. Hospitalization was lower in those who received MAB (18.3% vs 60.8%, p=0.00). Completion of vaccine series, defined as 2 doses of mRNA or 1 dose of Janssen vaccine prior to infection, was also associated with better survival (98.6% vs 80.3%, p=0.00), Figure 1. Hospitalization rate was lower in those who completed vaccination prior to infection with SARS-CoV-2 (27.1% vs 59.2%, p = 0.00). The combination of MAB therapy and completion of vaccination also decreased hospitalization compared to those who received MAB but did not complete vaccines series (14% vs 26.8%, Table 1). Subgroup analysis of 143 patients infected from December 2021 to Jan 26, 2022 which may have reflected the Omicron surge was performed (Table 2). Treatment with MAB was associated with a reduction in hospitalization (11.6%) compared to 44.6% in those who did not receive MAB. Conclusion(s): MAB treatment for COVID-19 and prior vaccination were associated with improved survival and decreased risk of hospitalization in kidney transplant recipients.

10.
American Journal of Transplantation ; 22(Supplement 3):406, 2022.
Article in English | EMBASE | ID: covidwho-2063340

ABSTRACT

Purpose: To compare antibody response to a third dose (D3) of SARS-CoV-2 vaccine in solid organ transplant recipients (SOTRs) with negative or low-positive antibody levels after 2-dose mRNA vaccination across D3 platforms. Method(s): From our observational study, 532 SOTRs who developed suboptimal antibody responses to 2-dose mRNA vaccination (Roche<50 U/mL or EUROIMMUN <1.1 AU) were selected. Belatacept recipients and persons with any COVID-19 diagnosis were excluded. We compared post-D3 antibody levels among SOTRs who received an mRNA vaccine for D3 (n=487) versus Ad.26.COV2.S for D3 (n=45). Poisson regression with robust standard error was used to study the association between vaccine platform and seroconversion, adjusting for immunosuppression, age, time since transplant, and liver transplant status. Result(s): Pre-D3, 342 SOTRs (64%) were seronegative, of whom 107 (31%) developed high-positive antibody levels post-D3. In contrast, of the 190 (36%) with low-positive pre-D3 antibody levels, 172 (91%) were high-positive post-D3 (Figure 1). Among SOTRs seronegative pre-D3, 1.8x more Ad.26.COV2.S D3 recipients seroconverted compared to mRNA D3 recipients (49.7% vs 27.8%, Fisher's exact=0.014) (Figure 2). Among the pre-D3 seronegative group, there was a 2x higher chance of developing high-positive post-D3 levels with Ad.26.COV2.S compared to mRNA D3 (IRR =1.42.02.9, p<0.001). This was despite the Ad.26. COV2.S D3 group having fewer younger patients and liver transplant recipients, factors that are associated with higher odds of positive antibody response. 165 SOTRs (31%) remained seronegative after D3 (22% of Ad.26.COV2.S recipients vs 32% of mRNA recipients). Conclusion(s): Heterologous boosting with Ad.26.COV2.S may be a promising vaccination option for SOTRs with poor response to the 2-dose mRNA series, particularly among those who are seronegative. (Table Presented).

11.
U.S ; Pharmacist. 47(9):41-46, 2022.
Article in English | EMBASE | ID: covidwho-2057848

ABSTRACT

The global health crisis of coronavirus disease 2019 (COVID-19) continues to challenge the standard of care in both the inpatient and outpatient setting. One area that has been heavily impacted is effective management of anticoagulation. COVID-19 has led to new and innovative ways to manage vitamin K-antagonist therapy in the ambulatory care clinics, with increased utilization of drive-through testing sites, home international normalized ratio monitoring, and conversions to direct oral anticoagulants. The COVID-19 infection has been found to have an increased risk in venous thromboembolism events due to its complex inflammatory response. Since 2019, multiple strategies have been trialed in the inpatient setting, including therapeutic, intermediate, and prophylaxis anticoagulation using heparin or low-molecular-weight heparin. Vaccines and therapeutics for COVID-19 have added complexity to the clinical situation, including an FDA warning for a vaccine-induced immune thrombocytopenia with the adenoviral vector vaccine (Ad26.COV2.S) and the potential for drug interactions between COVID-19 therapeutics and anticoagulation therapy. Copyright © 2022, Jobson Publishing Corporation. All rights reserved.

