Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtre
1.
Neurol Neuroimmunol Neuroinflamm ; 10(4)2023 07.
Article Dans Anglais | MEDLINE | ID: covidwho-2317258

Résumé

BACKGROUND AND OBJECTIVES: SARS-CoV-2 infection has been associated with a syndrome of long-term neurologic sequelae that is poorly characterized. We aimed to describe and characterize in-depth features of neurologic postacute sequelae of SARS-CoV-2 infection (neuro-PASC). METHODS: Between October 2020 and April 2021, 12 participants were seen at the NIH Clinical Center under an observational study to characterize ongoing neurologic abnormalities after SARS-CoV-2 infection. Autonomic function and CSF immunophenotypic analysis were compared with healthy volunteers (HVs) without prior SARS-CoV-2 infection tested using the same methodology. RESULTS: Participants were mostly female (83%), with a mean age of 45 ± 11 years. The median time of evaluation was 9 months after COVID-19 (range 3-12 months), and most (11/12, 92%) had a history of only a mild infection. The most common neuro-PASC symptoms were cognitive difficulties and fatigue, and there was evidence for mild cognitive impairment in half of the patients (MoCA score <26). The majority (83%) had a very disabling disease, with Karnofsky Performance Status ≤80. Smell testing demonstrated different degrees of microsmia in 8 participants (66%). Brain MRI scans were normal, except 1 patient with bilateral olfactory bulb hypoplasia that was likely congenital. CSF analysis showed evidence of unique intrathecal oligoclonal bands in 3 cases (25%). Immunophenotyping of CSF compared with HVs showed that patients with neuro-PASC had lower frequencies of effector memory phenotype both for CD4+ T cells (p < 0.0001) and for CD8+ T cells (p = 0.002), an increased frequency of antibody-secreting B cells (p = 0.009), and increased frequency of cells expressing immune checkpoint molecules. On autonomic testing, there was evidence for decreased baroreflex-cardiovagal gain (p = 0.009) and an increased peripheral resistance during tilt-table testing (p < 0.0001) compared with HVs, without excessive plasma catecholamine responses. DISCUSSION: CSF immune dysregulation and neurocirculatory abnormalities after SARS-CoV-2 infection in the setting of disabling neuro-PASC call for further evaluation to confirm these changes and explore immunomodulatory treatments in the context of clinical trials.


Sujets)
Lymphocytes T CD8+ , COVID-19 , Femelle , Mâle , Humains , COVID-19/complications , SARS-CoV-2 , Encéphale , Catécholamines
2.
Lancet Infect Dis ; 23(5): 578-588, 2023 05.
Article Dans Anglais | MEDLINE | ID: covidwho-2299258

