Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Intervalo de año de publicación
1.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-22281024

RESUMEN

Age is a major risk factor for hospitalization and death after SARS-CoV-2 infection, even in vaccinees. Suboptimal responses to a primary vaccination course have been reported in the elderly, but there is little information regarding the impact of age on responses to booster third doses. Here we show that individuals 70 or older who received a primary two dose schedule with AZD1222 and booster third dose with mRNA vaccine achieved significantly lower neutralizing antibody responses against SARS-CoV-2 spike pseudotyped virus compared to those younger than 70. One month after the booster neither the concentration of serum binding anti spike IgG antibody, nor the frequency of spike-specific B cells showed differences by age grouping. However, the impaired neutralization potency and breadth post-third dose in the elderly was associated with enrichment of circulating "atypical" spike-specific B cells expressing CD11c and FCRL5. Single cell RNA sequencing confirmed an expansion of TBX21-, ITGAX-expressing B cells in the elderly that enriched for B cell activation/receptor signalling pathway genes. Importantly we also observed impaired T cell responses to SARS-CoV-2 spike peptides in the elderly post-booster, both in terms of IFNgamma and IL2 secretion, as well as a decrease in T cell receptor signalling pathway genes. This expansion of atypical B cells and impaired T cell responses may contribute to the generation of less affinity-matured antibodies, with lower neutralizing capacity post-third dose in the elderly. Altogether, our data reveal the extent and potential mechanistic underpinning of the impaired vaccine responses present in the elderly after a booster dose, contributing to their increased susceptibility to COVID-19 infection.

2.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-22280033

RESUMEN

The effect of immune checkpoint blockade on COVID-19 immunity is unclear. In this study, we determine whether immune checkpoint blockade expanded age-associated B cells (ABCs) are similar to those present in other conditions, and whether they enhance or detract from the COVID-19 vaccine responses. First, we use single cell RNA sequencing (scRNAseq) to show that ABCs arising from distinct aetiologies have common transcriptional profiles and may be further subdivided according to expression of genes associated with different immune functions, including the autoimmune regulator (AIRE). Next, we perform detailed longitudinal profiling of the COVID-19 vaccination response in patients. Finally, we show that high pre-vaccination ABC frequency correlates with decreased levels of antigen-specific memory B cells, and reduced magnitude and longevity of neutralising capacity against authentic SARS-CoV-2 virus. Expansion of ABCs is a biomarker for individuals with cancer requiring additional or more frequent booster immunisation against COVID-19.

3.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-22276196

RESUMEN

Obesity is associated with an increased risk of severe Covid-19. However, the effectiveness of SARS-CoV-2 vaccines in people with obesity is unknown. Here we studied the relationship between body mass index (BMI), hospitalization and mortality due to Covid-19 amongst 3.5 million people in Scotland. Vaccinated people with severe obesity (BMI>40 kg/m2) were significantly more likely to experience hospitalization or death from Covid-19. Excess risk increased with time since vaccination. To investigate the underlying mechanisms, we conducted a prospective longitudinal study of the immune response in a clinical cohort of vaccinated people with severe obesity. Compared with normal weight people, six months after their second vaccine dose, significantly more people with severe obesity had unquantifiable titres of neutralizing antibody against authentic SARS-CoV-2 virus, reduced frequencies of antigen-experienced SARS-CoV-2 Spike-binding B cells, and a dissociation between anti-Spike antibody levels and neutralizing capacity. Neutralizing capacity was restored by a third dose of vaccine, but again declined more rapidly in people with severe obesity. We demonstrate that waning of SARS-CoV-2 vaccine-induced humoral immunity is accelerated in people with severe obesity and associated with increased hospitalization and mortality from breakthrough infections. Given the prevalence of obesity, our findings have significant implications for global public health.