12.
Journal of Neuromuscular Diseases ; 9:S35-S36, 2022.
Article in English | EMBASE | ID: covidwho-2043389

ABSTRACT

Several vaccine strategies are in use against the SARS-CoV2 virus which causes COVID-19. There are three major approaches to vaccine development: Using the whole virus (live virus, inactivated virus, or viral vector vaccines), using immunogenic parts of the virus, or using the genetic material of the virus. As of April 29, 2022, 10 vaccines with different mechanisms of action have received EUL by WHO.1 In the United States, 3 vaccines have received either emergency use authorization or FDA approval.2 The timing and number of injections in the primary series and timing and need for booster doses of these vaccines depend on the age of the person, underlying immunocompromised status.4 The vaccines have overall been demonstrated to be safe and effective in preventing COVID-19 infection and also in preventing serious COVID-19 infection. However they are rarely associated with some serious side effects. Thrombosis with thrombocytopenia syndrome (TTS) after Johnson & Johnson/ Janssen adenoviral vector vaccine is a rare complication that appears to affect women between the ages of 30 and 49 years. It has also been described with the Oxford/Astra Zeneca vaccine. Based on the available data there is no increased risk for TTS after mRNA vaccination, although cases of immune thrombocytopenia have bene reported after the Pfizer/ BioNTech and Moderna vaccines.3,4 An increased risk of Guillain-Barré syndrome has also been associated with this the Janssen vaccine.3 Myocarditis and pericarditis, particularly in male adolescents and young adults have been associated with the Pfizer and Moderna mRNA vaccines.5 Currently, antibody testing either post infection or post vaccination are not recommended for various reasons.6, 7 Preexposure prophylaxis with a combination of 2 long-acting antibodies, tixagevimab-cilgavimab target the receptor binding domain of the SARS-CoV-2 spike protein and inhibit virus attachment. In December 2021, the US FDA provided emergency use authorization for preexposure prophylaxis of COVID- 19 in patients 12 years or older weighing at least 40 kg, not currently infected with COVID-19 or have a known recent exposure and either have moderate to severe immunocompromise due to a medical condition or receipt of immunosuppressive treatments and may not mount an adequate immune response to COVID-19 vaccines, or in whom available vaccines are not recommended due to prior severe adverse reaction.8 This consists of 2 consecutive intramuscular injections at the same visit and the effi cacy appears to last for up to 6 months. It was granted marketing authorization in the EU in March 2022.

13.
HemaSphere ; 6:2974-2975, 2022.
Article in English | EMBASE | ID: covidwho-2032158

ABSTRACT

Background: SARS-COV2 infection is associated with inflammation, hypercoagulability and endothelial damage. Anti-SARS-COV2 vaccines have radically changed the course of the pandemic, however, reports on rare thrombotic events raise concern in the scientific community and the general population. Aims: In a prospectively enrolled cohort of adult subjects undergoing mRNA or adenovirus vector vaccination, we wanted to longitudinally evaluate the changes in levels of hemostatic biomarkers (i.e. activation of blood coagulation and perturbance of endothelium and fibrinolysis), together with the serological response, and occurrence of manifest thrombotic complications. Methods: Peripheral venous blood samples were collected at enrollment (day 0, D0) before the 1st vaccine dose, and on 15 (D15), 60 (D60), 90 (D90) and 180 (D180) days after the 1st dose. At each time point, hemostatic markers (i.e., fibrinogen, D-dimer, FVIII, von Willebrand Factor [vWF] antigen and activity, F1+2, thrombomodulin, protein C, protein S, FXIII, tPA, and PAI-1), and anti-Spike receptor-binding-domain protein (anti-S/RBD) IgG were measured. Follow up is currently continuing. Results: Fifty-three subjects (57% males) with a median age of 50 years (range 23-86) were enrolled into the study and followed-up for 6 months: 36 (68%) received BNT162b2, 6 (11%) mRNA-1273, and 8 (15%) ChAdOx1 nCoV-19 vaccines, in 2 doses over 21, 30 and 77 days, respectively;while 3 (6%) subjects received Ad26.COV2.S as single shot. Twenty individuals (38%) reported previous history of COVID-19, with a mean time from infection to vaccination of 10 months (4-18);only 1 required Hospitalization. Nine subjects presented cardiovascular risk factors and 4 a prior, non-active, cancer;3 were on anticoagulation for atrial fibrillation. The evaluation of the hemostatic biomarkers at the different time points showed variations in some of the parameters evaluated, with median values remaining within normal range levels. Specifically, compared to baseline, we observed a significant increase in thrombomodulin at D90 (p=0.001) and D180 (p=0.03), in parallel to a significant decrease in fibrinogen (D60), vWFAg (D60 and D180), FVIII (D60, D90 and D180), and TPA (D60 and D90) levels. The reduction of these biomarkers was particularly evident in individuals with a history of COVID-19. Of interest, this group of subjects was also characterized by significantly lower levels of PAI-1 both at baseline (7.18 ng/mL vs 17.53 ng/mL;p<0.0001), and at other time points (p<0.0001), and by an increase in F1+2 at D90 (p=0.02). The association between lower baseline PAI-1 levels with history of COVID-19 was confirmed by linear regression analysis (B= -10.351, p=0.013), and was independent by the time of infection resolution. Notably, no differences were observed in the hemostatic biomarkers according to vaccine types. All subjects positively responded to vaccination with a significant increase in anti-S/RBD IgG from baseline (D0) to each time point, especially COVID-19 subjects (D15, D60, and D90:p<0.0001;D180:p=0.031). No thrombotic or cardiovascular complications occurred during follow-up. Summary/Conclusion: No hypercoagulable state elicited by COVID-19 vaccination was observed, contrarily we detected an overall persistent reduction of coagulation activation over time. Subjects with previous SARS-COV2 infection had persistently low levels of PAI-1, supporting enhanced fibrinolysis activation. Compared with recent studies, our results provide a longer observation follow-up with all vaccine types and reassure on the safety of anti- SARS-COV2 vaccination.