Résumé

BACKGROUND: Human monoclonal antibodies might offer an important new approach to reduce malaria morbidity and mortality. In the first two parts of a three-part clinical trial, the antimalarial monoclonal antibody CIS43LS conferred high protection against parasitaemia at doses of 20 mg/kg or 40 mg/kg administered intravenously followed by controlled human malaria infection. The ability of CIS43LS to confer protection at lower doses or by the subcutaneous route is unknown. We aimed to provide data on the safety and optimisation of dose and route for the human antimalaria monoclonal antibody CIS43LS. METHODS: VRC 612 Part C was the third part of a three-part, first-in-human, phase 1, adaptive trial, conducted at the University of Maryland, Baltimore Center for Vaccine Development and Global Health, Baltimore, MD, USA. We enrolled adults aged 18-50 years with no previous malaria vaccinations or infections, in a sequential, dose-escalating manner. Eligible participants received the monoclonal antibody CIS43LS in a single, open-label dose of 1 mg/kg, 5 mg/kg, or 10 mg/kg intravenously, or 5 mg/kg or 10 mg/kg subcutaneously. Participants underwent controlled human malaria infection by the bites of five mosquitoes infected with Plasmodium falciparum 3D7 strain approximately 8 weeks after their monoclonal antibody inoculation. Six additional control participants who did not receive CIS43LS underwent controlled human malaria infection simultaneously. Participants were followed-up daily on days 7-18 and day 21, with qualitative PCR used for P falciparum detection. Participants who tested positive for P falciparum were treated with atovaquone-proguanil and those who remained negative were treated at day 21. Participants were followed-up until 24 weeks after dosing. The primary outcome was safety and tolerability of CIS43LS at each dose level, assessed in the as-treated population. Secondary outcomes included protective efficacy of CIS43LS after controlled human malaria infection. This trial is now complete and is registered with ClinicalTrials.gov, NCT04206332. FINDINGS: Between Sept 1, 2021, and Oct 29, 2021, 47 people were assessed for eligibility and 31 were enrolled (one subsequently withdrew and was replaced) and assigned to receive doses of 1 mg/kg (n=7), 5 mg/kg (n=4), and 10 mg/kg (n=3) intravenously and 5 mg/kg (n=4) and 10 mg/kg (n=4) subcutaneously, or to the control group (n=8). CIS43LS administration was safe and well tolerated; no serious adverse events occurred. CIS43LS protected 18 (82%) of 22 participants who received a dose. No participants developed parasitaemia following dosing at 5 mg/kg intravenously or subcutaneously, or at 10 mg/kg intravenously or subcutaneously. All six control participants and four of seven participants dosed at 1 mg/kg intravenously developed parasitaemia after controlled human malaria infection. INTERPRETATION: CIS43LS was safe and well tolerated, and conferred protection against P falciparum at low doses and by the subcutaneous route, providing evidence that this approach might be useful to prevent malaria across several clinical use cases. FUNDING: National Institute of Allergy and Infectious Diseases, National Institutes of Health.


Sujets)
Antipaludiques , Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Adulte , Animaux , Humains , Anticorps monoclonaux/usage thérapeutique , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/prévention et contrôle , Plasmodium falciparum , Vaccins contre le paludisme/usage thérapeutique
3.
Sci Transl Med ; 15(692): eade4790, 2023 04 19.
Article Dans Anglais | MEDLINE | ID: covidwho-2305673

Résumé

Influenza vaccines could be improved by platforms inducing cross-reactive immunity. Immunodominance of the influenza hemagglutinin (HA) head in currently licensed vaccines impedes induction of cross-reactive neutralizing stem-directed antibodies. A vaccine without the variable HA head domain has the potential to focus the immune response on the conserved HA stem. This first-in-human dose-escalation open-label phase 1 clinical trial (NCT03814720) tested an HA stabilized stem ferritin nanoparticle vaccine (H1ssF) based on the H1 HA stem of A/New Caledonia/20/1999. Fifty-two healthy adults aged 18 to 70 years old enrolled to receive either 20 µg of H1ssF once (n = 5) or 60 µg of H1ssF twice (n = 47) with a prime-boost interval of 16 weeks. Thirty-five (74%) 60-µg dose participants received the boost, whereas 11 (23%) boost vaccinations were missed because of public health restrictions in the early stages of the COVID-19 pandemic. The primary objective of this trial was to evaluate the safety and tolerability of H1ssF, and the secondary objective was to evaluate antibody responses after vaccination. H1ssF was safe and well tolerated, with mild solicited local and systemic reactogenicity. The most common symptoms included pain or tenderness at the injection site (n = 10, 19%), headache (n = 10, 19%), and malaise (n = 6, 12%). We found that H1ssF elicited cross-reactive neutralizing antibodies against the conserved HA stem of group 1 influenza viruses, despite previous H1 subtype head-specific immunity. These responses were durable, with neutralizing antibodies observed more than 1 year after vaccination. Our results support this platform as a step forward in the development of a universal influenza vaccine.