4.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-22274668

RESUMEN

BackgroundThere are no real world data on vaccine elicited neutralising antibody responses for the worlds most widely used vaccine, AZD1222, in African populations following scale up. Here, we measured i) baseline SARS-CoV-2 seroprevalence and levels of protective neutralizing antibodies prior to vaccination rollout using both flow cytometric based analysis of binding antibodies to nucleocapsid (N), coupled with virus neutralisation approaches and ii) neutralizing antibody responses to VOC prior to vaccination (January 2021) and after two-doses of AZD1222 vaccine administered with a 12 week interval in Lagos, Nigeria - a period when the Delta variant was circulating. MethodsHealth workers at multiple sites in Lagos were recruited to the study. For binding antibody measurement, IgG antibodies against SARS-COV-2 Wuhan-1 receptor-binding domain (RBD), trimeric spike protein (S), nucleocapsid protein (N) and Omicron S1 were measured using the Luminex-based SARS-CoV-2-IgG assay by flow cytometry. For plasma neutralising antibody measurement, SARS-CoV-2 lentiviral pseudovirus (PV) were prepared by transfecting 293T cells with Wuhan-614G wild type (WT), B.1.617.2 (Delta) and BA.1 (Omicron) plasmids in conjunction with HIV-1 expression vectors and luciferase encoding genome flanked by LTRs. We performed serial plasma dilutions from each time point and mixed plasma with PV before infecting HeLa-ACE2 cell lines, reading out luminescence and calculating ID50 (reciprocal dilution of sera required to inhibit 50% of PV infection). ResultsOur underlying study population receiving at least one dose of vaccine comprised 140 participants with a median age of 40 (interquartile range: 33, 48). 62/140 (44%) participants were anti-N IgG positive prior to administration of first vaccine dose. 49 had plasma samples available at baseline prior to vaccination and at two follow-up timepoints post two dose vaccination for neutralization assays. Half of the participants, 25/49 (51%) were IgG anti-N positive at baseline. Of the 24 individuals anti-N Ab negative at baseline, 12/24 had ID50 above the cut-off of 20. In these individuals, binding antibodies to S were also detectable, and neutralisation correlated with IgG anti-S, suggesting waning of N antibody after infection. Overall, neutralizing Ab titres to WT 1 month after second dose were 2579 and at 3 months post second-dose were 1695. As expected, lower levels of neutralization were observed against the Delta GMT 549 and Omicron variants 269 at 1 month. Positive anti-N IgG Ab status at baseline was associated with significantly higher titres of neutralizing antibodies following vaccination across all tested VOC. Those with anti-N Abs present at baseline did not experience waning of responses between months 1 and 3 post second dose. When data were analysed for negative anti-N IgG status at any timepoint, there was a significant decline in neutralization and binding antibodies between 1 month and 3 months post second-dose. The GMT in these individuals for Delta and Omicron was approximately 100, nearly a log lower in comparison to WT. We tested anti-N IgG in subjects who were anti-N IgG negative at baseline (n=78) and became positive between 1- and 3-months post second dose and found 7/49 (14%) with de-novo infection, with one additional participant demonstrating both reinfection and breakthrough infection to yield a total breakthrough rate of 8/49 (16%). Neutralising and binding Ab titres 1 month post vaccine, prior to breakthrough, were not associated with breakthrough infection. Neutralizing titres were higher at the last time point in individuals who had experienced vaccine breakthrough infection (with no evidence of infection prior to vaccine), indicating a boosting effect of infection in addition to vaccine. However, neutralisation and binding S antibodies against Omicron were low in those with either prior exposure or infection following two dose AZD1222. ConclusionsAZD1222 is immunogenic in this real world west African cohort with significant background seroprevalence and incidence of breakthrough infection over a short time period. Prior infection and breakthrough infection induced higher anti-SARS-CoV-2 Ab responses at 3 months post vaccine against all widely circulating VOC. However, responses to Omicron BA.1 were low at three months regardless of hybrid immunity from prior exposure or breakthrough infection. Booster doses after AZD1222 should be considered in the African setting, even after natural infection, as future variants may be more pathogenic as well as immune evasive in the context of waning immunity.