14.
Vaccines (Basel) ; 10(9)2022 Sep 01.
Article in English | MEDLINE | ID: covidwho-2010329

ABSTRACT

With the recent outbreak of the COVID-19 pandemic and emergency use authorization of anti-SARS-CoV-2 vaccines, reports of post-vaccine immune thrombocytopenia (ITP) have gained attention. With this systematic review, we aim to analyze the clinical characteristics, therapeutic strategies, and outcomes of patients presenting with ITP after receiving COVID-19 vaccination. Medline, Embase, and Ebsco databases were systematically explored from inception until 1 June 2022. Case reports and case series investigating the association between the anti-SARS-CoV-2 vaccine and ITP were included. We found a total of 66 patients. The mean age of presentation was 63 years with a female preponderance (60.6%). Sixteen patients had pre-existing ITP. The mean time from vaccine administration to symptom onset was 8.4 days. More ITP events were triggered by mRNA vaccines (BNT162b2 (n = 29) > mRNA-1273 (n = 13)) than with adenoviral vaccines (ChAdOx1-S AstraZeneca (n = 15) > Ad26.COV2-S (n = 9)). Most of the patients were treated with steroids or IVIG, or both. The overall outcome was promising, with no reported deaths. Our review attempts to increase awareness among physicians while evaluating patients presenting with thrombocytopenia after receiving the vaccine. In our solicited opinion, the rarity of these events and excellent outcomes for patients should not change views regarding the benefits provided by immunization.