Sujets)
COVID-19 , Vaccins antigrippaux , Grippe humaine , Adolescent , Adulte , Sujet âgé , Humains , Adulte d'âge moyen , Jeune adulte , Anticorps neutralisants , Anticorps antiviraux , Anticorps neutralisants à large spectre , Glycoprotéine hémagglutinine du virus influenza , Hémagglutinines , Pandémies
4.
Br J Haematol ; 199(5): 679-687, 2022 12.
Article Dans Anglais | MEDLINE | ID: covidwho-2277554

Résumé

Patients with severe aplastic anaemia (SAA) are often not vaccinated against viruses due to concerns of ineffective protective antibody response and potential for pathogenic global immune system activation, leading to relapse. We evaluated the impact of COVID-19 vaccination on haematological indices and disease status and characterized the humoural and cellular responses to vaccination in 50 SAA patients, who were previously treated with immunosuppressive therapy (IST). There was no significant difference in haemoglobin (p = 0.52), platelet count (p = 0.67), absolute lymphocyte (p = 0.42) and neutrophil (p = 0.98) counts prior to and after completion of vaccination series. Relapse after vaccination, defined as a progressive decline in counts requiring treatment, occurred in three patients (6%). Humoural response was detectable in 90% (28/31) of cases by reduction in an in-vitro Angiotensin II Converting Enzyme (ACE2) binding and neutralization assay, even in patients receiving ciclosporin (10/11, 90.1%). Comparison of spike-specific T-cell responses in 27 SAA patients and 10 control subjects revealed qualitatively similar CD4+ Th1-dominant responses to vaccination. There was no difference in CD4+ (p = 0.77) or CD8+ (p = 0.74) T-cell responses between patients on or off ciclosporin therapy at the time of vaccination. Our data highlight appropriate humoural and cellular responses in SAA previously treated with IST and true relapse after vaccination is rare.


Sujets)
Anémie aplasique , COVID-19 , Humains , Anémie aplasique/traitement médicamenteux , Ciclosporine/usage thérapeutique , Vaccins contre la COVID-19/usage thérapeutique , SARS-CoV-2 , Immunosuppresseurs/usage thérapeutique , COVID-19/prévention et contrôle , Récidive , Immunité , Vaccination
5.
Open Forum Infect Dis ; 10(3): ofad069, 2023 Mar.
Article Dans Anglais | MEDLINE | ID: covidwho-2259370

Résumé

Background: Hybrid immunity is associated with more durable protection against coronavirus disease 2019 (COVID-19). We describe the antibody responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in vaccinated and unvaccinated individuals. Methods: The 55 vaccine arm COVID-19 cases diagnosed during the blinded phase of the Coronavirus Efficacy trial were matched with 55 placebo arm COVID-19 cases. Pseudovirus neutralizing antibody (nAb) activity to the ancestral strain and binding antibody (bAb) responses to nucleocapsid and spike antigens (ancestral and variants of concern [VOCs]) were assessed on disease day 1 (DD1) and 28 days later (DD29). Results: The primary analysis set was 46 vaccine cases and 49 placebo cases with COVID-19 at least 57 days post-first dose. For vaccine group cases, there was a 1.88-fold rise in ancestral antispike bAbs 1 month post-disease onset, although 47% had no increase. The vaccine-to-placebo geometric mean ratios for DD29 antispike and antinucleocapsid bAbs were 6.9 and 0.04, respectively. DD29 mean bAb levels were higher for vaccine vs placebo cases for all VOCs. DD1 nasal viral load positively correlated with bAb levels in the vaccine group. Conclusions: Following COVID-19, vaccinated participants had higher levels and greater breadth of antispike bAbs and higher nAb titers than unvaccinated participants. These were largely attributable to the primary immunization series.

