202 results on '"Robert C. Read"'
Search Results
2. Distinct early cellular kinetics in participants protected against colonization upon Bordetella pertussis challenge
- Author
-
Annieck M. Diks, Hans de Graaf, Cristina Teodosio, Rick J. Groenland, Bas de Mooij, Muktar Ibrahim, Alison R. Hill, Robert C. Read, Jacques J.M. van Dongen, Magdalena A. Berkowska, and on behalf of the IMI-2 PERISCOPE Consortium
- Subjects
Immunology ,Infectious disease ,Medicine - Abstract
BACKGROUND To date, only limited data are available on the mechanisms of protection against colonization with Bordetella pertussis in humans.METHODS In this study, the cellular responses to B. pertussis challenge were monitored longitudinally using high-dimensional EuroFlow-based flow cytometry, allowing quantitative detection of more than 250 different immune cell subsets in the blood of 15 healthy donors.RESULTS Participants who were protected against colonization showed different early cellular responses compared with colonized participants. Especially prominent for colonization-protected participants were the early expansion of CD36– nonclassical monocytes on day 1 (D1), natural killer cells (D3), follicular T helper cells (D1–D3), and plasma cells (D3). Plasma cell expansion on D3 correlated negatively with the CFU load on D7 and D9 after challenge. Increased plasma cell maturation on D11–D14 was found in participants with seroconversion.CONCLUSION These early cellular immune responses following experimental infection can now be further characterized and potentially linked to an efficient mucosal immune response, preventing colonization. Ultimately, their presence may be used to evaluate whether new B. pertussis vaccine candidates are protective against B. pertussis colonization, e.g., by bacterial challenge after vaccination.TRIAL REGISTRATION ClinicalTrials.gov NCT03751514.FUNDING Innovative Medicines Initiative 2 Joint Undertaking and the EuroFlow Consortium.
- Published
- 2023
- Full Text
- View/download PDF
3. The role of public involvement in the design of the first SARS-CoV-2 human challenge study during an evolving pandemic
- Author
-
Maria Piggin, Emma Smith, Peter Mankone, Leah Ndegwa, Diane Gbesemete, Philippa Pristerà, Michael Bahrami-Hessari, Halle Johnson, Andrew P. Catchpole, Peter J.M. Openshaw, Christopher Chiu, Robert C. Read, Helen Ward, and Caroline Barker
- Subjects
Ethics ,Human challenge study ,Public involvement ,SARS-CoV-2 ,Infectious and parasitic diseases ,RC109-216 - Abstract
High quality health care research must involve patients and the public. This ensures research is important, relevant and acceptable to those it is designed to benefit. The world’s first human challenge study with SARS-CoV-2 undertook detailed public involvement to inform study design despite the urgency to review and establish the study. The work was integral to the UK Research Ethics Committee review and approval of the study. Discussion with individuals from ethnic minorities within the UK population supported decision-making around the study exclusion criteria. Public review of study materials for consent processes led to the addition of new information, comparisons and visual aids to help volunteers consider the practicalities and risks involved in participating. A discussion exploring the acceptability of a human challenge study with SARS-CoV-2 taking place in the UK, given the current context of the pandemic, identified overall support for the study. Public concern for the wellbeing of trial participants, as a consequence of isolation, was identified. We outline our approach to public involvement and its impact on study design.
- Published
- 2022
- Full Text
- View/download PDF
4. Public attitudes to a human challenge study with SARS-CoV-2: a mixed-methods study [version 1; peer review: 2 approved]
- Author
-
Michael Bahrami-Hessari, Emma Smith, Katherine Baker, Halle Johnson, Carmel McGrath, Ambar Qavi, Robert C. Read, Christopher Chiu, Caroline Barker, Helen Ward, Diane Gbesemete, Katharine Collet, Daniella Watson, Maria Piggin, Wendy Lawerence, and Philippa Pristerà
- Subjects
Ethics ,human challenge study ,consultation ,public ,acceptability ,COVID-19 ,eng ,Medicine ,Science - Abstract
Background: Human challenge studies involve the deliberate exposure of healthy volunteers to an infectious micro-organism in a highly controlled and monitored way. They are used to understand infectious diseases and have contributed to the development of vaccines. In early 2020, the UK started exploring the feasibility of establishing a human challenge study with SARS-CoV-2. Given the significant public interest and the complexity of the potential risks and benefits, it is vital that public views are considered in the design and approval of any such study and that investigators and ethics boards remain accountable to the public. Methods: Mixed methods study comprising online surveys conducted with 2,441 UK adults and in-depth virtual focus groups with 57 UK adults during October 2020 to explore the public’s attitudes to a human challenge study with SARS-CoV-2 taking place in the UK. Results: There was overall agreement across the surveys and focus groups that a human challenge study with SARS-CoV-2 should take place in the UK. Transparency of information, trust and the necessity to provide clear information on potential risks to study human challenge study participants were important. The perceived risks of taking part included the risk of developing long-term effects from COVID, impact on personal commitments and mental health implications of isolation. There were a number of practical realities to taking part that would influence a volunteer’s ability to participate (e.g. Wi-Fi, access to exercise, outside space and work, family and pet commitments). Conclusions: The results identified practical considerations for teams designing human challenge studies. Recommendations were grouped: 1) messaging to potential study participants, 2) review of the protocol and organisation of the study, and 3) more broadly, making the study more inclusive and relevant. This study highlights the value of public consultation in research, particularly in fields attracting public interest and scrutiny.
- Published
- 2022
- Full Text
- View/download PDF
5. ACCORD: A Multicentre, Seamless, Phase 2 Adaptive Randomisation Platform Study to Assess the Efficacy and Safety of Multiple Candidate Agents for the Treatment of COVID-19 in Hospitalised Patients: A structured summary of a study protocol for a randomised controlled trial
- Author
-
Tom Wilkinson, Rupert Dixon, Clive Page, Miles Carroll, Gareth Griffiths, Ling-Pei Ho, Anthony De Soyza, Timothy Felton, Keir E. Lewis, Karen Phekoo, James D. Chalmers, Anthony Gordon, Lorcan McGarvey, Jillian Doherty, Robert C. Read, Manu Shankar-Hari, Nuria Martinez-Alier, Michael O’Kelly, Graeme Duncan, Roelize Walles, James Sykes, Charlotte Summers, Dave Singh, and on behalf of the ACCORD Collaborators
- Subjects
COVID-19 ,randomised ,platform study ,master protocol ,phase II ,Medicine (General) ,R5-920 - Abstract
Abstract Objectives Stage 1: To evaluate the safety and efficacy of candidate agents as add-on therapies to standard of care (SoC) in patients hospitalised with COVID-19 in a screening stage. Stage 2: To confirm the efficacy of candidate agents selected on the basis of evidence from Stage 1 in patients hospitalised with COVID-19 in an expansion stage. Trial design ACCORD is a seamless, Phase 2, adaptive, randomised controlled platform study, designed to rapidly test candidate agents in the treatment of COVID-19. Designed as a master protocol with each candidate agent being included via its own sub-protocol, initially randomising equally between each candidate and a single contemporaneous SoC arm (which can adapt into 2:1). Candidate agents currently include bemcentinib, MEDI3506, acalabrutinib, zilucoplan and nebulised heparin. For each candidate a total of 60 patients will be recruited in Stage 1. If Stage 1 provides evidence of efficacy and acceptable safety the candidate will enter Stage 2 where a total of approximately 126 patients will be recruited into each study arm sub-protocol. Enrollees and outcomes will not be shared across the Stages; the endpoint, analysis and sample size for Stage 2 may be adjusted based on evidence from Stage 1. Additional arms may be added as new potential candidate agents are identified via candidate agent specific sub-protocols. Participants The study will include hospitalised adult patients (≥18 years) with confirmed SARS-CoV-2 infection, the virus that causes COVID-19, that clinically meet Grades 3 (hospitalised – mild disease, no oxygen therapy), Grades 4 (hospitalised, oxygen by mask or nasal prongs) and 5 (hospitalised, non-invasive ventilation or high flow oxygen) of the WHO Working Group on the Clinical Characteristics of COVID-19 9-point category ordinal scale. Participants will be recruited from England, Northern Ireland, Wales and Scotland. Intervention and comparator Comparator is current standard of care (SoC) for the treatment of COVID-19. Current candidate experimental arms include bemcentinib, MEDI3506, acalabrutinib, zilucoplan and nebulised heparin with others to be added over time. Bemcentinib could potentially reduce viral infection and blocks SARS-CoV-2 spike protein; MEDI3506 is a clinic-ready anti-IL-33 monoclonal antibody with the potential to treat respiratory failure caused by COVID; acalabrutinib is a BTK inhibitor which is anti-viral and anti-inflammatory; zilucoplan is a complement C5 inhibitor which may block the severe inflammatory response in COVID-19 and; nebulised heparin has been shown to bind with the spike protein. ACCORD is linked with the UK national COVID therapeutics task force to help prioritise candidate agents. Main outcomes Time to sustained clinical improvement of at least 2 points (from randomisation) on the WHO 9-point category ordinal scale, live discharge from the hospital, or considered fit for discharge (a score of 0, 1, or 2 on the ordinal scale), whichever comes first, by Day 29 (this will also define the “responder” for the response rate analyses). Randomisation An electronic randomization will be performed by Cenduit using Interactive Response Technology (IRT). Randomisation will be stratified by baseline severity grade. Randomisation will proceed with an equal allocation to each arm and a contemporaneous SoC arm (e.g. 1:1 if control and 1 experimental arm; 1:1:1 if two experimental candidate arms etc) but will be reviewed as the trial progresses and may be changed to 2:1 in favour of the candidate agents. Blinding (masking) The trial is open label and no blinding is currently planned in the study. Numbers to be randomised (sample size) This will be in the order of 60 patients per candidate agent for Stage 1, and 126 patients for Stage 2. However, sample size re-estimation may be considered after Stage 1. It is estimated that up to 1800 patients will participate in the overall study. Trial Status Master protocol version ACCORD-2-001 - Master Protocol (Amendment 1) 22nd April 2020, the trial has full regulatory approval and recruitment is ongoing in the bemcentinib (first patient recruited 6/5/2020), MEDI3506 (first patient recruited 19/5/2020), acalabrutinib (first patient recruited 20/5/2020) and zilucoplan (first patient recruited 19/5/2020) candidates (and SoC). The recruitment dates of each arm will vary between candidate agents as they are added or dropped from the trial, but will have recruited and reported within a year. Trial registration EudraCT 2020-001736-95 , registered 28th April 2020. Full protocol The full protocol (Master Protocol with each of the candidate sub-protocols) is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.
- Published
- 2020
- Full Text
- View/download PDF
6. Microevolution of Neisseria lactamica during nasopharyngeal colonisation induced by controlled human infection
- Author
-
Anish Pandey, David W. Cleary, Jay R. Laver, Andrew Gorringe, Alice M. Deasy, Adam P. Dale, Paul D. Morris, Xavier Didelot, Martin C. J. Maiden, and Robert C. Read
- Subjects
Science - Abstract
Carriage of Neisseria lactamica, a harmless coloniser of the human respiratory tract, is inversely correlated with Neisseria meningitidis infection. Here, Pandey et al. provide insights into micro-evolutionary processes in N. lactamica during controlled infection of healthy volunteers.