15.
Journal of Clinical Oncology ; 40(16), 2022.
Article in English | EMBASE | ID: covidwho-2009630

ABSTRACT

Background: Patients with hematologic malignancies have a lower vaccine response and higher rates for SARS CoV-2 morbidity and mortality. We present preliminary data focusing on humoral vaccine responses and correlates with disease subtype and treatment exposure. Methods: We analyzed data from 332 patients with a hematologic malignancy from May 1, 2021 - Jan 31, 2022 who received SARSCoV-2 vaccination and performed a prospective cohort serologic study with the Elecsys®Anti-SARSCoV-2-S test. Patients received homologous or heterologous vaccine combination of BNT162b2, mRNA1273, ChAdOx1 nCoV-19, and/or Ad26.COV2.S. Blood samples were obtained before any vaccination, 2-6 weeks after the second vaccine (2V), before third vaccine (3V), and 2-6 weeks after 3V. Results: The median age was 67 years (range 18-91years) with 41.9% female. At 2V, 11.5% and at 3V, 23.8% received heterologous vaccines. Treatment status at first vaccine dose significantly affected peak 2V antibody response (p < 0.05). Seropositive rate and median antibody titer after 2V for previously untreated patients were higher compared to patients on active therapy or had previously been treated. Treatment naïve (n = 60;seropositivity 85.1%;median titer 1306 U/mL;[Q1-Q3:11.4-> 2499]);first-line (1L) active therapy (n = 127;65.4%;41.25 U/mL;[ < 0.8-592.5]);second-line and beyond (2L+) active therapy (n = 56;60.7%;2.6U/mL;[ < 0.8-154]);previous treatment with 1L (n = 66;64.8%;118 U/mL;[ < 0.8-> 2499]);previous treatment with 2L+ (n = 23;59.1%;4U/mL;[ < 0.8-229.5]). Of 61 patients that were seronegative at 2V, 17 (27.9%) seroconverted after 3V. Anti-CD20 monoclonal antibody (mAb) containing therapy as the most recent treatment from 2V had the greatest impact on humoral response. Exposure to anti-CD20 mAb based regimens or as monotherapy revealed low antibody responses (n = 84;seropositivity 22.6%;median titer < 0.8 U/mL;Q1-Q3 [ < 0.8-< 0.8]). On analysis of indolent B-cell Non-Hodgkin Lymphomas whereby antiCD-20 mAb are often incorporated, treatment proximity to 2V impacted responses: < 3 months (n = 33;22%;< 0.8 U/ mL;[ < 0.8-< 0.8]) vs. 12-24 months (n = 4;60%;228 U/mL;[ < 0.8-232]). In contrast, tyrosine kinase inhibitor (n = 38;100%;858 U/mL;[221-> 2499]), proteosome inhibitor monotherapy (n = 4;100%;median titer 1520 U/mL;[462-> 2499]) were among the subgroups with the highest numerical responses, however, the addition of corticosteroids impacted vaccine response as seen in proteosome inhibitor with corticosteroids (n = 7;85.7%;6.6 U/mL;[1.8-115.2]). Conclusions: The humoral response from our single institution cohort identifies diminished responses depending on treatment status and the type of treatment including the proximity of treatment exposure to receipt of vaccination. Furthermore, vaccine boosters can induce antibody responses in patients who were previously seronegative.

16.
Journal of Clinical Oncology ; 40(16), 2022.
Article in English | EMBASE | ID: covidwho-2009524

ABSTRACT

Background: Patients (pts) with cancer are at increased risk of severe COVID-19. Both underlying malignancy and anti-cancer treatments influence the immune system, potentially impacting the level of vaccine protection achieved. Methods: A systematic literature search of PubMed, Embase, CENTRAL and conference proceedings (ASCO annual meetings and ESMO congress) up to 28/09/21, was conducted to identify studies reporting anti-SARS-CoV-2 spike protein immunoglobulin G seroconversion rates (SR) at any time point after complete COVID-19 immunization (mRNA- or adenoviral-based vaccines) in cancer pts. Complete immunization was defined as 1 dose of JNJ-78436735 vaccine or 2 doses of BNT162b2, mRNA-1273 or ChAdOx1 nCoV-19 vaccines. Subgroup analyses were performed to examine the impact of cancer diagnosis, disease stage, and anticancer therapies on the SR. Overall effects were pooled using random-effects models and reported as pooled SR with 95% confidence intervals (CI). Results: Of 1,548 identified records, 64 studies were included in this analysis reporting data from 10,511 subjects. The Table shows the SR in the overall population and specific subgroups. In pts with solid malignancies (SM), disease stage and primary site did not significantly impact the SR. In pts with hematologic malignancies (HM), SR were significantly lower in pts with chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL) compared to acute lymphoblastic leukemia (ALL), Hodgkin lymphoma (HL), and multiple myeloma (MM). Concerning the impact of cancer therapies on SR, pts with SM undergoing chemotherapy had numerically lower SR (N = 1,234, SR 87%, CI 81-92) compared to those treated with immune checkpoint inhibitors (N = 574, SR 94%, CI 88-97) or endocrine therapy (N = 326, SR 94%, CI 86-97) with or without another targeted therapy. Pts with HM treated with anti-CD20 therapy (within the last 12 months: N = 360, SR 7%, CI 2-20;or more than 12m: N = 175, SR 59%, CI 35-80), immune-modulating agents (BTK or BCL2 inhibitors) (N = 462, SR 47%, CI 32-64%) or other immunotherapies (anti-CD19/CART or anti-CD38) (N = 293, SR 37%, CI 23-53) had lower SR compared to pts treated with autologous (N = 353, SR 77%, CI 67- 85) or allogenic stem cell transplantation (N = 509, SR 77%, CI 68-84). Conclusions: SR varies between cancer types and anticancer therapies with some cancer pts having low protection against COVID- 19 even after complete vaccination.