6.
Nat Microbiol ; 7(12): 1996-2010, 2022 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-2185886

Résumé

Measuring immune correlates of disease acquisition and protection in the context of a clinical trial is a prerequisite for improved vaccine design. We analysed binding and neutralizing antibody measurements 4 weeks post vaccination as correlates of risk of moderate to severe-critical COVID-19 through 83 d post vaccination in the phase 3, double-blind placebo-controlled phase of ENSEMBLE, an international randomized efficacy trial of a single dose of Ad26.COV2.S. We also evaluated correlates of protection in the trial cohort. Of the three antibody immune markers we measured, we found most support for 50% inhibitory dilution (ID50) neutralizing antibody titre as a correlate of risk and of protection. The outcome hazard ratio was 0.49 (95% confidence interval 0.29, 0.81; P = 0.006) per 10-fold increase in ID50; vaccine efficacy was 60% (43%, 72%) at non-quantifiable ID50 (<2.7 IU50 ml-1) and increased to 89% (78%, 96%) at ID50 = 96.3 IU50 ml-1. Comparison of the vaccine efficacy by ID50 titre curves for ENSEMBLE-US, the COVE trial of the mRNA-1273 vaccine and the COV002-UK trial of the AZD1222 vaccine supported the ID50 titre as a correlate of protection across trials and vaccine types.


Sujets)
Ad26COVS1 , COVID-19 , Humains , COVID-19/prévention et contrôle , Vaccin ChAdOx1 nCoV-19 , Vaccin ARNm-1273 contre la COVID-19 , , Anticorps neutralisants
7.
Nat Commun ; 13(1): 7733, 2022 12 14.
Article Dans Anglais | MEDLINE | ID: covidwho-2160214

Résumé

An important consequence of infection with a SARS-CoV-2 variant is protective humoral immunity against other variants. However, the basis for such cross-protection at the molecular level is incompletely understood. Here, we characterized the repertoire and epitope specificity of antibodies elicited by infection with the Beta, Gamma and WA1 ancestral variants and assessed their cross-reactivity to these and the more recent Delta and Omicron variants. We developed a method to obtain immunoglobulin sequences with concurrent rapid production and functional assessment of monoclonal antibodies from hundreds of single B cells sorted by flow cytometry. Infection with any variant elicited similar cross-binding antibody responses exhibiting a conserved hierarchy of epitope immunodominance. Furthermore, convergent V gene usage and similar public B cell clones were elicited regardless of infecting variant. These convergent responses despite antigenic variation may account for the continued efficacy of vaccines based on a single ancestral variant.


Sujets)
COVID-19 , Région variable d'immunoglobuline , Humains , Épitopes/génétique , SARS-CoV-2/génétique , Clones cellulaires , Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , Glycoprotéine de spicule des coronavirus/génétique
8.
Cell ; 185(23): 4333-4346.e14, 2022 Nov 10.
Article Dans Anglais | MEDLINE | ID: covidwho-2041612

Résumé

SARS-CoV-2 mRNA booster vaccines provide protection from severe disease, eliciting strong immunity that is further boosted by previous infection. However, it is unclear whether these immune responses are affected by the interval between infection and vaccination. Over a 2-month period, we evaluated antibody and B cell responses to a third-dose mRNA vaccine in 66 individuals with different infection histories. Uninfected and post-boost but not previously infected individuals mounted robust ancestral and variant spike-binding and neutralizing antibodies and memory B cells. Spike-specific B cell responses from recent infection (<180 days) were elevated at pre-boost but comparatively less so at 60 days post-boost compared with uninfected individuals, and these differences were linked to baseline frequencies of CD27lo B cells. Day 60 to baseline ratio of BCR signaling measured by phosphorylation of Syk was inversely correlated to days between infection and vaccination. Thus, B cell responses to booster vaccines are impeded by recent infection.