- Published
- 2018
- Full Text
- View/download PDF
7. Neisseria lactamica Y92–1009 complete genome sequence
- Author
-
Anish K. Pandey, David W. Cleary, Jay R. Laver, Martin C. J. Maiden, Xavier Didelot, Andrew Gorringe, and Robert C. Read
- Subjects
SMRT cell sequencing ,Neisseria ,Short read sequencing ,Bacteria ,Genome assembly ,Nasopharyngeal microflora ,Genetics ,QH426-470 - Abstract
Abstract We present the high quality, complete genome assembly of Neisseria lactamica Y92–1009 used to manufacture an outer membrane vesicle (OMV)-based vaccine, and a member of the Neisseria genus. The strain is available on request from the Public Health England Meningococcal Reference Unit. This Gram negative, dipplococcoid bacterium is an organism of worldwide clinical interest because human nasopharyngeal carriage is related inversely to the incidence of meningococcal disease, caused by Neisseria meningitidis. The organism sequenced was isolated during a school carriage survey in Northern Ireland in 1992 and has been the subject of a variety of laboratory and clinical studies. Four SMRT cells on a RSII machine by Pacific Biosystems were used to produce a complete, closed genome assembly. Sequence data were obtained for a total of 30,180,391 bases from 2621 reads and assembled using the HGAP algorithm. The assembly was corrected using short reads obtained from an Illumina HiSeq 2000instrument. This resulted in a 2,146,723 bp assembly with approximately 460 fold mean coverage depth and a GC ratio of 52.3%.
- Published
- 2017
- Full Text
- View/download PDF
8. The environmental deposition of influenza virus from patients infected with influenza A(H1N1)pdm09: Implications for infection prevention and control
- Author
-
Benjamin Killingley, Jane Greatorex, Paul Digard, Helen Wise, Fayna Garcia, Harsha Varsani, Simon Cauchemez, Joanne E. Enstone, Andrew Hayward, Martin D. Curran, Robert C. Read, Wei S. Lim, Karl G. Nicholson, and Jonathan S. Nguyen-Van-Tam
- Subjects
Infectious and parasitic diseases ,RC109-216 ,Public aspects of medicine ,RA1-1270 - Abstract
Summary: In a multi-center, prospective, observational study over two influenza seasons, we sought to quantify and correlate the amount of virus recovered from the nares of infected subjects with that recovered from their immediate environment in community and hospital settings. We recorded the symptoms of adults and children with A(H1N1)pdm09 infection, took nasal swabs, and sampled touched surfaces and room air. Forty-two infected subjects were followed up. The mean duration of virus shedding was 6.2 days by PCR (Polymerase Chain Reaction) and 4.2 days by culture. Surface swabs were collected from 39 settings; 16 (41%) subject locations were contaminated with virus. Overall, 33 of the 671 (4.9%) surface swabs were PCR positive for influenza, of which two (0.3%) yielded viable virus. On illness Day 3, subjects yielding positive surface samples had significantly higher nasal viral loads (geometric mean ratio 25.7; 95% CI 1.75, 376.0, p = 0.021) and a positive correlation (r = 0.47, p = 0.006) was observed between subject nasal viral loads and viral loads recovered from the surfaces around them. Room air was sampled in the vicinity of 12 subjects, and PCR positive samples were obtained for five (42%) samples. Influenza virus shed by infected subjects did not detectably contaminate the vast majority of surfaces sampled. We question the relative importance of the indirect contact transmission of influenza via surfaces, though our data support the existence of super-spreaders via this route. The air sampling results add to the accumulating evidence that supports the potential for droplet nuclei (aerosol) transmission of influenza. Keywords: Influenza, Environmental, Deposition, Infection, Control
- Published
- 2016
- Full Text
- View/download PDF
9. Erratum: Blume, C., et al. Modulation of Human Airway Barrier Functions during Burkholderia thailandensis and Francisella tularensis Infection Running Title: Airway Barrier Functions during Bacterial Infections. Pathogens 2016, 5, 53
- Author
-
Cornelia Blume, Jonathan David, Rachel E. Bell, Jay R. Laver, Robert C. Read, Graeme C. Clark, Donna E. Davies, and Emily J. Swindle
- Subjects
n/a ,Medicine - Abstract
There is an error in the title [...]
- Published
- 2020
- Full Text
- View/download PDF
10. Summary of the international clinical guidelines for the management of hospital-acquired and ventilator-acquired pneumonia
- Author
-
Antoni Torres, Michael S. Niederman, Jean Chastre, Santiago Ewig, Patricia Fernandez-Vandellos, Hakan Hanberger, Marin Kollef, Gianluigi Li Bassi, Carlos M. Luna, Ignacio Martin-Loeches, J. Artur Paiva, Robert C. Read, David Rigau, Jean François Timsit, Tobias Welte, and Richard Wunderink
- Subjects
Medicine - Published
- 2018
- Full Text
- View/download PDF
11. Correction to: Neisseria lactamica Y92–1009 complete genome sequence
- Author
-
Anish K. Pandey, David W. Cleary, Jay R. Laver, Martin C. J. Maiden, Xavier Didelot, Andrew Gorringe, and Robert C. Read
- Subjects
Genetics ,QH426-470 - Abstract
Correction After publication of the original article [1] it was identified that an incorrect version of the manuscript has been published. This was caused by a technical error which led to a discrepancy between the editorially-accepted version of the manuscript, and the published version.
- Published
- 2018
- Full Text
- View/download PDF
12. Nosocomial Pandemic (H1N1) 2009, United Kingdom, 2009–2010
- Author
-
Joanne E. Enstone, Puja R. Myles, Peter J.M. Openshaw, Elaine M. Gadd, Wei Shen Lim, Malcolm G. Semple, Robert C. Read, Bruce L. Taylor, James McMenamin, Colin Armstrong, Barbara Bannister, Karl G. Nicholson, and Jonathan S. Nguyen-Van-Tam
- Subjects
Nosocomial infections ,influenza ,pandemic (H1N1) 2009 ,influenza A virus ,infection control ,hospitalization ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
To determine clinical characteristics of patients hospitalized in the United Kingdom with pandemic (H1N1) 2009, we studied 1,520 patients in 75 National Health Service hospitals. We characterized patients who acquired influenza nosocomially during the pandemic (H1N1) 2009 outbreak. Of 30 patients, 12 (80%) of 15 adults and 14 (93%) of 15 children had serious underlying illnesses. Only 12 (57%) of 21 patients who received antiviral therapy did so within 48 hours after symptom onset, but 53% needed escalated care or mechanical ventilation; 8 (27%) of 30 died. Despite national guidelines and standardized infection control procedures, nosocomial transmission remains a problem when influenza is prevalent. Health care workers should be routinely offered influenza vaccine, and vaccination should be prioritized for all patients at high risk. Staff should remain alert to the possibility of influenza in patients with complex clinical problems and be ready to institute antiviral therapy while awaiting diagnosis during influenza outbreaks.
- Published
- 2011
- Full Text
- View/download PDF
13. Pneumolysin Activates Macrophage Lysosomal Membrane Permeabilization and Executes Apoptosis by Distinct Mechanisms without Membrane Pore Formation
- Author
-
Martin A. Bewley, Michael Naughton, Julie Preston, Andrea Mitchell, Ashleigh Holmes, Helen M. Marriott, Robert C. Read, Timothy J. Mitchell, Moira K. B. Whyte, and David H. Dockrell
- Subjects
Microbiology ,QR1-502 - Abstract
ABSTRACT Intracellular killing of Streptococcus pneumoniae is complemented by induction of macrophage apoptosis. Here, we show that the toxin pneumolysin (PLY) contributes both to lysosomal/phagolysosomal membrane permeabilization (LMP), an upstream event programing susceptibility to apoptosis, and to apoptosis execution via a mitochondrial pathway, through distinct mechanisms. PLY is necessary but not sufficient for the maximal induction of LMP and apoptosis. PLY’s ability to induce both LMP and apoptosis is independent of its ability to form cytolytic pores and requires only the first three domains of PLY. LMP involves TLR (Toll-like receptor) but not NLRP3/ASC (nucleotide-binding oligomerization domain [Nod]-like receptor family, pyrin domain-containing protein 3/apoptosis-associated speck-like protein containing a caspase recruitment domain) signaling and is part of a PLY-dependent but phagocytosis-independent host response that includes the production of cytokines, including interleukin-1 beta (IL-1β). LMP involves progressive and selective permeability to 40-kDa but not to 250-kDa fluorescein isothiocyanate (FITC)-labeled dextran, as PLY accumulates in the cytoplasm. In contrast, the PLY-dependent execution of apoptosis requires phagocytosis and is part of a host response to intracellular bacteria that also includes NO generation. In cells challenged with PLY-deficient bacteria, reconstitution of LMP using the lysomotrophic detergent LeuLeuOMe favored cell necrosis whereas PLY reconstituted apoptosis. The results suggest that PLY contributes to macrophage activation and cytokine production but also engages LMP. Following bacterial phagocytosis, PLY triggers apoptosis and prevents macrophage necrosis as a component of a broad-based antimicrobial strategy. This illustrates how a key virulence factor can become the focus of a multilayered and coordinated innate response by macrophages, optimizing pathogen clearance and limiting inflammation. IMPORTANCE Streptococcus pneumoniae, the commonest cause of bacterial pneumonia, expresses the toxin pneumolysin, which can make holes in cell surfaces, causing tissue damage. Macrophages, resident immune cells essential for responses to bacteria in tissues, activate a program of cell suicide called apoptosis, maximizing bacterial clearance and limiting harmful inflammation. We examined pneumolysin’s role in activating this response. We demonstrate that pneumolysin did not directly form holes in cells to trigger apoptosis and show that pneumolysin has two distinct roles which require only part of the molecule. Pneumolysin and other bacterial factors released by bacteria that have not been eaten by macrophages activate macrophages to release inflammatory factors but also make the cell compartment containing ingested bacteria leaky. Once inside the cell, pneumolysin ensures that the bacteria activate macrophage apoptosis, rather than necrosis, enhancing bacterial killing and limiting inflammation. This dual response to pneumolysin is critical for an effective immune response to S. pneumoniae.
- Published
- 2014
- Full Text
- View/download PDF
14. Modulation of Human Airway Barrier Functions during Burkholderia thailandensis and Francisella tularensis Infection Running Title: Airway Barrier Functions during Bacterial Infections
- Author
-
Cornelia Blume, Jonathan David, Rachel E. Bell, Jay R. Laver, Robert C. Read, Graeme C. Clark, Donna E. Davies, and Emily J. Swindle
- Subjects
airway epithelium ,bacterial infection ,Burkholderia thailandensis ,Fransicella tularensis ,barrier functions ,bacterial passage ,fluticasone propionate ,Medicine - Abstract
The bronchial epithelium provides protection against pathogens from the inhaled environment through the formation of a highly-regulated barrier. In order to understand the pulmonary diseases melioidosis and tularemia caused by Burkholderia thailandensis and Fransicella tularensis, respectively, the barrier function of the human bronchial epithelium were analysed. Polarised 16HBE14o- or differentiated primary human bronchial epithelial cells (BECs) were exposed to increasing multiplicities of infection (MOI) of B. thailandensis or F. tularensis Live Vaccine Strain and barrier responses monitored over 24–72 h. Challenge of polarized BECs with either bacterial species caused an MOI- and time-dependent increase in ionic permeability, disruption of tight junctions, and bacterial passage from the apical to the basolateral compartment. B. thailandensis was found to be more invasive than F. tularensis. Both bacterial species induced an MOI-dependent increase in TNF-α release. An increase in ionic permeability and TNF-α release was induced by B. thailandensis in differentiated BECs. Pretreatment of polarised BECs with the corticosteroid fluticasone propionate reduced bacterial-dependent increases in ionic permeability, bacterial passage, and TNF-α release. TNF blocking antibody Enbrel® reduced bacterial passage only. BEC barrier properties are disrupted during respiratory bacterial infections and targeting with corticosteroids or anti-TNF compounds may represent a therapeutic option.