17.
Annals of the Rheumatic Diseases ; 81:1686-1687, 2022.
Article in English | EMBASE | ID: covidwho-2009043

ABSTRACT

Background: Patients (pts) with autoimmune diseases are at higher risk of infections, including those by SARS-COV-2. There is no general agreement regarding priority criteria for anti-COVID vaccine access for pts with autoimmune rheumatic diseases (ARD). Few studies have addressed the issue of anti-COVID vaccination in these pts, but many are available on the safety, immunogenicity, efficacy, and possible contraindications of traditional vaccines in pts with ARD. These studies may represent the basis on which to recommend the anti-COVID-19 vaccines. Objectives: Patients (pts) with autoimmune diseases are at higher risk of infections, including those by SARS-COV-2. There is no general agreement regarding priority criteria for anti-COVID vaccine access for pts with autoimmune rheumatic diseases (ARD). Few studies have addressed the issue of anti-COVID vaccination in these pts, but many are available on the safety, immunogenicity, efficacy, and possible contraindications of traditional vaccines in pts with ARD. These studies may represent the basis on which to recommend the anti-COVID-19 vaccines. Methods: A telephone survey investigating the AE of SARS-CoV-2 vaccinations on pts with systemic lupus erythematosus, systemic sclerosis, infammatory arthritis (rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis), idiopathic infammatory myopathies, ANCA-associated vasculitis was administered. Data extraction included diagnosis, disease activity status, demographics, disease duration, therapy, comor-bidities, and laboratory tests. Vaccinated participants are asked to report whether they experienced any local or systemic AE following vaccination, and if so, to report on the severity and duration of the AE. Mild AE were defned as unpleasant reactions that did not limit daily activities, moderate AE as those that limited daily activities, and severe AE-required medical attention. Serious AE were defned as reactions that resulted in hospital admission. Results: ChAdOx1 nCoV-19 and BNT162b2 are the most common vaccines in our pts. 98 (39,84 %) of 246 pts received the BNT162b2 vaccine, 95 (38,62 %)-ChA-dOx1 nCoV-19 vaccine, 47 (19,10%)-CX-024414 and 6 (2,44 %) were vaccinated with Ad26.COV2-S. 127 (51,63%) pts had at least one mild AE and 51 (20,73 %) pts reported moderate AE. Severe AE were rare-4 (1,63 %) pts and no serious AE were reported. The most commonly reported AE is pain (40,24 %), redness (30,49 %), swelling (18,7 %) at the injection site, which was consistent across all vaccines for both frst and second doses. Systemic AE occurred in 104 (43,27 %) pts, most frequently fatigue (29,67 %), headache (27,13 %) and muscle ache (24,39 %). The symptoms started mostly during the frst day post-vaccination and lasted for no more than two days. Joint complaints were reported in 8,94 %, but only a small proportion of pts (2,84 %) reported a deterioration of their autoimmune disease up to 3 months after COVID-19 vaccination. Age was a signifcant effect modifer in the association between autoimmune status and the risk of moderate or severe AE. Vaccination with ChAdOx1 nCoV-19, female sex, age between 35-50 years were independently associated with an increased likelihood of reporting any AE. The current results support the safety of different COVID-19 vaccines in pts with ARD. This information can help fght vaccine hesitancy in this population. Conclusion: Our data indicated that COVID-19 vaccines are well tolerated by pts with ARD. We did not observe any serious AE, but the number of pts included in our study is too low to draw conclusions about rare serious events. Additionally, our data suggest that COVID-19 vaccinations do not seem to trigger autoimmune disease fares, which is in accordance with data from previous small studies that assessed consequences of vaccines in pts with ARD.