Sujets)
Lymphocytes B , COVID-19 , Vaccins antiviraux , Humains , Anticorps neutralisants , Anticorps antiviraux , COVID-19/prévention et contrôle , Vaccins contre la COVID-19 , SARS-CoV-2 , Vaccination , Lymphocytes B/immunologie
9.
Proc Natl Acad Sci U S A ; 119(28): e2204607119, 2022 07 12.
Article Dans Anglais | MEDLINE | ID: covidwho-1908385

Résumé

Messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are highly effective at inducing protective immunity. However, weak antibody responses are seen in some individuals, and cellular correlates of immunity remain poorly defined, especially for B cells. Here we used unbiased approaches to longitudinally dissect primary antibody, plasmablast, and memory B cell (MBC) responses to the two-dose mRNA-1273 vaccine in SARS-CoV-2-naive adults. Coordinated immunoglobulin A (IgA) and IgG antibody responses were preceded by bursts of spike-specific plasmablasts after both doses but earlier and more intensely after dose 2. While antibody and B cell cellular responses were generally robust, they also varied within the cohort and decreased over time after a dose-2 peak. Both antigen-nonspecific postvaccination plasmablast frequency after dose 1 and their spike-specific counterparts early after dose 2 correlated with subsequent antibody levels. This correlation between early plasmablasts and antibodies remained for titers measured at 6 months after vaccination. Several distinct antigen-specific MBC populations emerged postvaccination with varying kinetics, including two MBC populations that correlated with 2- and 6-month antibody titers. Both were IgG-expressing MBCs: one less mature, appearing as a correlate after the first dose, while the other MBC correlate showed a more mature and resting phenotype, emerging as a correlate later after dose 2. This latter MBC was also a major contributor to the sustained spike-specific MBC response observed at month 6. Thus, these plasmablasts and MBCs that emerged after both the first and second doses with distinct kinetics are potential determinants of the magnitude and durability of antibodies in response to mRNA-based vaccination.


Sujets)
Vaccin ARNm-1273 contre la COVID-19 , Production d'anticorps , Lymphocytes B , COVID-19 , ARN messager , SARS-CoV-2 , Vaccin ARNm-1273 contre la COVID-19/administration et posologie , Vaccin ARNm-1273 contre la COVID-19/immunologie , Lymphocytes B/immunologie , COVID-19/prévention et contrôle , Humains , Immunité cellulaire , Immunoglobuline A/sang , Immunoglobuline A/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , ARN messager/administration et posologie , ARN messager/immunologie , SARS-CoV-2/immunologie , Vaccination
10.
Science ; 373(6556)2021 Aug 13.
Article Dans Anglais | MEDLINE | ID: covidwho-1559379

Résumé

The emergence of highly transmissible SARS-CoV-2 variants of concern (VOCs) that are resistant to therapeutic antibodies highlights the need for continuing discovery of broadly reactive antibodies. We identified four receptor binding domain-targeting antibodies from three early-outbreak convalescent donors with potent neutralizing activity against 23 variants, including the B.1.1.7, B.1.351, P.1, B.1.429, B.1.526, and B.1.617 VOCs. Two antibodies are ultrapotent, with subnanomolar neutralization titers [half-maximal inhibitory concentration (IC50) 0.3 to 11.1 nanograms per milliliter; IC80 1.5 to 34.5 nanograms per milliliter). We define the structural and functional determinants of binding for all four VOC-targeting antibodies and show that combinations of two antibodies decrease the in vitro generation of escape mutants, suggesting their potential in mitigating resistance development.


Sujets)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/immunologie , Angiotensin-converting enzyme 2/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/métabolisme , Anticorps neutralisants/composition chimique , Anticorps neutralisants/métabolisme , Anticorps antiviraux/composition chimique , Anticorps antiviraux/métabolisme , Affinité des anticorps , Réaction antigène-anticorps , COVID-19/virologie , Humains , Échappement immunitaire , Fragments Fab d'immunoglobuline/immunologie , Fragments Fab d'immunoglobuline/métabolisme , Mutation , Tests de neutralisation , Domaines protéiques , Récepteurs du coronavirus/antagonistes et inhibiteurs , Récepteurs du coronavirus/métabolisme , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme
11.
Science ; 374(6573): 1343-1353, 2021 Dec 10.
Article Dans Anglais | MEDLINE | ID: covidwho-1483979