- Published
- 2016
- Full Text
- View/download PDF
15. Correction: Correlation of Group C Meningococcal Conjugate Vaccine Response with B- and T-Lymphocyte Activity.
- Author
-
James B. Wing, Lynne Smart, Ray Borrow, Jamie Findlow, Helen Findlow, Andrew Lees, Robert C. Read, and Andrew W. Heath
- Subjects
Medicine ,Science - Published
- 2012
- Full Text
- View/download PDF
16. Effect of colonisation with Neisseria lactamica on cross-reactive anti-meningococcal B-cell responses: a randomised, controlled, human infection trial
- Author
-
Adam P Dale, Anastasia A Theodosiou, Diane F Gbesemete, Jonathan M Guy, Eleanor F Jones, Alison R Hill, Muktar M Ibrahim, Hans de Graaf, Muhammad Ahmed, Saul N Faust, Andrew R Gorringe, Marta E Polak, Jay R Laver, and Robert C Read
- Subjects
Adult ,Microbiology (medical) ,Infectious Diseases ,Immunoglobulin G ,Virology ,Immunoglobulin A, Secretory ,Leukocytes, Mononuclear ,Humans ,Saline Solution ,Neisseria meningitidis ,Microbiology ,Neisseria lactamica ,Phosphates - Abstract
Pharyngeal colonisation by the commensal bacterium Neisseria lactamica inhibits colonisation by Neisseria meningitidis and has an inverse epidemiological association with meningococcal disease. The mechanisms that underpin this relationship are unclear, but could involve the induction of cross-reactive immunity. In this study, we aimed to evaluate whether colonisation with N lactamica induces N lactamica-specific B-cell responses that are cross-reactive with N meningitidis.In this randomised, placebo-controlled, human infection trial at University Hospital Southampton Clinical Research Facility (Southampton, UK), healthy adults aged 18-45 years were randomly assigned (2:1) to receive intranasal inoculation with either 10Of 50 participants assessed for eligibility between Sept 5, 2018, and March 3, 2019, 31 were randomly assigned (n=20 N lactamica, n=11 PBS). Among the 17 participants who were colonised with N lactamica, the median baselines compared with peak post-colonisation N lactamica-specific plasma-cell frequencies (per 10Natural immunity to N meningitidis after colonisation with N lactamica might be due to cross-reactive adaptive responses. Exploitation of this microbial mechanism with a genetically modified live vector could protect against N meningitidis colonisation and disease.Wellcome Trust, Medical Research Council, and NIHR Southampton Biomedical Research Centre.
- Published
- 2022
- Full Text
- View/download PDF
17. Persistence of immune response in heterologous COVID vaccination schedules in the Com-COV2 study - a single-blind, randomised trial incorporating mRNA, viral-vector and protein-adjuvant vaccines
- Author
-
Robert H. Shaw, Melanie Greenland, Arabella S.V. Stuart, Parvinder K. Aley, Nick J. Andrews, J. Claire Cameron, Sue Charlton, Elizabeth A. Clutterbuck, Andrea M. Collins, Tom Darton, Tanya Dinesh, Christopher J.A. Duncan, Saul N. Faust, Daniela M. Ferreira, Adam Finn, Anna L. Goodman, Christopher A. Green, Bassam Hallis, Paul T. Heath, Helen Hill, Teresa Lambe, Vincenzo Libri, Patrick J. Lillie, Ella Morey, Yama F. Mujadidi, Ruth Payne, Emma L. Plested, Samuel Provstgaard-Morys, Maheshi N. Ramasamy, Mary Ramsay, Robert C. Read, Hannah Robinson, Gavin R. Screaton, Nisha Singh, David P.J. Turner, Paul J. Turner, Rachel White, Jonathan S. Nguyen-Van-Tam, Xinxue Liu, and Matthew D. Snape
- Subjects
Microbiology (medical) ,Infectious Diseases - Abstract
Background: Heterologous COVID vaccine priming schedules are immunogenic and effective. This report aims to understand the persistence of immune response to the viral vectored, mRNA and protein-based COVID-19 vaccine platforms used in homologous and heterologous priming combinations, which will inform the choice of vaccine platform in future vaccine development. Methods: Com-COV2 was a single-blinded trial in which adults ≥ 50 years, previously immunised with single dose ‘ChAd’ (ChAdOx1 nCoV-19, AZD1222, Vaxzevria, Astrazeneca) or ‘BNT’ (BNT162b2, tozinameran, Comirnaty, Pfizer/BioNTech), were randomised 1:1:1 to receive a second dose 8–12 weeks later with either the homologous vaccine, or ‘Mod’ (mRNA-1273, Spikevax, Moderna) or ‘NVX’ (NVX-CoV2373, Nuvaxovid, Novavax). Immunological follow-up and the secondary objective of safety monitoring were performed over nine months. Analyses of antibody and cellular assays were performed on an intention-to-treat population without evidence of COVID-19 infection at baseline or for the trial duration. Findings: In April/May 2021, 1072 participants were enrolled at a median of 9.4 weeks after receipt of a single dose of ChAd (N = 540, 45% female) or BNT (N = 532, 39% female) as part of the national vaccination programme. In ChAd-primed participants, ChAd/Mod had the highest anti-spike IgG from day 28 through to 6 months, although the heterologous vs homologous geometric mean ratio (GMR) dropped from 9.7 (95% CI (confidence interval): 8.2, 11.5) at D28 to 6.2 (95% CI: 5.0, 7.7) at D196. The heterologous/homologous GMR for ChAd/NVX similarly dropped from 3.0 (95% CI:2.5,3.5) to 2.4 (95% CI:1.9, 3.0). In BNT-primed participants, decay was similar between heterologous and homologous schedules with BNT/Mod inducing the highest anti-spike IgG for the duration of follow-up. The adjusted GMR (aGMR) for BNT/Mod compared with BNT/BNT increased from 1.36 (95% CI: 1.17, 1.58) at D28 to 1.52 (95% CI: 1.21, 1.90) at D196, whilst for BNT/NVX this aGMR was 0.55 (95% CI: 0.47, 0.64) at day 28 and 0.62 (95% CI: 0.49, 0.78) at day 196. Heterologous ChAd-primed schedules produced and maintained the largest T-cell responses until D196. Immunisation with BNT/NVX generated a qualitatively different antibody response to BNT/BNT, with the total IgG significantly lower than BNT/BNT during all follow-up time points, but similar levels of neutralising antibodies. Interpretation: Heterologous ChAd-primed schedules remain more immunogenic over time in comparison to ChAd/ChAd. BNT-primed schedules with a second dose of either mRNA vaccine also remain more immunogenic over time in comparison to BNT/NVX. The emerging data on mixed schedules using the novel vaccine platforms deployed in the COVID-19 pandemic, suggest that heterologous priming schedules might be considered as a viable option sooner in future pandemics. ISRCTN:27841311 EudraCT:2021-001275-16.
- Published
- 2023
18. Corrigendum to 'Persistence of immunogenicity after seven COVID-19 vaccines given as third dose boosters following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK: Three month analyses of the COV-BOOST trial' [J Infect 84(6) (2022) 795–813, 5511]
- Author
-
Xinxue Liu, Alasdair PS Munro, Shuo Feng, Leila Janani, Parvinder K Aley, Gavin Babbage, David Baxter, Marcin Bula, Katrina Cathie, Krishna Chatterjee, Wanwisa Dejnirattisai, Kate Dodd, Yvanne Enever, Ehsaan Qureshi, Anna L. Goodman, Christopher A Green, Linda Harndahl, John Haughney, Alexander Hicks, Agatha A. van der Klaauw, Jonathan Kwok, Vincenzo Libri, Martin J Llewelyn, Alastair C McGregor, Angela M. Minassian, Patrick Moore, Mehmood Mughal, Yama F Mujadidi, Kyra Holliday, Orod Osanlou, Rostam Osanlou, Daniel R Owens, Mihaela Pacurar, Adrian Palfreeman, Daniel Pan, Tommy Rampling, Karen Regan, Stephen Saich, Teona Serafimova, Dinesh Saralaya, Gavin R Screaton, Sunil Sharma, Ray Sheridan, Ann Sturdy, Piyada Supasa, Emma C Thomson, Shirley Todd, Chris Twelves, Robert C. Read, Sue Charlton, Bassam Hallis, Mary Ramsay, Nick Andrews, Teresa Lambe, Jonathan S Nguyen-Van-Tam, Victoria Cornelius, Matthew D Snape, and Saul N Faust
- Subjects
Microbiology (medical) ,Infectious Diseases - Published
- 2023
- Full Text
- View/download PDF
19. Neisseria lactamica controlled human infection model
- Author
-
Jay R. Laver, Robert C. Read, Diane Gbesemete, Adam P. Dale, Bidmos, Fadil, Bossé, Janine, and Langford, Paul
- Subjects
biology ,Neisseria meningitidis ,biology.organism_classification ,medicine.disease_cause ,law.invention ,Microbiology ,Prolonged exposure ,Immune system ,medicine.anatomical_structure ,stomatognathic system ,Antigen ,law ,Neisseria lactamica ,otorhinolaryngologic diseases ,medicine ,Colonization ,Polymerase chain reaction ,Respiratory tract - Abstract
Neisseria lactamica is a nonpathogenic commensal of the human upper respiratory tract that has been associated with protection against N. meningitidis colonization and disease. We have previously utilized the N. lactamica controlled human infection model to investigate the protective effect of N. lactamica colonization on N. meningitidis colonization, the nature of cross-reactive immune responses mounted toward N. meningitidis following N. lactamica colonization, and the microevolution of N. lactamica over a 5-month colonization period. More recently, we have assessed the possibility of utilizing genetically modified strains of N. lactamica to enable use of the commensal as a vehicle for prolonged exposure of the nasopharynx of humans to antigens of interest, expressed in carried organisms. A controlled infection with N. lactamica expressing the meningococcal antigen NadA has been executed and the results demonstrate that this strategy is effective at generating immune responses to the target antigen. Throughout this chapter, we outline in a step-by-step manner the methodologies utilized when performing controlled human infection with N. lactamica including procedures relating to: (1) the dilution of N. lactamica stock vials to derive intranasal inocula, (2) the delivery of intranasal inocula to human volunteers, (3) the determination of N. lactamica colonization status following intranasal inoculation using oropharyngeal swabbing and nasal wash sampling, (4) the microbiological procedures utilized to identify N. lactamica colonization among study volunteers, and (5) the identification of N. lactamica colonies as strain Y92-1009 using polymerase chain reaction.