18.
Annals of the Rheumatic Diseases ; 81:1665-1666, 2022.
Article in English | EMBASE | ID: covidwho-2008843

ABSTRACT

Background: Currently there is little information on the efficacy and safety of SARS-CoV-2 vaccination in patients with immune-mediated diseases and/or under immunosuppressive treatment in our country, where different types of vaccines and mix regimens are used. For this reason, the Argentine Society of Rheumatology (SAR) with the Argentine Society of Psoriasis (SOARPSO) set out to develop a national register of patients with rheumatic and immune-mediated infammatory diseases (IMIDs) who have received a SARS-CoV-2 vaccine in order to assess their efficacy and safety in this population. Objectives: To assess SARS-CoV-2 vaccine efficacy and safety in patients with rheumatic and IMIDs. Methods: SAR-CoVAC is a national, multicenter and observational registry. Adult patients with a diagnosis of rheumatic or IMIDs who have been vaccinated for SARS-CoV-2 were consecutively included between June 1st and September 17th, 2021. Sociodemographic data, comorbidities, underlying rheumatic or IMIDs, treatments received and their modifcation prior to vaccination and history of SARS-CoV-2 infection were recorded. In addition, the date and place of vaccination, type of vaccine applied, scheme and indication will be registered. Finally, adverse events (AE), as well as SARS-CoV-2 infection after the application of the vaccine were documented Results: A total of 1234 patients were included, 79% were female, with a mean age of 57.8 (SD 14.1) years. The most frequent diseases were rheumatoid arthritis (41.2%), osteoarthritis (14.5%), psoriasis (12.7%) and spondy-loarthritis (12.3%). Most of them were in remission (28.5%) and low disease activity (41.4%). At the time of vaccination, 21% were receiving glucocorti-coid treatment, 35.7% methotrexate, 29.7% biological (b) Disease Modifying Anti-Rheumatic Drugs (DMARDs) and 5.4% JAK inhibitors. Before vaccine application 16.9% had had a SARS-CoV-2 infection. Regarding the frst dose of the vaccine, the most of the patients (51.1%) received Gam-COV-ID-Vac, followed by ChAdOx1 nCoV-19 (32.8%) and BBIBP-CorV (14.5%). In a lesser proportion, BNT162b2 (0.6%), Ad26.COV2.S (0.2%) and Coro-naVac (0.2%) vaccines were used. Almost half of them (48.8%) completed the scheme, 12.5% were mix regimenes, the most frequent being Gam-COVID-Vac/mRNA-1273. The median time between doses was 51days (IQR 53). More than a quarter (25.9%) of the patients reported at least one AE after the frst dose and 15.9% after the second. The fu-like syndrome and local hypersensitivity were the most frequent manifestations. There was one case of mild anaphylaxis. No patient was hospitalized. Altogether, the incidence of AE was 246.5 events/1000 doses. BBIBP-CorV presented signifcantly lower incidence of AE in comparison with the other types of vaccines. (118.5 events/1000 doses, p<0.002 in all cases) Regarding efficacy, 63 events of SARS-CoV-2 infection were reported after vaccination, 19% occurred before 14 days post-vaccination, 57.1% after the frst dose (>14 days) and 23.8% after the second. In most cases (85.9%) the infection was asymptomatic or had an outpatient course and 2 died due to COVID-19. Conclusion: In this national cohort of patients with rheumatic and IMIDs vaccinated for SARS-CoV-2, the most widely used vaccines were Gam-COVID-Vac and ChAdOx1 nCoV-19, approximately half completed the schedule and in most cases homologously. A quarter of the patients presented some AE, while 5.1% presented SARS-CoV-2 infection after vaccination, in most cases mild.