Résumé

Neutralizing antibody responses gradually wane against several variants of concern (VOCs) after vaccination with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine messenger RNA-1273 (mRNA-1273). We evaluated the immune responses in nonhuman primates that received a primary vaccination series of mRNA-1273 and were boosted about 6 months later with either homologous mRNA-1273 or heterologous mRNA-1273.ß, which encompasses the spike sequence of the B.1.351 Beta variant. After boost, animals had increased neutralizing antibody responses across all VOCs, which was sustained for at least 8 weeks after boost. Nine weeks after boost, animals were challenged with the SARS-CoV-2 Beta variant. Viral replication was low to undetectable in bronchoalveolar lavage and significantly reduced in nasal swabs in all boosted animals, suggesting that booster vaccinations may be required to sustain immunity and protection.


Sujets)
Vaccin ARNm-1273 contre la COVID-19/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Immunogénicité des vaccins , SARS-CoV-2/immunologie , , Vaccin ARNm-1273 contre la COVID-19/administration et posologie , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/analyse , Anticorps antiviraux/sang , Liquide de lavage bronchoalvéolaire/immunologie , Liquide de lavage bronchoalvéolaire/virologie , COVID-19/immunologie , COVID-19/virologie , Vaccins contre la COVID-19/administration et posologie , Immunité muqueuse , Rappel de vaccin , Macaca mulatta , Cellules B mémoire/immunologie , Nez/immunologie , Nez/virologie , ARN viral/analyse , SARS-CoV-2/génétique , SARS-CoV-2/isolement et purification , SARS-CoV-2/physiologie , Lymphocytes T auxiliaires folliculaires/immunologie , Lymphocytes auxiliaires Th1/immunologie , Réplication virale
12.
N Engl J Med ; 385(9): 803-814, 2021 08 26.
Article Dans Anglais | MEDLINE | ID: covidwho-1373469

Résumé

BACKGROUND: Additional interventions are needed to reduce the morbidity and mortality caused by malaria. METHODS: We conducted a two-part, phase 1 clinical trial to assess the safety and pharmacokinetics of CIS43LS, an antimalarial monoclonal antibody with an extended half-life, and its efficacy against infection with Plasmodium falciparum. Part A of the trial assessed the safety, initial side-effect profile, and pharmacokinetics of CIS43LS in healthy adults who had never had malaria. Participants received CIS43LS subcutaneously or intravenously at one of three escalating dose levels. A subgroup of participants from Part A continued to Part B, and some received a second CIS43LS infusion. Additional participants were enrolled in Part B and received CIS43LS intravenously. To assess the protective efficacy of CIS43LS, some participants underwent controlled human malaria infection in which they were exposed to mosquitoes carrying P. falciparum sporozoites 4 to 36 weeks after administration of CIS43LS. RESULTS: A total of 25 participants received CIS43LS at a dose of 5 mg per kilogram of body weight, 20 mg per kilogram, or 40 mg per kilogram, and 4 of the 25 participants received a second dose (20 mg per kilogram regardless of initial dose). No safety concerns were identified. We observed dose-dependent increases in CIS43LS serum concentrations, with a half-life of 56 days. None of the 9 participants who received CIS43LS, as compared with 5 of 6 control participants who did not receive CIS43LS, had parasitemia according to polymerase-chain-reaction testing through 21 days after controlled human malaria infection. Two participants who received 40 mg per kilogram of CIS43LS and underwent controlled human malaria infection approximately 36 weeks later had no parasitemia, with serum concentrations of CIS43LS of 46 and 57 µg per milliliter at the time of controlled human malaria infection. CONCLUSIONS: Among adults who had never had malaria infection or vaccination, administration of the long-acting monoclonal antibody CIS43LS prevented malaria after controlled infection. (Funded by the National Institute of Allergy and Infectious Diseases; VRC 612 ClinicalTrials.gov number, NCT04206332.).