5.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-473248

RESUMEN

The SARS-CoV-2 Omicron BA.1 variant emerged in late 2021 and is characterised by multiple spike mutations across all spike domains. Here we show that Omicron BA.1 has higher affinity for ACE2 compared to Delta, and confers very significant evasion of therapeutic monoclonal and vaccine-elicited polyclonal neutralising antibodies after two doses. mRNA vaccination as a third vaccine dose rescues and broadens neutralisation. Importantly, antiviral drugs remdesevir and molnupiravir retain efficacy against Omicron BA.1. We found that in human nasal epithelial 3D cultures replication was similar for both Omicron and Delta. However, in lower airway organoids, Calu-3 lung cells and gut adenocarcinoma cell lines live Omicron virus demonstrated significantly lower replication in comparison to Delta. We noted that despite presence of mutations predicted to favour spike S1/S2 cleavage, the spike protein is less efficiently cleaved in live Omicron virions compared to Delta virions. We mapped the replication differences between the variants to entry efficiency using spike pseudotyped virus (PV) entry assays. The defect for Omicron PV in specific cell types correlated with higher cellular RNA expression of TMPRSS2, and accordingly knock down of TMPRSS2 impacted Delta entry to a greater extent as compared to Omicron. Furthermore, drug inhibitors targeting specific entry pathways demonstrated that the Omicron spike inefficiently utilises the cellular protease TMPRSS2 that mediates cell entry via plasma membrane fusion. Instead, we demonstrate that Omicron spike has greater dependency on cell entry via the endocytic pathway requiring the activity of endosomal cathepsins to cleave spike. Consistent with suboptimal S1/S2 cleavage and inability to utilise TMPRSS2, syncytium formation by the Omicron spike was dramatically impaired compared to the Delta spike. Overall, Omicron appears to have gained significant evasion from neutralising antibodies whilst maintaining sensitivity to antiviral drugs targeting the polymerase. Omicron has shifted cellular tropism away from TMPRSS2 expressing cells that are enriched in cells found in the lower respiratory and GI tracts, with implications for altered pathogenesis.

6.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21266112

RESUMEN

COVID-19 has been associated with many neurological complications including stroke, delirium and encephalitis. Furthermore, many individuals experience a protracted post-viral syndrome which is dominated by neuropsychiatric symptoms, and is seemingly unrelated to COVID-19 severity. The true frequency and underlying mechanisms of neurological injury are unknown, but exaggerated host inflammatory responses appear to be a key driver of severe COVID-19 more broadly. We sought to investigate the dynamics of, and relationship between, serum markers of brain injury (neurofilament light [NfL], Glial Fibrillary Acidic Protein [GFAP] and total Tau) and markers of dysregulated host response including measures of autoinflammation (proinflammatory cytokines) and autoimmunity. Brain injury biomarkers were measured using the Quanterix Simoa HDx platform, cytokine profiling by Luminex (R&D) and autoantibodies by a custom protein microarray. During hospitalisation, patients with COVID-19 demonstrated elevations of NfL and GFAP in a severity-dependant manner, and there was evidence of ongoing active brain injury at follow-up 4 months later. Raised NfL and GFAP were associated with both elevations of pro-inflammatory cytokines and the presence of autoantibodies; autoantibodies were commonly seen against lung surfactant proteins as well as brain proteins such as myelin associated glycoprotein, but reactivity was seen to a large number of different antigens. Furthermore, a distinct process characterised by elevation of serum total Tau was seen in patients at follow-up, which appeared to be independent of initial disease severity and was not associated with dysregulated immune responses in the same manner as NfL and GFAP.

7.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21257572

RESUMEN

Precision monitoring of antibody responses during the COVID-19 pandemic is increasingly important during large scale vaccine rollout and rise in prevalence of Severe Acute Respiratory Syndrome-related Coronavirus-2 (SARS-CoV-2) variants of concern (VOC). Equally important is defining Correlates of Protection (CoP) for SARS-CoV-2 infection and COVID-19 disease. Data from epidemiological studies and vaccine trials identified virus neutralising antibodies (Nab) and SARS-CoV-2 antigen-specific (notably RBD, and S) binding antibodies as candidate CoP. In this study, we used the World Health Organisation (WHO) international standard to benchmark neutralising antibody responses and a large panel of binding antibody assays to compare convalescent sera obtained from: a) COVID-19 patients; b) SARS-CoV-2 seropositive healthcare workers (HCW) and c) seronegative HCW. The ultimate aim of this study, was to identify biomarkers of humoral immunity that could be used as candidate CoP in internationally accepted unitage. Whenever suitable, the antibody levels of the samples studied were expressed in International Units (INU) for virus neutralisation assays or International Binding Antibody Units (BAU) for ELISA tests. In this work we used commercial and non-commercial antibody binding assays; a lateral flow test for detection of SARS-CoV-2-specific IgG / IgM; a high throughput multiplexed particle flow cytometry assay for SARS-CoV-2 Spike (S), Nucleocapsid (N) and Receptor Binding Domain (RBD) proteins); a multiplex antigen semi-automated immuno-blotting assay measuring IgM, IgA and IgG; a pseudotyped microneutralisation test (pMN) and electroporation-dependent neutralisation assay (EDNA). Our results indicate that overall, severe COVID-19 patients showed statistically significantly higher levels of SARS-CoV-2-specific neutralising antibodies (average 1029 IU/ml) than those observed in seropositive HCW with mild or asymptomatic infections (379 IU/ml) and that clinical severity scoring, based on WHO guidelines was tightly correlated with neutralisation and RBD / S binding assays. In addition, there was a positive correlation between severity, N-antibody assays and intracellular virus neutralisation.