- Published
- 2022
20. Effect of priming interval on reactogenicity, peak immunological response, and waning after homologous and heterologous COVID-19 vaccine schedules: exploratory analyses of Com-COV, a randomised control trial
- Author
-
Robert H Shaw, Xinxue Liu, Arabella S V Stuart, Melanie Greenland, Parvinder K Aley, Nick J Andrews, J Claire Cameron, Sue Charlton, Elizabeth A Clutterbuck, Andrea M Collins, Wanwisa Dejnirattisai, Tanya Dinesh, Saul N Faust, Daniela M Ferreira, Adam Finn, Christopher A Green, Bassam Hallis, Paul T Heath, Helen Hill, Teresa Lambe, Rajeka Lazarus, Vincenzo Libri, Fei Long, Yama F Mujadidi, Emma L Plested, Ella R Morey, Samuel Provstgaard-Morys, Maheshi N Ramasamy, Mary Ramsay, Robert C Read, Hannah Robinson, Gavin R Screaton, Nisha Singh, David P J Turner, Paul J Turner, Iason Vichos, Laura L Walker, Rachel White, Jonathan S Nguyen-Van-Tam, Matthew D Snape, Alasdair P.S. Munro, Jazz Bartholomew, Laura Presland, Sarah Horswill, Sarah Warren, Sophie Varkonyi-Clifford, Stephen Saich, Kirsty Adams, Marivic Ricamara, Nicola Turner, Nicole Y. Yee Ting, Sarah Whittley, Tommy Rampling, Amisha Desai, Claire H. Brown, Ehsaan Qureshi, Karishma Gokani, Kush Naker, Johanna K. Kellett Wright, Rachel L. Williams, Tawassal Riaz, Florentina D. Penciu, Amy Carson, Claudio Di Maso, Gracie Mead, Elizabeth G. Howe, Mujtaba Ghulam Farooq, Rabiullah Noristani, Xin L. Yao, Neil J. Oldfield, Daniel Hammersley, Sue Belton, Simon Royal, Alberto San Francisco Ramos, Cecilia Hultin, Eva P. Galiza, Rebecca Crook, Marcin Bula, Fred Fyles, Hassan Burhan, Flora Maelin, Elen Hughes, Emmanuel Okenyi, Group, Com-COV Study, and National Institute for Health and Care Research
- Subjects
Pulmonary and Respiratory Medicine ,Adult ,COVID-19 Vaccines ,wa_115 ,SARS-CoV-2 ,COVID-19/prevention & control ,Com-COV Study Group ,Immunization, Secondary ,COVID-19 ,Covid19 ,1103 Clinical Sciences ,Antibodies, Viral ,qw_806 ,qw_805 ,1117 Public Health and Health Services ,ChAdOx1 nCoV-19 ,Immunoglobulin G ,wc_506 ,Humans ,BNT162 Vaccine ,COVID-19 Vaccines/adverse effects ,1199 Other Medical and Health Sciences - Abstract
Background Priming COVID-19 vaccine schedules have been deployed at variable intervals globally, which might influence immune persistence and the relative importance of third-dose booster programmes. Here, we report exploratory analyses from the Com-COV trial, assessing the effect of 4-week versus 12-week priming intervals on reactogenicity and the persistence of immune response up to 6 months after homologous and heterologous priming schedules using the vaccines BNT162b2 (tozinameran, Pfizer/BioNTech) and ChAdOx1 nCoV-19 (AstraZeneca). Methods Com-COV was a participant-masked, randomised immunogenicity trial. For these exploratory analyses, we used the trial's general cohort, in which adults aged 50 years or older were randomly assigned to four homologous and four heterologous vaccine schedules using BNT162b2 and ChAdOx1 nCoV-19 with 4-week or 12-week priming intervals (eight groups in total). Immunogenicity analyses were done on the intention-to-treat (ITT) population, comprising participants with no evidence of SARS-CoV-2 infection at baseline or for the trial duration, to assess the effect of priming interval on humoral and cellular immune response 28 days and 6 months post-second dose, in addition to the effects on reactogenicity and safety. The Com-COV trial is registered with the ISRCTN registry, 69254139 (EudraCT 2020–005085–33). Findings Between Feb 11 and 26, 2021, 730 participants were randomly assigned in the general cohort, with 77–89 per group in the ITT analysis. At 28 days and 6 months post-second dose, the geometric mean concentration of anti-SARS-CoV-2 spike IgG was significantly higher in the 12-week interval groups than in the 4-week groups for homologous schedules. In heterologous schedule groups, we observed a significant difference between intervals only for the BNT162b2–ChAdOx1 nCoV-19 group at 28 days. Pseudotyped virus neutralisation titres were significantly higher in all 12-week interval groups versus 4-week groups, 28 days post-second dose, with geometric mean ratios of 1·4 (95% CI 1·1–1·8) for homologous BNT162b2, 1·5 (1·2–1·9) for ChAdOx1 nCoV-19–BNT162b2, 1·6 (1·3–2·1) for BNT162b2–ChAdOx1 nCoV-19, and 2·4 (1·7–3·2) for homologous ChAdOx1 nCoV-19. At 6 months post-second dose, anti-spike IgG geometric mean concentrations fell to 0·17–0·24 of the 28-day post-second dose value across all eight study groups, with only homologous BNT162b2 showing a slightly slower decay for the 12-week versus 4-week interval in the adjusted analysis. The rank order of schedules by humoral response was unaffected by interval, with homologous BNT162b2 remaining the most immunogenic by antibody response. T-cell responses were reduced in all 12-week priming intervals compared with their 4-week counterparts. 12-week schedules for homologous BNT162b2 and ChAdOx1 nCoV-19–BNT162b2 were up to 80% less reactogenic than 4-week schedules. Interpretation These data support flexibility in priming interval in all studied COVID-19 vaccine schedules. Longer priming intervals might result in lower reactogenicity in schedules with BNT162b2 as a second dose and higher humoral immunogenicity in homologous schedules, but overall lower T-cell responses across all schedules. Future vaccines using these novel platforms might benefit from schedules with long intervals. Funding UK Vaccine Taskforce and National Institute for Health and Care Research.
- Published
- 2022
- Full Text
- View/download PDF
21. Antibiotics for lower respiratory tract infection in children presenting in primary care in England (ARTIC PC): a double-blind, randomised, placebo-controlled trial
- Author
-
Guiqing Yao, Anthony Harnden, Shihua Zhu, Kim Harman, Jane Whitehurst, Charlotte Hookham, Catherine J. Woods, Geraldine Leydon, Samuel Coenen, Christopher C Butler, Pete Smith, Kerenza Hood, Beth Stuart, James Raftery, Natalie Thompson, Mandy Wan, Gilly O'Reilly, Kay Wang, Nick A Francis, Paul Little, Mike Thomas, Joseph Little, Joanne Euden, Samantha Richards-Hall, Michael Moore, Saul N. Faust, Robert C. Read, Mike Sharland, Taeko Becque, Theo J M Verheij, Alastair D Hay, and Kate Rowley
- Subjects
Male ,medicine.medical_specialty ,Population ,Placebo-controlled study ,Administration, Oral ,Placebo ,Double-Blind Method ,Internal medicine ,Lower respiratory tract infection ,medicine ,Humans ,education ,Child ,Respiratory Tract Infections ,education.field_of_study ,Respiratory tract infections ,Primary Health Care ,business.industry ,Hazard ratio ,Amoxicillin ,Infant ,General Medicine ,Articles ,medicine.disease ,Asthma ,Anti-Bacterial Agents ,Pneumonia ,Treatment Outcome ,England ,Child, Preschool ,Female ,Human medicine ,business ,medicine.drug - Abstract
Background: Antibiotic resistance is a global public health threat. Antibiotics are very commonly prescribed for children presenting with uncomplicated lower respiratory tract infections (LRTIs), but there is little evidence from randomised controlled trials of the effectiveness of antibiotics, both overall or among key clinical subgroups. In ARTIC PC, we assessed whether amoxicillin reduces the duration of moderately bad symptoms in children presenting with uncomplicated (non-pneumonic) LRTI in primary care, overall and in key clinical subgroups. Methods: ARTIC PC was a double-blind, randomised, placebo-controlled trial done at 56 general practices in England. Eligible children were those aged 6 months to 12 years presenting in primary care with acute uncomplicated LRTI judged to be infective in origin, where pneumonia was not suspected clinically, with symptoms for less than 21 days. Patients were randomly assigned in a 1:1 ratio to receive amoxicillin 50 mg/kg per day or placebo oral suspension, in three divided doses orally for 7 days. Patients and investigators were masked to treatment assignment. The primary outcome was the duration of symptoms rated moderately bad or worse (measured using a validated diary) for up to 28 days or until symptoms resolved. The primary outcome and safety were assessed in the intention-to-treat population. The trial is registered with the ISRCTN Registry (ISRCTN79914298). Findings: Between Nov 9, 2016, and March 17, 2020, 432 children (not including six who withdrew permission for use of their data after randomisation) were randomly assigned to the antibiotics group (n=221) or the placebo group (n=211). Complete data for symptom duration were available for 317 (73%) patients; missing data were imputed for the primary analysis. Median durations of moderately bad or worse symptoms were similar between the groups (5 days [IQR 4–11] in the antibiotics group vs 6 days [4–15] in the placebo group; hazard ratio [HR] 1·13 [95% CI 0·90–1·42]). No differences were seen for the primary outcome between the treatment groups in the five prespecified clinical subgroups (patients with chest signs, fever, physician rating of unwell, sputum or chest rattle, and short of breath). Estimates from complete-case analysis and a per-protocol analysis were similar to the imputed data analysis. Interpretation: Amoxicillin for uncomplicated chest infections in children is unlikely to be clinically effective either overall or for key subgroups in whom antibiotics are commonly prescribed. Unless pneumonia is suspected, clinicians should provide safety-netting advice but not prescribe antibiotics for most children presenting with chest infections.
- Published
- 2021
- Full Text
- View/download PDF
22. Controlled human infection with
- Author
-
Anastasia A, Theodosiou, Jay R, Laver, Adam P, Dale, David W, Cleary, Christine E, Jones, and Robert C, Read
- Subjects
Adult ,Microbiota ,Infant, Newborn ,Infant ,Mothers ,Pilot Projects ,Neisseria meningitidis ,Pregnancy ,Child, Preschool ,RNA, Ribosomal, 16S ,Humans ,Pharynx ,Female ,Child ,Neisseria lactamica - Abstract
Infant upper respiratory microbiota are derived partly from the maternal respiratory tract, and certain microbiota are associated with altered risk of infections and respiratory disease.20 healthy pregnant women will receive nasal inoculation withThis study has been approved by the London Central Research Ethics Committee (21/PR/0373). Findings will be published in peer-reviewed open-access journals as soon as possible.NCT04784845.
- Published
- 2022
23. Overcoming Waning Immunity in Pertussis Vaccines: Workshop of the National Institute of Allergy and Infectious Diseases
- Author
-
Kristina T. Lu, Robert C. Read, Nicola P. Klein, Eric T. Harvill, Pejman Rohani, Marcela F. Pasetti, Kathryn M. Edwards, Peter Sebo, Xin-Xing Gu, F. Heath Damron, Purnima Dubey, Kingston H. G. Mills, and Mariette Barbier
- Subjects
Bordetella pertussis ,medicine.medical_specialty ,Allergy ,Whooping Cough ,Immunology ,Acellular pertussis vaccines ,Article ,03 medical and health sciences ,Vaccines, Acellular ,0302 clinical medicine ,National Institute of Allergy and Infectious Diseases (U.S.) ,Immunity ,medicine ,Animals ,Humans ,Immunology and Allergy ,Waning immunity ,Intensive care medicine ,Pertussis Vaccine ,biology ,Transmission (medicine) ,business.industry ,Public health ,biology.organism_classification ,medicine.disease ,United States ,Acellular vaccines ,business ,030215 immunology - Abstract
Despite high vaccine coverage in many parts of the world, pertussis is resurging in a number of areas in which acellular vaccines are the primary vaccine administered to infants and young children. This is attributed in part to the suboptimal and short-lived immunity elicited by acellular pertussis vaccines and to their inability to prevent nasal colonization and transmission of the etiologic agent Bordetella pertussis. In response to this escalating public health concern, the National Institute of Allergy and Infectious Diseases held the workshop “Overcoming Waning Immunity in Pertussis Vaccines” in September 2019 to identify issues and possible solutions for the defects in immunity stimulated by acellular pertussis vaccines. Discussions covered aspects of the current problem, gaps in knowledge and possible paths forward. This review summarizes presentations and discussions of some of the key points that were raised by the workshop.
- Published
- 2020
- Full Text
- View/download PDF
24. Antibiotics for lower respiratory tract infection in children presenting in primary care (ARTIC-PC): the predictive value of molecular testing
- Author
-
Paul Little, Robert C. Read, Taeko Becque, Nick A. Francis, Alastair D. Hay, Beth Stuart, Gilly O'Reilly, Natalie Thompson, Kerenza Hood, Saul Faust, Kay Wang, Michael Moore, and Theo Verheij
- Subjects
Microbiology (medical) ,Infectious Diseases ,Bacteria ,Molecular Diagnostic Techniques ,Primary Health Care ,Viruses ,Amoxicillin ,Humans ,General Medicine ,Child ,Respiratory Tract Infections ,Anti-Bacterial Agents - Abstract
Objectives\udThis study aimed to assess whether the presence of bacteria or viruses in the upper airway of children presenting with uncomplicated lower respiratory tract infection (LRTI) predicts the benefit of antibiotics.\udMethods\udChildren between 6 months and 12 years presenting to UK general practices with an acute LRTI were randomized to receive amoxicillin 50 mg/kg/d for 7 days or placebo. Children not randomized (ineligible or clinician/parental choice) could participate in a parallel observational study. The primary outcome was the duration of symptoms rated moderately bad or worse. Throat swabs were taken and analyzed for the presence of bacteria and viruses by multiplex PCR.\udResults\udSwab results were available for most participants in the trial (306 of 432; 71%) and in the observational (182 of 326; 59%) studies. Bacterial pathogens potentially sensitive to amoxicillin (Haemophilus influenzae, Moraxella catarrhalis, Streptococcus pneumoniae) were detected among 51% of the trial placebo group and 49% of the trial antibiotic group. The median difference in the duration of symptoms rated moderately bad or worse between antibiotic and placebo was similar when potentially antibiotic-susceptible bacteria were present (median: –1 day; 99% CI, –12.3 to 10.3) or not present (median: –1 day; 99% CI, –4.5 to 2.5). Furthermore, bacterial genome copy number did not predict benefit. There were similar findings for all secondary outcomes and when including the data from the observational study.\udDiscussion\udThere was no clear evidence that antibiotics improved clinical outcomes conditional on the presence or concentration of bacteria or viruses in the upper airway. Before deploying microbiologic point-of-care tests for children with uncomplicated LRTI in primary care, rigorous validating trials are needed.