Sujets)
Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Antipaludiques/usage thérapeutique , Paludisme à Plasmodium falciparum/prévention et contrôle , Adulte , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux/pharmacocinétique , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/pharmacocinétique , Anticorps antiprotozoaires/sang , Antipaludiques/administration et posologie , Antipaludiques/effets indésirables , Antipaludiques/pharmacocinétique , Relation dose-effet des médicaments , Volontaires sains , Humains , Perfusions veineuses/effets indésirables , Injections sous-cutanées/effets indésirables , Adulte d'âge moyen , Plasmodium falciparum/immunologie , Plasmodium falciparum/isolement et purification
13.
Immunohorizons ; 5(6): 466-476, 2021 06 17.
Article Dans Anglais | MEDLINE | ID: covidwho-1359325

Résumé

Lasting immunity will be critical for overcoming COVID-19. However, the factors associated with the development of high titers of anti-SARS-CoV-2 Abs and how long those Abs persist remain incompletely defined. In particular, an understanding of the relationship between COVID-19 symptoms and anti-SARS-CoV-2 Abs is limited. To address these unknowns, we quantified serum anti-SARS- CoV-2 Abs in clinically diverse COVID-19 convalescent human subjects 5 wk (n = 113) and 3 mo (n = 79) after symptom resolution with three methods: a novel multiplex assay to quantify IgG against four SARS-CoV-2 Ags, a new SARS-CoV-2 receptor binding domain-angiotensin converting enzyme 2 inhibition assay, and a SARS-CoV-2 neutralizing assay. We then identified clinical and demographic factors, including never-before-assessed COVID-19 symptoms, that consistently correlate with high anti-SARS-CoV-2 Ab levels. We detected anti-SARS-CoV-2 Abs in 98% of COVID-19 convalescent subjects 5 wk after symptom resolution, and Ab levels did not decline at 3 mo. Greater disease severity, older age, male sex, higher body mass index, and higher Charlson Comorbidity Index score correlated with increased anti-SARS-CoV-2 Ab levels. Moreover, we report for the first time (to our knowledge) that COVID-19 symptoms, most consistently fever, body aches, and low appetite, correlate with higher anti-SARS-CoV-2 Ab levels. Our results provide robust and new insights into the development and persistence of anti-SARS-CoV-2 Abs.


Sujets)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , Immunoglobuline G/immunologie , SARS-CoV-2/immunologie , Adulte , Anticorps neutralisants/sang , Anticorps antiviraux/sang , COVID-19/épidémiologie , COVID-19/virologie , Études de cohortes , Femelle , Hospitalisation/statistiques et données numériques , Humains , Immunoglobuline G/sang , Modèles linéaires , Mâle , Adulte d'âge moyen , Analyse multifactorielle , Pandémies , SARS-CoV-2/physiologie , Indice de gravité de la maladie , Facteurs temps
14.
J Infect Dis ; 224(1): 49-59, 2021 07 02.
Article Dans Anglais | MEDLINE | ID: covidwho-1294731

Résumé

BACKGROUND: We investigated frequency of reinfection with seasonal human coronaviruses (HCoVs) and serum antibody response following infection over 8 years in the Household Influenza Vaccine Evaluation (HIVE) cohort. METHODS: Households were followed annually for identification of acute respiratory illness with reverse-transcription polymerase chain reaction-confirmed HCoV infection. Serum collected before and at 2 time points postinfection were tested using a multiplex binding assay to quantify antibody to seasonal, severe acute respiratory syndrome coronavirus (SARS-CoV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike proteins and SARS-CoV-2 spike subdomains and N protein. RESULTS: Of 3418 participants, 40% were followed for ≥3 years. A total of 1004 HCoV infections were documented; 303 (30%) were reinfections of any HCoV type. The number of HCoV infections ranged from 1 to 13 per individual. The mean time to reinfection with the same type was estimated at 983 days for 229E, 578 days for HKU1, 615 days for OC43, and 711 days for NL63. Binding antibody levels to seasonal HCoVs were high, with little increase postinfection, and were maintained over time. Homologous, preinfection antibody levels did not significantly correlate with odds of infection, and there was little cross-response to SARS-CoV-2 proteins. CONCLUSIONS: Reinfection with seasonal HCoVs is frequent. Binding anti-spike protein antibodies do not correlate with protection from seasonal HCoV infection.