8.
Preprint en Inglés | bioRxiv | ID: ppbiorxiv-435957

RESUMEN

Efforts to define serological correlates of protection against COVID-19 have been hampered by the lack of a simple, scalable, standardised assay for SARS-CoV-2 infection and antibody neutralisation. Plaque assays remain the gold standard, but are impractical for high-throughput screening. In this study, we show that expression of viral proteases may be used to quantitate infected cells. Our assays exploit the cleavage of specific oligopeptide linkers, leading to the activation of cell-based optical biosensors. First, we characterise these biosensors using recombinant SARS-CoV-2 proteases. Next, we confirm their ability to detect viral protease expression during replication of authentic virus. Finally, we generate reporter cells stably expressing an optimised luciferase-based biosensor, enabling viral infection to be measured within 24 h in a 96- or 384-well plate format, including variants of concern. We have therefore developed a luminescent SARS-CoV-2 reporter cell line, and demonstrated its utility for the relative quantitation of infectious virus and titration of neutralising antibodies. Author summaryTechniques for measuring infection with SARS-CoV-2 in the laboratory are laborious and time-consuming, and different laboratories use different approaches. There is therefore no generally agreed way to quantitate neutralising antibodies against SARS-CoV-2, which block infection with the virus and protect people from COVID-19. In this study, we describe a new way to measure SARS-CoV-2 infection, which is much simpler and faster than existing methods. It relies on the production of a specific protease enzyme by the virus, which is able to cleave and activate an engineered protein biosensor in infected cells. This biosensor emits light in the presence of viral infection, and the amount of light released is used as a readout for the amount of infectious SARS-CoV-2 present. The signal is very sensitive, so the number of infected cells required is very small, and the method can be scaled-up to test many samples at once. In particular, we demonstrate how it can be used to detect different variants of SARS-CoV-2, and quantitate neutralising antibodies against these viruses.

9.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21251054

RESUMEN

Two dose mRNA vaccination provides excellent protection against SARS-CoV-2. However, there are few data on vaccine efficacy in elderly individuals above the age of 801. Additionally, new variants of concern (VOC) with reduced sensitivity to neutralising antibodies have raised fears for vulnerable groups. Here we assessed humoral and cellular immune responses following vaccination with mRNA vaccine BNT162b22 in elderly participants prospectively recruited from the community and younger health care workers. Median age was 72 years and 51% were females amongst 140 participants. Neutralising antibody responses after the first vaccine dose diminished with increasing age, with a marked drop in participants over 80 years old. Sera from participants below and above 80 showed significantly lower neutralisation potency against B.1.1.7, B.1.351 and P.1. variants of concern as compared to wild type. Those over 80 were more likely to lack any neutralisation against VOC compared to younger participants following first dose. The adjusted odds ratio for inadequate neutralisation activity against the B.1.1.7, P.1 and B.1.351 variant in the older versus younger age group was 4.3 (95% CI 2.0-9.3, p<0.001), 6.7 (95% CI 1.7-26.3, p=0.008) and 1.7 (95% CI 0.5-5.7, p=0.41). Binding IgG and IgA antibodies were lower in the elderly, as was the frequency of SARS-CoV-2 Spike specific B-memory cells. We observed a trend towards lower somatic hypermutation in participants with suboptimal neutralisation, and elderly participants demonstrated clear reduction in class switched somatic hypermutation, driven by the IgA1/2 isotype. SARS-CoV-2 Spike specific T-cell IFN{gamma} and IL-2 responses fell with increasing age, and both cytokines were secreted primarily by CD4 T cells. We conclude that the elderly are a high risk population that warrant specific measures in order to mitigate against vaccine failure, particularly where variants of concern are circulating.