- Published
- 2022
25. Safety, tolerability and viral kinetics during SARS-CoV-2 human challenge
- Author
-
Ben Killingley, Alex Mann, Mariya Kalinova, Alison Boyers, Niluka Goonawardane, Jie Zhou, Kate Lindsell, Samanjit S. Hare, Jonathan Brown, Rebecca Frise, Emma Smith, Claire Hopkins, Nicolas Noulin, Brandon Londt, Tom Wilkinson, Stephen Harden, Helen McShane, Mark Baillet, Anthony Gilbert, Michael Jacobs, Christine Charman, Priya Mande, Jonathan S. Nguyen-Van-Tam, Malcolm G. Semple, Robert C. Read, Neil M. Ferguson, Peter J. Openshaw, Garth Rapeport, Wendy S. Barclay, Andrew P. Catchpole, and Christopher Chiu
- Abstract
To establish a novel SARS-CoV-2 human challenge model, 36 volunteers aged 18-29 years without evidence of previous infection or vaccination were inoculated with 10 TCID50 of a wild-type virus (SARS-CoV-2/human/GBR/484861/2020) intranasally. Two participants were excluded from per protocol analysis due to seroconversion between screening and inoculation. Eighteen (~53%) became infected, with viral load (VL) rising steeply and peaking at ~5 days post-inoculation. Virus was first detected in the throat but rose to significantly higher levels in the nose, peaking at ~8.87 log10 copies/ml (median, 95% CI [8.41,9.53). Viable virus was recoverable from the nose up to ~10 days post-inoculation, on average. There were no serious adverse events. Mild-to-moderate symptoms were reported by 16 (89%) infected individuals, beginning 2-4 days post-inoculation. Anosmia/dysosmia developed more gradually in 12 (67%) participants. No quantitative correlation was noted between VL and symptoms, with high VLs even in asymptomatic infection, followed by the development of serum spike-specific and neutralising antibodies. However, lateral flow results were strongly associated with viable virus and modelling showed that twice-weekly rapid tests could diagnose infection before 70-80% of viable virus had been generated. Thus, in this first SARS-CoV-2 human challenge study, no serious safety signals were detected and the detailed characteristics of early infection and their public health implications were shown. ClinicalTrials.gov identifier: NCT04865237.
- Published
- 2022
- Full Text
- View/download PDF
26. Safety and immunogenicity of seven COVID-19 vaccines as a third dose (booster) following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK (COV-BOOST): a blinded, multicentre, randomised, controlled, phase 2 trial
- Author
-
Phedra Marius, Lynne Grundy, Nabeela Nazir Ahmed, Margaret Irwin, Jeanette Thorpe, Hannah Robinson, Helen Thorp, Maria Moon, Sadaf Farooqi, Nick Andrews, Louise Haskell, Bea Choi, Helen Beckett, Sharon Davies-Dear, Victoria Cornelius, Tracey Dare, Sunder Chita, Stephen Singh, Chris Twelves, John Haughney, Patrick S. Moore, Maja dabagh, Xinxue Liu, S Bibi, Suzanne Wilkins, Mohammed Khan, Charlotte Trinham, Emily Brunt, Edwin Justice, Hanna Nguyen, Andrew Gowland, Andrew Riordan, Tanveer Bawa, Daniel Pan, Ceri Davies, Suahil Aslam, Chris A Rogers, Dileep Kumar, Yvanne Enever, Siobhan Roche, Karen Bisnauthsing, Hayley Tulloch, Andrew Ustianowski, Steve Hurdover, Ehsaan Qureshi, Akamino Egbo, Ingrid Seath, Jo Salkeld, Carla Ferreira Da Silva, Ray Sheridan, Samantha Keenan, Shama Hamal, Jo Piper, Kerry Godwin, Sara Bennett, Liliana Cifuentes, Nicholas Ronan, Nicki Lakeman, Lona Tudor Jones, Ian Bentley, Rachel White, Chloe McDonnell, Nina Parungao, Emma Plested, Kyra Holliday, Lisa Berry, Christine Minnis, Victoria Graham, Christopher J Edwards, Beth Giddins, Tara Watson, Suzie Colquhoun, Johanna Mouland, Marion K Campbell, Rostam Osanlou, Carlota Pereira Dias Alves, Simon Fowler, Becky Mansfield, Sally Batham, Orod Osanlou, Arpan Guha, Stephen Saich, Kush Naker, Marcin Bula, Igor Starinskij, Bassam Hallis, Sonia Baryschpolec, Shirley Todd, Agatha A. van der Klaauw, Claire Brown, Emma Snashall, Andrew Seaton, Helen Radford, John Hladkiwskyj, Rachael Drake-Brockman, Matilda lang, Linda Harndahl, Holly Burton, Tim Whitbred, Sue Charlton, Mushiya Mpelembue, Anna Stewart, Anil Shenoy, Zalina Rashid-Gardner, Joseph Newman, John Gavin, Mary Savage, Julie Evans, Aidan Lingwood, Lauren Allen, Parvinder K. Aley, Rebecca Lyon, Rachel Bousfield, Robert C. Read, Joanne Spencer, David Baxter, Anastasia de la Haye, James Calderwood, Emily Chiplin, Evgenia Kourampa, Helen Gutteridge, Jade Gouriet, Trishna Champaneri, Javier Magan, Luke Vamplew, Abigail Oliver, Sally Reeder, Sunil Sharma, Nicola Turner, Yukari Sakagami, Mikayala King, Steve Thomas, Chanice Knight, Samantha Broadhead, Erica Peters, Dennyl Vail, Marta Merida-Morillas, Emily Locke, Krishna Chatterjee, Debbie Suggitt, Sara Fraser, Mihaela Pacurar, Kerry Hughes, Jessica Hailstone, Eleni Ladikou, Leah Richmond, Wythehi Ambihapathy, Kari Nightingale, Chris Cooper, Victoria Wenn, Kimberley Driver, Rachel Hughes, Filipa Dos Santos, Michael Singh, Ben Gardside, Donna Wixted, Jessica Lewis-Taylor, Jason Domingo, Scott Elliott, Wiesia Woodyatt, Jonathan Kwok, Subarna Roy, Amisha Desai, Iryna Boubriak, Helen Haydock, Arabella Stuart, Amy Ross-Russell, Rossana Romani, Lauren Fox, Gillian McMillan, Angela M. Minassian, Ann Sturdy, R. A. James, Valerie Renals, Stephanie Leung, Lillian Goncalves cordeiro, Fran Westwell, Robert Shaw, Anna L. Goodman, Katrina Cathie, Ryan Stephen Elliott, Adrian Palfreeman, Phillip Brown, Kim En Lee, Farida khan, Suzanne Tasker, Anna Hardy, Elisa Nanino, Donald van Welsenes, Adam Farrier, Antonette Andrews, Jacqueline Brandon, Alicja Kownacka, Jennifer Murira, Kate Dodd, Emily Horsfall, Chantelle Moorbey, Alison Hogan, Lynda Wagstaff, Gita Patel, Rebecca Cutts, Matthew D. Snape, Karen Regan, Beverley Longhurst, Saul N. Faust, Vincenzo Libri, Andrea Mazzella, Michael Stackpoole, Carool Osuji, Jonathan Baker, Teona Serafimova, Tumena Corrah, Sophie E. Moore, Sarah Warren, Christopher Herbert, Laura Presland, Daniel R. Owens, Colin Hale, Beth Jackson, Fran Hall, Debbie Branney, Martha Nabunjo, Mehmood Mughal, Laura Longshaw, Holly Baker, Elizabeth A. Clutterbuck, Eloise Summerton, Rowena Weighell, Fiona Makia, Alexander Hicks, Leila Janani, Matthew Stokes, Amanda Buadi, E. Thomson, Jennifer Gibney, Jane Hall, Tricia Coughlan, Bridget Tandy, Kelly Littlewood, Christopher A Green, Mary Ramsay, Lorinda Pickup, Karren Buttigieg, Gavin Babbage, Todd Rawlins, Simon Tunstall, Dominique Barker, Martin J. Llewelyn, James Cullinane, Judith Bell, Elizabeth Gordon, Andrew L. Freedman, Martin Wiselka, Mohammed Kamal, Sarah Whittley, Natalie Baker, Jorden Frankham, Malathi Munusamy, Karen Underwood, Dinesh Saralaya, Olivia Chalwin, Tommy Rampling, Rachael Phillips, Sarah Garrahy, Yee Ting Nicole Yim, Charlotte Sabine, Haniah Habash-Bailey, Ashley Whittington, Benjamin Welham, Patrick Kinch, Avril Bonnaud, Jonathan Macdonald, NinaSimone Hopkins, Kim Storton, Stephen Hughes, Enya Cooney, Alasdair Munro, Christine Cole, John Paul Seenan, Kim Appleby, Laurence John, David J. Smith, Lara Barcella, Imam Shaik, Kate Ellis, Olumide Adebambo, Jane Stockport, Gertraud Morshead, Paminder Lall, Stephen E. Cox, Daniel Hansen, Jonathan Perkins, Yama F Mujadidi, Thomas Honey, Alan Magee, Jonathan S. Nguyen-Van-Tam, Mwila Kasanyinga, Marivic Ricamara, Jaimie Wilson-Goldsmith, Alastair McGregor, Djamila Shamtally, Helena Baker, Tom Eadsforth, Dee Mullan, Karishma Gokani, Kirsty Adams, and Dominic Galvin
- Subjects
Adult ,Male ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Population ,Immunization, Secondary ,Department of Error ,Group A ,Group B ,COV-BOOST study group ,Medicine, General & Internal ,Immunogenicity, Vaccine ,General & Internal Medicine ,ChAdOx1 nCoV-19 ,Internal medicine ,Safety, immunogenicity, COVID-19, vaccines, booster ,Humans ,Medicine ,Adverse effect ,education ,Pandemics ,BNT162 Vaccine ,11 Medical and Health Sciences ,Aged ,Aged, 80 and over ,education.field_of_study ,Science & Technology ,Booster (rocketry) ,Reactogenicity ,SARS-CoV-2 ,business.industry ,Immunogenicity ,COVID-19 ,General Medicine ,Articles ,Middle Aged ,United Kingdom ,Female ,Patient Safety ,business ,Life Sciences & Biomedicine - Abstract
Background: \ud Few data exist on the comparative safety and immunogenicity of different COVID-19 vaccines given as a third (booster) dose. To generate data to optimise selection of booster vaccines, we investigated the reactogenicity and immunogenicity of seven different COVID-19 vaccines as a third dose after two doses of ChAdOx1 nCov-19 (Oxford–AstraZeneca; hereafter referred to as ChAd) or BNT162b2 (Pfizer–BioNtech, hearafter referred to as BNT).\ud \ud Methods: \ud COV-BOOST is a multicentre, randomised, controlled, phase 2 trial of third dose booster vaccination against COVID-19. Participants were aged older than 30 years, and were at least 70 days post two doses of ChAd or at least 84 days post two doses of BNT primary COVID-19 immunisation course, with no history of laboratory-confirmed SARS-CoV-2 infection. 18 sites were split into three groups (A, B, and C). Within each site group (A, B, or C), participants were randomly assigned to an experimental vaccine or control. Group A received NVX-CoV2373 (Novavax; hereafter referred to as NVX), a half dose of NVX, ChAd, or quadrivalent meningococcal conjugate vaccine (MenACWY) control (1:1:1:1). Group B received BNT, VLA2001 (Valneva; hereafter referred to as VLA), a half dose of VLA, Ad26.COV2.S (Janssen; hereafter referred to as Ad26) or MenACWY (1:1:1:1:1). Group C received mRNA1273 (Moderna; hereafter referred to as m1273), CVnCov (CureVac; hereafter referred to as CVn), a half dose of BNT, or MenACWY (1:1:1:1). Participants and all investigatory staff were blinded to treatment allocation. Coprimary outcomes were safety and reactogenicity and immunogenicity of anti-spike IgG measured by ELISA. The primary analysis for immunogenicity was on a modified intention-to-treat basis; safety and reactogenicity were assessed in the intention-to-treat population. Secondary outcomes included assessment of viral neutralisation and cellular responses. This trial is registered with ISRCTN, number 73765130.\ud \ud Findings: \ud Between June 1 and June 30, 2021, 3498 people were screened. 2878 participants met eligibility criteria and received COVID-19 vaccine or control. The median ages of ChAd/ChAd-primed participants were 53 years (IQR 44–61) in the younger age group and 76 years (73–78) in the older age group. In the BNT/BNT-primed participants, the median ages were 51 years (41–59) in the younger age group and 78 years (75–82) in the older age group. In the ChAd/ChAD-primed group, 676 (46·7%) participants were female and 1380 (95·4%) were White, and in the BNT/BNT-primed group 770 (53·6%) participants were female and 1321 (91·9%) were White. Three vaccines showed overall increased reactogenicity: m1273 after ChAd/ChAd or BNT/BNT; and ChAd and Ad26 after BNT/BNT. For ChAd/ChAd-primed individuals, spike IgG geometric mean ratios (GMRs) between study vaccines and controls ranged from 1·8 (99% CI 1·5–2·3) in the half VLA group to 32·3 (24·8–42·0) in the m1273 group. GMRs for wild-type cellular responses compared with controls ranged from 1·1 (95% CI 0·7–1·6) for ChAd to 3·6 (2·4–5·5) for m1273. For BNT/BNT-primed individuals, spike IgG GMRs ranged from 1·3 (99% CI 1·0–1·5) in the half VLA group to 11·5 (9·4–14·1) in the m1273 group. GMRs for wild-type cellular responses compared with controls ranged from 1·0 (95% CI 0·7–1·6) for half VLA to 4·7 (3·1–7·1) for m1273. The results were similar between those aged 30–69 years and those aged 70 years and older. Fatigue and pain were the most common solicited local and systemic adverse events, experienced more in people aged 30–69 years than those aged 70 years or older. Serious adverse events were uncommon, similar in active vaccine and control groups. In total, there were 24 serious adverse events: five in the control group (two in control group A, three in control group B, and zero in control group C), two in Ad26, five in VLA, one in VLA-half, one in BNT, two in BNT-half, two in ChAd, one in CVn, two in NVX, two in NVX-half, and one in m1273.\ud \ud Interpretation: \ud All study vaccines boosted antibody and neutralising responses after ChAd/ChAd initial course and all except one after BNT/BNT, with no safety concerns. Substantial differences in humoral and cellular responses, and vaccine availability will influence policy choices for booster vaccination.\ud \ud Funding: \ud UK Vaccine Taskforce and National Institute for Health Research.