Sujets)
Infections à coronavirus/épidémiologie , Coronavirus , Caractéristiques familiales , Vaccins antigrippaux/immunologie , Grippe humaine/épidémiologie , Grippe humaine/prévention et contrôle , Syndrome respiratoire aigu sévère/épidémiologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , COVID-19/épidémiologie , COVID-19/virologie , Co-infection/épidémiologie , Coronavirus/classification , Coronavirus/génétique , Coronavirus/immunologie , Infections à coronavirus/diagnostic , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Réactions croisées/immunologie , Humains , Vaccins antigrippaux/administration et posologie , Grippe humaine/virologie , Estimation de Kaplan-Meier , Michigan/épidémiologie , Modèles des risques proportionnels , Surveillance de la santé publique , Réinfection/épidémiologie , RT-PCR , SARS-CoV-2 , Saisons , Études séroépidémiologiques , Syndrome respiratoire aigu sévère/diagnostic , Syndrome respiratoire aigu sévère/immunologie , Syndrome respiratoire aigu sévère/virologie , Charge virale
15.
JCI Insight ; 6(8)2021 04 22.
Article Dans Anglais | MEDLINE | ID: covidwho-1197300

Résumé

Preexisting cross-reactivity to SARS-CoV-2 occurs in the absence of prior viral exposure. However, this has been difficult to quantify at the population level due to a lack of reliably defined seroreactivity thresholds. Using an orthogonal antibody testing approach, we estimated that about 0.6% of nontriaged adults from the greater Vancouver, Canada, area between May 17 and June 19, 2020, showed clear evidence of a prior SARS-CoV-2 infection, after adjusting for false-positive and false-negative test results. Using a highly sensitive multiplex assay and positive/negative thresholds established in infants in whom maternal antibodies have waned, we determined that more than 90% of uninfected adults showed antibody reactivity against the spike protein, receptor-binding domain (RBD), N-terminal domain (NTD), or the nucleocapsid (N) protein from SARS-CoV-2. This seroreactivity was evenly distributed across age and sex, correlated with circulating coronaviruses' reactivity, and was partially outcompeted by soluble circulating coronaviruses' spike. Using a custom SARS-CoV-2 peptide mapping array, we found that this antibody reactivity broadly mapped to spike and to conserved nonstructural viral proteins. We conclude that most adults display preexisting antibody cross-reactivity against SARS-CoV-2, which further supports investigation of how this may impact the clinical severity of COVID-19 or SARS-CoV-2 vaccine responses.


Sujets)
Anticorps neutralisants/sang , Anticorps antiviraux/sang , Antigènes viraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , Adulte , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Colombie-Britannique/épidémiologie , COVID-19/sang , COVID-19/diagnostic , COVID-19/prévention et contrôle , Dépistage sérologique de la COVID-19/statistiques et données numériques , Vaccins contre la COVID-19/administration et posologie , Réactions croisées/immunologie , Études transversales , Femelle , Géographie , Volontaires sains , Humains , Immunité humorale , Dosage immunologique/statistiques et données numériques , Mâle , Adulte d'âge moyen , Études prospectives , SARS-CoV-2/isolement et purification , Indice de gravité de la maladie
SÉLECTION CITATIONS
Détails de la recherche