10.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-21249840

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) transmission is uncontrolled in many parts of the world, compounded in some areas by higher transmission potential of the B1.1.7 variant now seen in 50 countries. It is unclear whether responses to SARS-CoV-2 vaccines based on the prototypic strain will be impacted by mutations found in B.1.1.7. Here we assessed immune responses following vaccination with mRNA-based vaccine BNT162b2. We measured neutralising antibody responses following a single immunization using pseudoviruses expressing the wild-type Spike protein or the 8 amino acid mutations found in the B.1.1.7 spike protein. The vaccine sera exhibited a broad range of neutralising titres against the wild-type pseudoviruses that were modestly reduced against B.1.1.7 variant. This reduction was also evident in sera from some convalescent patients. Decreased B.1.1.7 neutralisation was also observed with monoclonal antibodies targeting the N-terminal domain (9 out of 10), the Receptor Binding Motif (RBM) (5 out of 31), but not in neutralising mAbs binding outside the RBM. Introduction of the E484K mutation in a B.1.1.7 background to reflect newly emerging viruses in the UK led to a more substantial loss of neutralising activity by vaccine-elicited antibodies and mAbs (19 out of 31) over that conferred by the B.1.1.7 mutations alone. E484K emergence on a B.1.1.7 background represents a threat to the vaccine BNT162b.

11.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20248765

RESUMEN

In a study of 207 SARS-CoV2-infected individuals with a range of severities followed over 12 weeks from symptom onset, we demonstrate that an early robust bystander CD8 T cell immune response, without systemic inflammation, is characteristic of asymptomatic or mild disease. Those presenting to hospital had delayed bystander responses and systemic inflammation already evident at around symptom onset. Such early evidence of inflammation suggests immunopathology may be inevitable in some individuals, or that preventative intervention might be needed before symptom onset. Viral load does not correlate with the development of this pathological response, but does with its subsequent severity. Immune recovery is complex, with profound persistent cellular abnormalities correlating with a change in the nature of the inflammatory response, where signatures characteristic of increased oxidative phosphorylation and reactive-oxygen species-associated inflammation replace those driven by TNF and IL-6. These late immunometabolic inflammatory changes and unresolved immune defects may have clinical implications.

12.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20241927

RESUMEN

SARS-CoV-2 Spike protein is critical for virus infection via engagement of ACE2, and amino acid variation in Spike is increasingly appreciated. Given both vaccines and therapeutics are designed around Wuhan-1 Spike, this raises the theoretical possibility of virus escape, particularly in immunocompromised individuals where prolonged viral replication occurs. Here we report chronic SARS-CoV-2 with reduced sensitivity to neutralising antibodies in an immune suppressed individual treated with convalescent plasma, generating whole genome ultradeep sequences by both short and long read technologies over 23 time points spanning 101 days. Although little change was observed in the overall viral population structure following two courses of remdesivir over the first 57 days, N501Y in Spike was transiently detected at day 55 and V157L in RdRp emerged. However, following convalescent plasma we observed large, dynamic virus population shifts, with the emergence of a dominant viral strain bearing D796H in S2 and{Delta} H69/{Delta}V70 in the S1 N-terminal domain NTD of the Spike protein. As passively transferred serum antibodies diminished, viruses with the escape genotype diminished in frequency, before returning during a final, unsuccessful course of convalescent plasma. In vitro, the Spike escape double mutant bearing{Delta} H69/{Delta}V70 and D796H conferred decreased sensitivity to convalescent plasma, whilst maintaining infectivity similar to wild type. D796H appeared to be the main contributor to decreased susceptibility, but incurred an infectivity defect. The{Delta} H69/{Delta}V70 single mutant had two-fold higher infectivity compared to wild type and appeared to compensate for the reduced infectivity of D796H. Consistent with the observed mutations being outside the RBD, monoclonal antibodies targeting the RBD were not impacted by either or both mutations, but a non RBD binding monoclonal antibody was less potent against{Delta} H69/{Delta}V70 and the double mutant. These data reveal strong selection on SARS-CoV-2 during convalescent plasma therapy associated with emergence of viral variants with reduced susceptibility to neutralising antibodies.