- Published
- 2021
27. Safety, tolerability and viral kinetics during SARS-CoV-2 human challenge in young adults
- Author
-
Ben Killingley, Alex J. Mann, Mariya Kalinova, Alison Boyers, Niluka Goonawardane, Jie Zhou, Kate Lindsell, Samanjit S. Hare, Jonathan Brown, Rebecca Frise, Emma Smith, Claire Hopkins, Nicolas Noulin, Brandon Löndt, Tom Wilkinson, Stephen Harden, Helen McShane, Mark Baillet, Anthony Gilbert, Michael Jacobs, Christine Charman, Priya Mande, Jonathan S. Nguyen-Van-Tam, Malcolm G. Semple, Robert C. Read, Neil M. Ferguson, Peter J. Openshaw, Garth Rapeport, Wendy S. Barclay, Andrew P. Catchpole, Christopher Chiu, BEIS - Department for Business, Energy and Industrial Strategy, and Wellcome Trust
- Subjects
Biochemistry & Molecular Biology ,Science & Technology ,SARS-CoV-2 ,Immunology ,COVID-19 ,Cell Biology ,General Medicine ,Research & Experimental Medicine ,Viral Load ,Antibodies, Viral ,DISEASE ,General Biochemistry, Genetics and Molecular Biology ,MODEL ,Kinetics ,Young Adult ,Treatment Outcome ,Medicine, Research & Experimental ,INFLUENZA ,INFECTION ,Humans ,LOAD ,Life Sciences & Biomedicine ,11 Medical and Health Sciences - Abstract
Since its emergence in 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused hundreds of millions of cases and continues to circulate globally. To establish a novel SARS-CoV-2 human challenge model that enables controlled investigation of pathogenesis, correlates of protection and efficacy testing of forthcoming interventions, 36 volunteers aged 18–29 years without evidence of previous infection or vaccination were inoculated with 10 TCID50of a wild-type virus (SARS-CoV-2/human/GBR/484861/2020) intranasally in an open-label, non-randomized study (ClinicalTrials.gov identifierNCT04865237; funder, UK Vaccine Taskforce). After inoculation, participants were housed in a high-containment quarantine unit, with 24-hour close medical monitoring and full access to higher-level clinical care. The study’s primary objective was to identify an inoculum dose that induced well-tolerated infection in more than 50% of participants, with secondary objectives to assess virus and symptom kinetics during infection. All pre-specified primary and secondary objectives were met. Two participants were excluded from the per-protocol analysis owing to seroconversion between screening and inoculation, identified post hoc. Eighteen (~53%) participants became infected, with viral load (VL) rising steeply and peaking at ~5 days after inoculation. Virus was first detected in the throat but rose to significantly higher levels in the nose, peaking at ~8.87 log10copies per milliliter (median, 95% confidence interval (8.41, 9.53)). Viable virus was recoverable from the nose up to ~10 days after inoculation, on average. There were no serious adverse events. Mild-to-moderate symptoms were reported by 16 (89%) infected participants, beginning 2–4 days after inoculation, whereas two (11%) participants remained asymptomatic (no reportable symptoms). Anosmia or dysosmia developed more slowly in 15 (83%) participants. No quantitative correlation was noted between VL and symptoms, with high VLs present even in asymptomatic infection. All infected individuals developed serum spike-specific IgG and neutralizing antibodies. Results from lateral flow tests were strongly associated with viable virus, and modeling showed that twice-weekly rapid antigen tests could diagnose infection before 70–80% of viable virus had been generated. Thus, with detailed characterization and safety analysis of this first SARS-CoV-2 human challenge study in young adults, viral kinetics over the course of primary infection with SARS-CoV-2 were established, with implications for public health recommendations and strategies to affect SARS-CoV-2 transmission. Future studies will identify the immune factors associated with protection in those participants who did not develop infection or symptoms and define the effect of prior immunity and viral variation on clinical outcome.
- Published
- 2021
28. A recombinant commensal bacteria elicits heterologous antigen-specific immune responses during pharyngeal carriage
- Author
-
Muhammad Ahmed, Holly E. Humphries, Jonathan M. Guy, Martin C. J. Maiden, Adam P. Dale, David W. Cleary, Hans de Graaf, Andrew Gorringe, Zoe C. Pounce, Carl N. Webb, Lauren Allen, Saul N. Faust, Konstantinos Belogiannis, Robert C. Read, Alison R Hill, Graham Berreen, Jay R. Laver, Eleanor F. Roche, Muktar Ibrahim, Anish K. Pandey, and Diane Gbesemete
- Subjects
0301 basic medicine ,Adult ,Heterologous vaccine ,biology ,030106 microbiology ,Meningococcal Vaccines ,General Medicine ,biology.organism_classification ,Antibodies, Bacterial ,Immunoglobulin G ,Microbiology ,Bacterial adhesin ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Antigen ,Antigens, Heterophile ,Neisseria lactamica ,biology.protein ,Humans ,Neisseria ,Seroconversion ,Immunologic Memory ,Ecosystem - Abstract
The human nasopharynx contains a stable microbial ecosystem of commensal and potentially pathogenic bacteria, which can elicit protective primary and secondary immune responses. Experimental intranasal infection of human adults with the commensal Neisseria lactamica produced safe, sustained pharyngeal colonization. This has potential utility as a vehicle for sustained release of antigen to the human mucosa, but commensals in general are thought to be immunologically tolerated. Here, we show that engineered N. lactamica, chromosomally transformed to express a heterologous vaccine antigen, safely induces systemic, antigen-specific immune responses during carriage in humans. When the N. lactamica expressing the meningococcal antigen Neisseria Adhesin A (NadA) was inoculated intranasally into human volunteers, all colonized participants carried the bacteria asymptomatically for at least 28 days, with most (86%) still carrying the bacteria at 90 days. Compared to an otherwise isogenic but phenotypically wild-type strain, colonization with NadA-expressing N. lactamica generated NadA-specific immunoglobulin G (IgG)- and IgA-secreting plasma cells within 14 days of colonization and NadA-specific IgG memory B cells within 28 days of colonization. NadA-specific IgG memory B cells were detected in peripheral blood of colonized participants for at least 90 days. Over the same period, there was seroconversion against NadA and generation of serum bactericidal antibody activity against a NadA-expressing meningococcus. The controlled infection was safe, and there was no transmission to adult bedroom sharers during the 90-day period. Genetically modified N. lactamica could therefore be used to generate beneficial immune responses to heterologous antigens during sustained pharyngeal carriage.