19.
Journal of Clinical Oncology ; 40(16), 2022.
Article in English | EMBASE | ID: covidwho-2005705

ABSTRACT

Background: Patients (pts) with Hodgkin lymphoma (HL) may be at an increased risk from COVID-19- related complications due to immunosuppression and common therapies with pulmonary toxicities (e.g. bleomycin, radiation). Additionally, they may experience unexpected adverse events (AEs) from the novel SARS-CoV-2 vaccines due to the intrinsic abnormalities in cellular immunity. Exploring their perspective on the COVID-19 pandemic and subjective AEs from the novel vaccines may ultimately allow providers to develop improved services based on needs of this specific population. Methods: We conducted a single-institution, questionnaire-based study in pts treated after 1/1/2015, for diagnosis of HL. Pts were identified through the electronic medical records at our institution. After signing an informed consent, pts answered 19 questions designed to reflect on their COVID-19 pandemic experience. An additional section aimed to determine the percentage of pts with a local or systemic AEs within 7 days of novel SARS-CoV-2 vaccine dose. Results: We enrolled 30 pts with median age 28.5 years (18-66);16 (53%) were male, and 26 (87%) were on active HL therapy. With data cutoff 1/28/ 2022, 27 pts (90%) completed the questionnaire. A total of 8 (27%) patients had COVID-19 infection with 75% of those occurring after the vaccination. None required hospitalization and none reported residual pulmonary issues. Eighteen (67%) had a member of their household test positive for COVID-19. Most pts (67%) did not require cancellation of their appointments due to COVID-19-related issues during the pandemic. 85% of pts participated in at least 1 telehealth medical appointment and the majority (72%) were satisfied with their telehealth experience for cancer care. A total of 24 (80%) pts received the novel SARS-CoV-2 vaccines (58% had Pfizer-BioNTech BNT162b2, 38% Moderna mRNA-1273, 4% Ad26.COV2.S) and 23 pts completed the AE section of the questionnaire. The vaccines were generally well-tolerated. Six pts (26%) reported at least one grade 3 AE, but none had grade 4 local or systemic AEs. The most common AE was pain at injection site (65% after dose 1, 55% after dose 2, 69% after dose 3). AEs that occurred in over 25% of pts during any of the injections included redness/swelling/pain at the injection site, fatigue, headache, muscle pain, and fever. Of 23 eligible pts, 13 (57%) received 3rd dose of the vaccine. No unexpected toxicities or autoimmune issues were noted by participants in this study following vaccination with a median follow up of 10 months after the 1st vaccine dose. Conclusions: Based on responses from our study, HL pts adjusted to the challenges of the COVID-19 pandemic with minimal disruptions to their cancer care, showed positive attitudes toward telehealth, and appear without any unexpected toxicities from the novel vaccines.

20.
Journal of Clinical Oncology ; 40(16), 2022.
Article in English | EMBASE | ID: covidwho-2005688

ABSTRACT

Background: Coranavirus disease 2019 (COVID-19) has affected more than 220 millon people worldwide. The establishment of safe and effective vaccine coverage is crucial in these patients. We aim to investigate the safety, adverse effects and immune response (cellular and humoral) in cancer patients under active treatment and risk criteria. Methods: 56 samples from cancer patients under active treatment for cancer were collected before and after vaccination Demographic, clinical and biochemical data were collected and a post-vaccination symptomatology questionnaire was performed. For the determination of anti-SARS-Cov-2 antibodies (Immunoglobulin G and immunoglobulin M) we used the ELISA technique (LIASON SARS-CoV-2 S1/S2 IgG test and LIASON SARS-CoV-2 IgM test, DiaSorin, Saluggia, Italy). A study of the lymphocyte population was performed by flow cytometry. Results: We enrolled 52 patients with cancer who received mRNA vaccines (mRNA-1273 and BNT162b2), 2 patients with AZD1222vaccine and 2 with Ad26.COV2.S vaccine. All patients were SARS-COV-2 naive as determined by a negative anti-SARS-COVID-2 IgG test baseline. The median follow-up time was 50 days after receip of a second vaccine dosis. All subject received anti- cancer therapy. The most common anti-cancer treatment received by this cohort of patients was cytotoxic chemoterapy (44.6%), immunotherapy (25%), and monoclonal antibody therapy (14.3%). Overall, a high rate of seroconvertion(anti-IgG) (94,5%) was observed in our cohort, 1 patients with chemotherapy (1,8%) and 2 patients with immunotherapy (3,6%) were negatives. No significant differences in antibody titer were observed according to therapy. Thirty percent presented an antibody titer lower than 1000 U/ml, 27 (48%) patients developed an anti-SARS-CoV2 titer between 1000-4000 U/ml, and only 9 (16%) subjects presented a titer higher than 4000 U/ml. Studies of the lymphocyte profile of vaccinated patients showed no significant changes in the subtypes, except for peripheral memory CD3+ CD8+ lymphocytes which were significantly increased (p = 0.0001) after the second dose of anti-SARS-CoV-2 vaccine. Interestingly, cell apoptosis was significantly reduced in almost all T lymphocyte subtypes studied in vaccinated patients. Finally, analysis of blood cells showed a statistically significant increase (p = 0.0402) of eosinophils in vaccinated patients compared to baseline data. Conclusions: A personalized approach to vaccination can be proposed to cancer patients, especially depending on the type of tumor and the specific oncological treatments received. We are currently recruiting patients (n = 150) who have received the third dose of vaccine and plan to follow up at 6 months for humoral and cellular immune response. Final results will be reported at the ASCO meeting.

SELECTION OF CITATIONS
SEARCH DETAIL