13.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20145318

RESUMEN

With the first 2020 surge of the COVID-19 pandemic, many health care workers (HCW) were re-deployed to critical care environments to support intensive care teams to look after high numbers of patients with severe COVID-19. There was considerable anxiety of increased risk of COVID19 for staff working in these environments. Using a multiplex platform to assess serum IgG responses to SARS-CoV-2 N, S and RBD proteins, and detailed symptom reporting, we screened over 500 HCW (25% of the total workforce) in a quaternary level hospital to explore the relationship between workplace and evidence of exposure to SARS-CoV-2. Whilst 45% of the cohort reported symptoms that they consider may have represented COVID-19, overall seroprevalence was 14% with anosmia and fever being the most discriminating symptoms for seropositive status. There was a significant difference in seropositive status between staff working in clinical and non-clinical roles (9% patient facing critical care, 15% patient facing non-critical care, 22% nonpatient facing). In the seropositive cohort, symptom severity increased with age for men and not for women. In contrast, there was no relationship between symptom severity and age or sex in the seronegative cohort reporting possible COVID-19 symptoms. Of the 12 staff screened PCR positive (10 symptomatic), 3 showed no evidence of seroconversion in convalescence. ConclusionThe current approach to Personal Protective Equipment (PPE) appears highly effective in protecting staff from patient acquired infection in the critical care environment including protecting staff managing interhospital transfers of COVID-19 patients. The relationship between seroconversion and disease severity in different demographics warrants further investigation. Longitudinally paired virological and serological surveillance, with symptom reporting are urgently required to better understand the role of antibody in the outcome of HCW exposure during subsequent waves of COVID-19 in health care environments.

14.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20220699

RESUMEN

BackgroundThe COVID-19 pandemic continues to grow at an unprecedented rate. Healthcare workers (HCWs) are at higher risk of SARS-CoV-2 infection than the general population but risk factors for HCW infection are not well described. MethodsWe conducted a prospective sero-epidemiological study of HCWs at a UK teaching hospital using a SARS-CoV-2 immunoassay. Risk factors for seropositivity were analysed using multivariate logistic regression. Findings410/5,698 (7{middle dot}2%) staff tested positive for SARS-CoV-2 antibodies. Seroprevalence was higher in those working in designated COVID-19 areas compared with other areas (9{middle dot}47% versus 6{middle dot}16%) Healthcare assistants (aOR 2{middle dot}06 [95%CI 1{middle dot}14-3{middle dot}71]; p=0{middle dot}016) and domestic and portering staff (aOR 3{middle dot}45 [95% CI 1{middle dot}07-11{middle dot}42]; p=0{middle dot}039) had significantly higher seroprevalence than other staff groups after adjusting for age, sex, ethnicity and COVID-19 working location. Staff working in acute medicine and medical sub-specialities were also at higher risk (aOR 2{middle dot}07 [95% CI 1{middle dot}31-3{middle dot}25]; p<0{middle dot}002). Staff from Black, Asian and minority ethnic (BAME) backgrounds had an aOR of 1{middle dot}65 (95% CI 1{middle dot}32 - 2{middle dot}07; p<0{middle dot}001) compared to white staff; this increased risk was independent of COVID-19 area working. The only symptoms significantly associated with seropositivity in a multivariable model were loss of sense of taste or smell, fever and myalgia; 31% of staff testing positive reported no prior symptoms. InterpretationRisk of SARS-CoV-2 infection amongst HCWs is heterogeneous and influenced by COVID-19 working location, role, age and ethnicity. Increased risk amongst BAME staff cannot be accounted for solely by occupational factors. FundingWellcome Trust, Addenbrookes Charitable Trust, National Institute for Health Research, Academy of Medical Sciences, the Health Foundation and the NIHR Cambridge Biomedical Research Centre. Research in context