- Published
- 2021
29. Safety and immunogenicity of heterologous versus homologous prime-boost schedules with an adenoviral vectored and mRNA COVID-19 vaccine (Com-COV): a single-blind, randomised, non-inferiority trial
- Author
-
Sophie Varkonyi-Clifford, Sue Charlton, Anna England, Matthew D. Snape, Parvinder K. Aley, Elizabeth A. Clutterbuck, Pauline Ambrose, Eva P. Galiza, Adam Finn, Laura L. Walker, Emma Plested, Ehsaan Qureshi, Chanice Knight, David P. J. Turner, Lisa Hitchins, Mujtaba Ghulam Farooq, Christopher A Green, Melanie Greenland, J. Claire Cameron, Stephanie Leung, Karen Buttigieg, Jonathan S. Nguyen-Van-Tam, Cecilia Hultin, Florentina D Penciu, Jazz Bartholomew, Yama F Mujadidi, Nisha Singh, Sarah Horswill, Tommy Rampling, Kush Naker, Rabiullah Noristani, Xinxue Liu, Marivic Ricamara, Tawassal Riaz, Phill Brown, Rajeka Lazarus, Imam Shaik, Johanna K Kellett Wright, Kerry Godwin, Kelly Davies, Robert Shaw, Paminder Lall, Neil J. Oldfield, Farah Shiham, Sue Belton, Karishma Gokani, Carla Solórzano, Mary Ramsay, Kirsty Adams, Ross Fothergill, Hannah Robinson, Claudio Di Maso, Rachel Williams, Lauren Allen, Paul T. Heath, Sarah Whittley, Stephen Saich, E Howe, Bassam Hallis, Alberto San Francisco Ramos, Simon Royal, Alasdair Munro, Tanya Dinesh, Vincenzo Libri, Helen Hill, Arabella Stuart, Samuel Provstgaard-Morys, Xin L Yao, Andrea M. Collins, Rachel White, Paul Turner, Teresa Lambe, Nick Andrews, Iason Vichos, Daniela M. Ferreira, Natalie Baker, Nicole Y Yee Ting, Claire Brown, M N Ramasamy, Sarah Warren, Amisha Desai, Fei Long, Laura Presland, Hannah Sainsbury, Daniel Hammersley, Saul N. Faust, Robert C. Read, Nicola Turner, Group, Com-COV Study, and Department of Health
- Subjects
Male ,COVID-19/prevention & control ,Antibodies, Viral ,law.invention ,Antibodies, Viral/blood ,Immunogenicity, Vaccine ,Randomized controlled trial ,law ,qv_771 ,wc_505 ,Single-Blind Method ,11 Medical and Health Sciences ,COVID-19 Vaccines/administration & dosage ,education.field_of_study ,Immunogenicity ,General Medicine ,Middle Aged ,Intention to Treat Analysis ,Vaccination ,Spike Glycoprotein, Coronavirus ,Female ,Life Sciences & Biomedicine ,medicine.medical_specialty ,COVID-19 Vaccines ,Population ,REACTOGENICITY ,Equivalence Trials as Topic ,qw_806 ,Medicine, General & Internal ,General & Internal Medicine ,Internal medicine ,ChAdOx1 nCoV-19 ,parasitic diseases ,medicine ,Humans ,education ,BNT162 Vaccine ,Immunization Schedule ,Aged ,Science & Technology ,Intention-to-treat analysis ,Reactogenicity ,business.industry ,Com-COV Study Group ,Comment ,qu_58 ,COVID-19 ,EFFICACY ,Clinical trial ,Regimen ,Immunoglobulin G/blood ,Immunoglobulin G ,Spike Glycoprotein, Coronavirus/immunology ,business - Abstract
Background Use of heterologous prime-boost COVID-19 vaccine schedules could facilitate mass COVID-19 immunisation. However, we have previously reported that heterologous schedules incorporating an adenoviral vectored vaccine (ChAdOx1 nCoV-19, AstraZeneca; hereafter referred to as ChAd) and an mRNA vaccine (BNT162b2, Pfizer–BioNTech; hereafter referred to as BNT) at a 4-week interval are more reactogenic than homologous schedules. Here, we report the safety and immunogenicity of heterologous schedules with the ChAd and BNT vaccines. Methods Com-COV is a participant-blinded, randomised, non-inferiority trial evaluating vaccine safety, reactogenicity, and immunogenicity. Adults aged 50 years and older with no or well controlled comorbidities and no previous SARS-CoV-2 infection by laboratory confirmation were eligible and were recruited at eight sites across the UK. The majority of eligible participants were enrolled into the general cohort (28-day or 84-day prime-boost intervals), who were randomly assigned (1:1:1:1:1:1:1:1) to receive ChAd/ChAd, ChAd/BNT, BNT/BNT, or BNT/ChAd, administered at either 28-day or 84-day prime-boost intervals. A small subset of eligible participants (n=100) were enrolled into an immunology cohort, who had additional blood tests to evaluate immune responses; these participants were randomly assigned (1:1:1:1) to the four schedules (28-day interval only). Participants were masked to the vaccine received but not to the prime-boost interval. The primary endpoint was the geometric mean ratio (GMR) of serum SARS-CoV-2 anti-spike IgG concentration (measured by ELISA) at 28 days after boost, when comparing ChAd/BNT with ChAd/ChAd, and BNT/ChAd with BNT/BNT. The heterologous schedules were considered non-inferior to the approved homologous schedules if the lower limit of the one-sided 97·5% CI of the GMR of these comparisons was greater than 0·63. The primary analysis was done in the per-protocol population, who were seronegative at baseline. Safety analyses were done among participants receiving at least one dose of a study vaccine. The trial is registered with ISRCTN, 69254139. Findings Between Feb 11 and Feb 26, 2021, 830 participants were enrolled and randomised, including 463 participants with a 28-day prime-boost interval, for whom results are reported here. The mean age of participants was 57·8 years (SD 4·7), with 212 (46%) female participants and 117 (25%) from ethnic minorities. At day 28 post boost, the geometric mean concentration of SARS-CoV-2 anti-spike IgG in ChAd/BNT recipients (12 906 ELU/mL) was non-inferior to that in ChAd/ChAd recipients (1392 ELU/mL), with a GMR of 9·2 (one-sided 97·5% CI 7·5 to ∞). In participants primed with BNT, we did not show non-inferiority of the heterologous schedule (BNT/ChAd, 7133 ELU/mL) against the homologous schedule (BNT/BNT, 14 080 ELU/mL), with a GMR of 0·51 (one-sided 97·5% CI 0·43 to ∞). Four serious adverse events occurred across all groups, none of which were considered to be related to immunisation. Interpretation Despite the BNT/ChAd regimen not meeting non-inferiority criteria, the SARS-CoV-2 anti-spike IgG concentrations of both heterologous schedules were higher than that of a licensed vaccine schedule (ChAd/ChAd) with proven efficacy against COVID-19 disease and hospitalisation. Along with the higher immunogenicity of ChAd/BNT compared with ChAD/ChAd, these data support flexibility in the use of heterologous prime-boost vaccination using ChAd and BNT COVID-19 vaccines. Funding UK Vaccine Task Force and National Institute for Health Research.
- Published
- 2021
- Full Text
- View/download PDF
30. Pharyngeal carriage of inoculated recombinant commensal bacteria generates antigen-specific immunological memory
- Author
-
Diane Gbesemete, Zoe C. Pounce, Konstantinos Belogiannis, Graham Berreen, Andrew Gorringe, Saul N. Faust, Martin C. J. Maiden, Jay R. Laver, Muktar Ibrahim, Eleanor F. Roche, Holly E. Humphries, Lauren Allen, Robert C. Read, David W. Cleary, Hans de Graaf, Alison R Hill, Adam P. Dale, Anish K. Pandey, and Carl N. Webb
- Subjects
0303 health sciences ,biology ,030306 microbiology ,Heterologous ,Meningococcal vaccine ,biology.organism_classification ,3. Good health ,Microbiology ,Bacterial adhesin ,03 medical and health sciences ,Immune system ,Antigen ,stomatognathic system ,Neisseria lactamica ,otorhinolaryngologic diseases ,Colonization ,Neisseria ,030304 developmental biology - Abstract
The human nasopharynx is colonized by commensal bacteria and pathobionts, which comprise a complex microbial ecosystem capable of generating primary and secondary immune responses. Experimental intranasal infection of human adults with the commensal Neisseria lactamica results in safe, sustained colonization. Herein is described a novel technology to chromosomally transform N. lactamica with heterologous antigen, for the purpose of safe delivery to the mucosal surface and the generation of an antigen-specific immune response. N. lactamica was transformed to express the meningococcal vaccine antigen Neisseria Adhesin A (NadA) and was inoculated intranasally into humans at a dose of 105 colony-forming units. NadA-expressing N. lactamica colonized these individuals and was carried asymptomatically for 3 months. Colonization with NadA-expressing N. lactamica generated NadA-specific IgG-secreting plasma cells within 14 days of colonization and both NadA-specific IgG and NadA-specific IgG memory B cells within 28 days of colonization. NadA-specific IgG memory B cells circulate in the bloodstream of colonized participants for at least 90 days. Genetically transformed N. lactamica has the potential to be a safe bacterial vehicle to generate beneficial immune responses to a wide range of heterologous antigens during sustained pharyngeal carriage.
- Published
- 2021
31. The infant pharyngeal microbiomes: origin, impact and manipulation
- Author
-
Christine E. Jones, Robert B Dorey, Anastasia A Theodosiou, and Robert C. Read
- Subjects
0301 basic medicine ,Microbiology (medical) ,Maternal Health ,Respiratory System ,Respiratory Tract Diseases ,030106 microbiology ,Environment ,Infections ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,Flora (microbiology) ,Humans ,Medicine ,030212 general & internal medicine ,Microbiome ,Clinical phenotype ,Bacteria ,Milk, Human ,Whole Genome Sequencing ,business.industry ,Microbiota ,Infant, Newborn ,Infant ,Asthma ,Anti-Bacterial Agents ,Temporal diversity ,Infectious Diseases ,Pharynx ,Taxonomic resolution ,business - Abstract
Purpose of review: there has been an exponential increase in research intoinfant microbiome evolution, and it appears that pharyngeal microbiota are associated with clinical phenotypes(e.g. infection and asthma). While broad consensus views are emerging, significant challenges and uncertainties remain.Recent findings: infant pharyngeal microbiome research is limited by low biomass, high temporal diversity, and lack of agreed standards for sampling, DNA sequencing, and taxonomic reporting. Analysis of amplicon sequence variants and improvedcost and availability of whole genome sequencing are promising options for improving taxonomic resolution of such studies. Infant respiratory microbiomes arise, at least in part, from maternal flora (e.g. the respiratory tract and breastmilk), and are associated with environmental and clinical factors (e.g.mode of feeding and delivery, siblings, daycare attendance, birth season, and antibiotic usage). Interventional research to modify the infant pharyngeal microbiota hasrecentlybeen reported, using dietary supplements. Summary: further work is needed to improve characterisation ofthe infant pharyngeal microbiomes, including routes of bacterial acquisition, role of environmental factors, and associations with disease phenotypes. Methodological standards are desirable to facilitate more reproducible, comparable research. Improved understanding may enable manipulation of infant pharyngeal microbiota to improve clinical outcomes.
- Published
- 2020
32. G602(P) Is human challenge an acceptable methodology in pregnancy: an interview study
- Author
-
Robert B Dorey, Anastasia A Theodosiou, TF Vandrevala, Christine E. Jones, and Robert C. Read
- Subjects
Pregnancy ,education.field_of_study ,medicine.medical_specialty ,business.industry ,Population ,medicine.disease ,Risk perception ,Neonatal infection ,Qualitative analysis ,Family medicine ,medicine ,Interview study ,education ,business - Abstract
Introduction Human challenge studies are becoming increasingly common as a method for investigating vaccines and the relationships between commensal microbes and humans. To date, these studies have been conducted exclusively in healthy adult populations. There is a significant burden from infectious diseases in the pregnant and neonatal population. Human challenge studies in pregnancy may help us elucidate the mechanisms underpinning risk from microbial transfer from mother to infant and help with vaccine development. However, it is unclear whether these studies would be acceptable to pregnant women. Therefore, we undertook an interview study to investigate whether human challenge studies would be acceptable to a pregnant population. Methods We recruited healthy pregnant women from a maternity hospital until data saturation was reached. We invited them to take part in an interview study that explored perception of risk and acceptability of a hypothetical human challenge study in pregnancy. We conducted qualitative analysis of interview transcripts to identify themes and patterns present. Results A significant proportion of the participants interviewed found a human challenge study in pregnancy to be acceptable. They reported that their participation in such a study could be encouraged by detailed explanations of risk and how study design would mitigate these. They reported that a degree of uncertainty would not dissuade them from participating. We found that participants were polarised in their opinions of this hypothetical study and early in the interview formed opinions about acceptability. Finally, once these opinions were formed reassurance did not change participants initial impressions of acceptability. Conclusion Conducting a human challenge study in pregnancy would be acceptable to some pregnant women and may offer a means to investigate the specific environment that surrounds commensalisation of neonates. Furthermore, this may offer new avenues to explore vaccine development for the prevention of neonatal infection. While participants would want the maximum amount of information available before considering whether to participate, initial aversion to this methodology it is unlikely to be changed.