Evidence before this studySpecific risk factors for SARS-CoV-2 infection in healthcare workers (HCWs) are not well defined. Additionally, it is not clear how population level risk factors influence occupational risk in defined demographic groups. Only by identifying these factors can we mitigate and reduce the risk of occupational SARS-CoV-2 infection. We performed a review of the evidence for HCW-specific risk factors for SARS-CoV-2 infection. We searched PubMed with the terms "SARS-CoV-2" OR "COVID-19" AND "Healthcare worker" OR "Healthcare Personnel" AND "Risk factor" to identify any studies published in any language between December 2019 and September 2020. The search identified 266 studies and included a meta-analysis and two observational studies assessing HCW cohort seroprevalence data. Seroprevalence and risk factors for HCW infections varied between studies, with contradictory findings. In the two serological studies, one identified a significant increased risk of seroprevalence in those working with COVID-19 patients (Eyre et al 2020), as well as associations with job role and department. The other study (Dimcheff et al 2020) found no significant association between seropositivity and any identified demographic or occupational factor. A meta-analysis of HCW (Gomez-Ochoa et al 2020) assessed >230,000 participants as a pooled analysis, including diagnoses by both RT-PCR and seropositivity for SARS-CoV-2 antibodies and found great heterogeneity in study design and reported contradictory findings. Of note, they report a seropositivity rate of 7% across all studies reporting SARS-CoV-2 antibodies in HCWs. Nurses were the most frequently affected healthcare personnel and staff working in non-emergency inpatient settings were the most frequently affected group. Our search found no prospective studies systematically evaluating HCW specific risk factors based entirely on seroprevalence data. Added value of this studyOur prospective cohort study of almost 6,000 HCWs at a large UK teaching hospital strengthens previous findings from UK-based cohorts in identifying an increased risk of SARS-CoV-2 exposure amongst HCWs. Specifically, factors associated with SARS-CoV-2 exposure include caring for confirmed COVID-19 cases and identifying as being within specific ethnic groups (BAME staff). We further delineated the risk amongst BAME staff and demonstrate that occupational factors alone do not account for all of the increased risk amongst this group. We demonstrate for the first time that healthcare assistants represent a key at-risk occupational group, and challenge previous findings of significantly higher risk amongst nursing staff. Seroprevalence in staff not working in areas with confirmed COVID-19 patients was only marginally higher than that of the general population within the same geographical region. This observation could suggest the increased risk amongst HCWs arises through occupational exposure to confirmed cases and could account for the overall higher seroprevalence in HCWs, rather than purely the presence of staff in healthcare facilities. Over 30% of seropositive staff had not reported symptoms consistent with COVID-19, and in those who did report symptoms, differentiating COVID-19 from other causes based on symptom data alone was unreliable. Implications of all the available evidenceInternational efforts to reduce the risk of SARS-CoV-2 infection amongst HCWs need to be prioritised. The risk of SARS-CoV-2 infection amongst HCWs is heterogenous but also follows demonstrable patterns. Potential mechanisms to reduce the risk for staff working in areas with confirmed COVID-19 patients include improved training in hand hygiene and personal protective equipment (PPE), better access to high quality PPE, and frequent asymptomatic testing. Wider asymptomatic testing in healthcare facilities has the potential to reduce spread of SARS-CoV-2 within hospitals, thereby reducing patient and staff risk and limiting spread between hospitals and into the wider community. The increased risk of COVID-19 amongst BAME staff cannot be explained by purely occupational factors; however, the increased risk amongst minority ethnic groups identified here was stark and necessitates further evaluation.

15.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20197590

RESUMEN

BackgroundInternational guidelines for testing potentially immunosuppressed cancer patients receiving non-surgical anticancer therapies for SARS-CoV-2 (COVID-19) are currently lacking. The value of routinely testing staff treating cancer patients is not known. MethodsPatient-facing oncology department staff at work during the COVID-19 pandemic consented to have a nasopharyngeal swab SARS-CoV-2 antigen test by polymerase chain reaction (PCR) and blood tests for SARS-CoV-2 antibody using a laboratory Luminex-based assay and a rapid point-of-care (POC) assay on 2 occasions 28 days apart in June and July 2020. Findings434 participants were recruited: nurses (58{middle dot}3%), doctors (21{middle dot}2%), radiographers (10{middle dot}4%) and administrators (10{middle dot}1%). 82% were female; median age 40-years (range 19-66). 26{middle dot}3% reported prior symptoms suggestive of SARS-CoV-2 infection and 1{middle dot}4% tested PCR-positive prior to June 2020. All were PCR-negative at both study day 1 and 28. 18{middle dot}4% were SARS-CoV-2 sero-positive on day 1 by Luminex, of whom 42{middle dot}5% also tested positive by POC. 47{middle dot}5% of Luminex sero-positives had antibodies to both nucleocapsid (N) and surface (S) antigens. Nurses (21{middle dot}3%) and doctors (17{middle dot}4%) had higher prevalence trends of Luminex sero-positivity compared with administrators (13{middle dot}6%) and radiographers (8{middle dot}9%) (p=0.2). 38% of sero-positive participants reported previous symptoms suggestive of SARS-CoV-2 infection, a 1{middle dot}9-fold higher odds than sero-negative participants (p=0{middle dot}01). 400 participants re-tested on day 28: 13{middle dot}3% were Luminex sero-positive of whom 92{middle dot}5% were previously positive and 7{middle dot}5% newly positive. Nurses (16{middle dot}5%) had the highest seroprevalence trend amongst staff groups (p=0{middle dot}07). 32{middle dot}5% of day 1 sero-positives became sero-negative by day 28: the majority being previously reactive to the N-antigen only (p<0{middle dot}0001). InterpretationThe high prevalence of SARS-CoV-2 IgG sero-positivity in oncology nurses, and the high decline of positivity over 4 weeks supports regular antigen and antibody testing in this staff group for SARS-CoV-2 as part of routine patient care prior to availability of a vaccine. FundingACT, NHS Evidence before this studyTo identify studies involving oncology healthcare workers and SARS-CoV-2 exposure during the COVID-19 pandemic, we searched PubMed and Medrxiv for articles published between January 1 and July 31 using the following search terms "COVID-19", "SARS-CoV-2", "oncology staff", "healthcare workers" without language restriction. To date, no large study has specifically reported and tracked patient-facing oncology staff SARS-CoV-2 exposure. Added value of this studyTo the best of our knowledge, this is the first study specifically investigating SARS-CoV-2 exposure in UK patient-facing oncology staff who were at work during the peak of the COVID-19 pandemic between March and June 2020. 18{middle dot}4% of staff were SARS-CoV-2 antibody positive at the start of June 2020 suggesting prior SARS-CoV-2 infection, while 32{middle dot}5% of those antibody-positive cases became antibody-negative 28 days after the first sample collection. The highest seroprevalence rates at both time points were recorded in nurses. Implications of all the available evidenceThese results justify incorporating SARS-CoV-2 PCR and antibody testing of oncology nurses into international guidelines for managing cancer patients treated with non-surgical anticancer treatments prior to availability of a functional vaccine.

16.
Preprint en Inglés | medRxiv | ID: ppmedrxiv-20136838

RESUMEN

BackgroundThe global SARS-CoV-2 (COVID-19) pandemic has caused substantial worldwide mortality. At present, there is no data regarding oncologist-specific SARS-CoV-2 infection/immunity rates in the United Kingdom (UK) which might impact planning for the management of potentially immunosuppressed cancer patients. Here, we present the first results from the COVID-19 Serology in Oncology Staff (CSOS) study with the aim of informing non-surgical oncology management guidelines. MethodsPatient-facing staff working in an oncology department during the COVID-19 pandemic at a large district general hospital in the East of England were invited to participate. Samples were collected during the first week of June 2020: blood for SARS-COV-2 antibody testing using a rapid lateral flow point of care (POC) assay and a laboratory Luminex based assay, as well as a nasopharyngeal swab for SARS-CoV-2 PCR testing. Participant characteristics were also collected. ResultsSeventy participants were recruited: nurses (45/70; 64.3%), doctors (15/70; 21.2%), and other patient-facing staff (10/70; 14.3%). The majority were female (61/70; 87.1%) with a mean age of 42 years (median 41; range 23-64 years). A minority were smokers (9/70; 10%) or had chronic underlying health conditions (16/70; 22.9%), the commonest being asthma. All participants were nasopharyngeal-swab PCR negative, although 4/70 (5.7%) had previously tested positive by NHS testing undertaken during the preceding months. 15/70 (21.4%) had positive SARS-CoV-2 antibodies using the Luminex test. Nurses had the highest incidence of positive antibodies (13/45; 28.9%), with a lower incidence in doctors (2/15; 13.3%) although this difference was not statistically significant (Fischers exact test p=0.3). No receptionists had positive antibody tests. All four participants with a previously reported positive PCR test were antibody-positive. 9/15 (60%) of antibody-positive participants reported previous symptoms suggestive of SARS-CoV-2 infection: a 3.6-fold higher odds than antibody-negative participants, of whom 16/55 reported symptoms (p=0.03). The mean duration of symptoms was 11 days (median 11; range 1-35 days) and the mean time from resolution of reported previous symptoms to antibody testing was 48.4 days (median 46; range 1-123 days). ConclusionThis study establishes the SARS-CoV-2 exposure and carriage rate amongst patient-facing staff working in the oncology department of a large UK general hospital during the pandemic. These results may help inform UK national oncology patient management prior to the development of a viable vaccine or treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...