- Published
- 2020
- Full Text
- View/download PDF
33. Exploring the acceptability of controlled human infection with SARSCoV2—a public consultation
- Author
-
Diane Gbesemete, H. de Graaf, Wendy Lawrence, Daniella Watson, Robert C. Read, and Mary Barker
- Subjects
0301 basic medicine ,Open science ,medicine.medical_specialty ,Population ,SARSCoV2 ,lcsh:Medicine ,Public opinion ,Public interest ,03 medical and health sciences ,0302 clinical medicine ,Controlled human infection model ,SAFER ,Correspondence ,medicine ,030212 general & internal medicine ,Social isolation ,education ,education.field_of_study ,business.industry ,Public consultation ,lcsh:R ,COVID-19 ,General Medicine ,Focus group ,030104 developmental biology ,Family medicine ,medicine.symptom ,business - Abstract
Rapid development of an effective vaccine for SARSCoV2 is a global priority. A controlled human infection model (CHIM) would accelerate the efficacy assessment of candidate vaccines. This strategy would require deliberate exposure of volunteers to SARSCoV2 with no currently available treatment and a small but definite risk of serious illness or death. This raises complex questions about the social and ethical acceptability of risk to individuals, given the potential benefit to the wider population, and as such, a study cannot be done without public involvement. We conducted a structured public consultation with 57 individuals aged 20–40 years to understand public attitudes to a CHIM, and pre-requisites for enrolment. The overall response to this strategy was positive, and many would volunteer altruistically. Carefully controlled infection is viewed as safer than natural exposure to wild virus. The prolonged social isolation required for the proposed CHIM is considered an obstacle but not insurmountable, with reasonable compensation and supportive care. Given the significant level of public interest, a CHIM should be done as open science with regular, controlled dissemination of information into the public domain. Importantly, there was a strong view that the final decision whether to conduct a CHIM should be in the hands of qualified and experienced clinician-scientists and the authorities.
- Published
- 2020
34. Reducing risks from coronavirus transmission in the home-the role of viral load
- Author
-
Richard Amlôt, Jennifer Bostock, Paul Little, Cathy Rice, Tim Chadborn, Robert C. Read, and Lucy Yardley
- Subjects
Betacoronavirus/genetics ,Coronavirus disease 2019 (COVID-19) ,Infection Control/methods ,media_common.quotation_subject ,Health Behavior ,Pneumonia, Viral ,medicine.disease_cause ,law.invention ,Betacoronavirus ,Promotion (rank) ,law ,Risk Factors ,Environmental health ,Pandemic ,Infection control ,Medicine ,Animals ,Humans ,Pneumonia, Viral/transmission ,Pandemics ,media_common ,Coronavirus ,Family Health ,Infection Control ,Coronavirus Infections/transmission ,business.industry ,SARS-CoV-2 ,COVID-19 ,Covid19 ,General Medicine ,Viral Load ,Home Care Services ,Transmission (mechanics) ,Caregivers ,Models, Animal ,Physical and Mental Health ,Health behavior ,business ,Coronavirus Infections ,Viral load - Abstract
Paul Little and colleagues call for better promotion of simple measures that can help reduce the spread and severity of infection among those living with people who have covid-19.Most people with covid-19 are cared for at home, increasing the likely exposure of household members. Although the evidence is limited, high infection rates among health workers have been attributed to more frequent contact with infected patients, and higher viral load —the size of the infecting dose of virus. This has led to demands for better personal protection equipment (PPE). Less attention, however, has been given to family members and others caring for people with covid-19 in the community. Providing them with the same level of PPE as in hospitals is not practicable, but promotion of simple evidence based interventions may lower the risk of infection transmission and help reduce morbidity and demand on hospitals.
- Published
- 2020
- Full Text
- View/download PDF
35. Controlled Human Infection With Bordetella pertussis Induces Asymptomatic, Immunizing Colonization
- Author
-
Robert C. Read, Alison R Hill, Andrew T M Vaughan, Diane Gbesemete, Muktar Ibrahim, Guy A. M. Berbers, Hans de Graaf, Anne-Marie Buisman, Andrew Gorringe, Kent E. Kester, Dimitri A. Diavatopoulos, Saul N. Faust, and Andrew Preston
- Subjects
0301 basic medicine ,Microbiology (medical) ,Adult ,Bordetella pertussis ,Adolescent ,Whooping Cough ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Azithromycin ,Asymptomatic ,Immunoglobulin G ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Nasopharynx ,Medicine ,Humans ,Colonization ,030212 general & internal medicine ,Seroconversion ,Articles and Commentaries ,Pertussis Vaccine ,biology ,business.industry ,Transmission (medicine) ,Middle Aged ,biology.organism_classification ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Carriage ,Immunology ,biology.protein ,medicine.symptom ,business ,medicine.drug - Abstract
Background Bordetella pertussis is among the leading causes of vaccine-preventable deaths and morbidity globally. Human asymptomatic carriage as a reservoir for community transmission of infections might be a target of future vaccine strategies, but has not been demonstrated. Our objective was to demonstrate that asymptomatic nasopharyngeal carriage of Bordetella pertussis is inducible in humans and to define the microbiological and immunological features of presymptomatic infection. Methods Healthy subjects aged 18–45 years with an antipertussis toxin immunoglobin G (IgG) concentration of Results There were 34 participants challenged, in groups of 4 or 5. The dose was gradually escalated from 103 colony-forming units (0% colonized) to 105 colony-forming units (80% colonized). Minor symptoms were reported in a minority of participants. Azithromycin eradicated colonization in 48 hours in 88% of colonized individuals. Antipertussis toxin IgG seroconversion occurred in 9 out of 19 colonized participants and in none of the participants who were not colonized. Nasal wash was a more sensitive method to detect colonization than pernasal swabs. No shedding of Bordetella pertussis was detected in systematically collected environmental samples. Conclusions Bordetella pertussis colonization can be deliberately induced and leads to a systemic immune response without causing pertussis symptoms. Clinical Trials Registration NCT03751514.
- Published
- 2020
36. Public attitudes to a human challenge study with SARS-CoV-2: a mixed-methods study
- Author
-
Caroline Barker, Katharine Collet, Diane Gbesemete, Maria Piggin, Daniella Watson, Philippa Pristerà, Wendy Lawerence, Emma Smith, Michael Bahrami-Hessari, Halle Johnson, Katherine Baker, Ambar Qavi, Carmel McGrath, Christopher Chiu, Robert C. Read, and Helen Ward
- Subjects
viruses ,virus diseases ,Medicine (miscellaneous) ,biochemical phenomena, metabolism, and nutrition ,digestive system diseases ,General Biochemistry, Genetics and Molecular Biology - Abstract
Background: Human challenge studies involve the deliberate exposure of healthy volunteers to an infectious micro-organism in a highly controlled and monitored way. They are used to understand infectious diseases and have contributed to the development of vaccines. In early 2020, the UK started exploring the feasibility of establishing a human challenge study with SARS-CoV-2. Given the significant public interest and the complexity of the potential risks and benefits, it is vital that public views are considered in the design and approval of any such study and that investigators and ethics boards remain accountable to the public. Methods: Mixed methods study comprising online surveys conducted with 2,441 UK adults and in-depth virtual focus groups with 57 UK adults during October 2020 to explore the public’s attitudes to a human challenge study with SARS-CoV-2 taking place in the UK. Results: There was overall agreement across the surveys and focus groups that a human challenge study with SARS-CoV-2 should take place in the UK. Transparency of information, trust and the necessity to provide clear information on potential risks to study human challenge study participants were important. The perceived risks of taking part included the risk of developing long-term effects from COVID, impact on personal commitments and mental health implications of isolation. There were a number of practical realities to taking part that would influence a volunteer’s ability to participate (e.g. Wi-Fi, access to exercise, outside space and work, family and pet commitments). Conclusions: The results identified practical considerations for teams designing human challenge studies. Recommendations were grouped: 1) messaging to potential study participants, 2) review of the protocol and organisation of the study, and 3) more broadly, making the study more inclusive and relevant. This study highlights the value of public consultation in research, particularly in fields attracting public interest and scrutiny.
- Published
- 2022
- Full Text
- View/download PDF
37. Genomes of Escherichia coli bacteraemia isolates originating from urinary tract foci contain more virulence-associated genes than those from non-urinary foci and neutropaenic hosts
- Author
-
Jay R. Laver, Konstantinos Belogiannis, Richard Hesp, Adam P. Dale, Anish K. Pandey, Robert C. Read, and Clifford C. Shone
- Subjects
Adult ,Male ,0301 basic medicine ,Microbiology (medical) ,Neutropenia ,Virulence Factors ,030106 microbiology ,Bacteremia ,Disease ,Biology ,Antimicrobial resistance ,medicine.disease_cause ,Genome ,Article ,Virulence factor ,Microbiology ,Young Adult ,Neutropaenia ,03 medical and health sciences ,Antibiotic resistance ,Escherichia coli ,medicine ,Humans ,Prospective Studies ,Gene ,Escherichia coli Infections ,Phylogeny ,Aged ,Aged, 80 and over ,ExPEC ,Whole genome sequencing ,Virulence ,Whole Genome Sequencing ,Sequence Analysis, DNA ,Middle Aged ,United Kingdom ,3. Good health ,Infectious Diseases ,Urinary Tract Infections ,Monoclonal ,Bacteraemia ,Female ,Immunocompetence ,Genome, Bacterial - Abstract
OBJECTIVES: Escherichia coli is the leading cause of bacteraemia. In an era of emerging multi-drug-resistant strains, development of effective preventative strategies will be informed by knowledge of strain diversity associated with specific infective syndromes/patient groups. We hypothesised that the number of virulence factor (VF) genes amongst bacteraemia isolates from neutropaenic patients would be lower than isolates from immunocompetent patients.METHODS: Immunocompetent and neutropaenic adults with E. coli bacteraemia were recruited prospectively and the source of bacteraemia determined. VF gene profiles were established in silico following whole genome sequencing.RESULTS: Isolates from individual patients were monoclonal. Strains from immunocompetent patients with urinary tract infective foci (UTIF) harboured more VF genes (median number of VF genes 16, range 8-24) than isolates from both immunocompetent patients with non-UTIF (10, 2-22, p = 0.0058) and neutropaenic patients with unknown focus of infection (NPUFI) (8, 3-13, p < 0.0001). Number of VF genes (OR 1.21, 95% CIs 1.01-1.46, p = 0.039) and urinary catheter/recurrent urinary tract infection (OR 12.82, 95% CIs 1.24-132.65, p = 0.032) were independent predictors of bacteraemia secondary to UTIF vs. non-UTIF in immunocompetent patients. papA, papC, papE/F, papG, agn43, tia, iut, fyuA, kpsM and sat were significantly more prevalent amongst UTIF- vs non-UTIF-originating isolates amongst immunocompetent patients, while papC, papE/F, papG, agn43, tia, fyuA, hlyA, usp and clb were significantly more prevalent amongst UTIF- vs NPUFI-associated isolates.CONCLUSIONS: Bacteraemia-associated E. coli strains originating from UTIF have distinct VF gene profiles from strains associated with non-UTIF- and NPUFI. This diversity must be addressed in the design of future vaccines to ensure adequate coverage of strains responsible for site-specific disease.
- Published
- 2018
- Full Text
- View/download PDF
38. ERS syllabus for postgraduate training in respiratory infections: a guide for comprehensive training
- Author
-
Nathalie Tabin, Felix C. Ringshausen, George Dimopoulos, Robert C. Read, Stefano Aliberti, Gernot Rohde, Giovanni Sotgiu, Amy Farr, Giovanni Battista Migliori, Michael R. Loebinger, Francesco Blasi, Antoni Torres, Mark Woodhead, and James D. Chalmers
- Subjects
lcsh:RC705-779 ,Pulmonary and Respiratory Medicine ,Medical education ,business.industry ,05 social sciences ,Editorials ,lcsh:Diseases of the respiratory system ,Training (civil) ,Syllabus ,03 medical and health sciences ,0302 clinical medicine ,030228 respiratory system ,0502 economics and business ,Medicine ,Postgraduate training ,business ,050203 business & management - Abstract
Respiratory infections, including tuberculosis, represent one of the leading causes of morbidity and mortality across the world. They represent the deadliest communicable diseases causing 3.0 million deaths worldwide in 2016 [1]. The burden of respiratory infections can be unbearable for some health systems: they represent one of the most common reasons for doctor visits, regardless of age and sex [2]., ERS has developed a syllabus for postgraduate training in respiratory infections to guide programme designers http://ow.ly/xJ0R30m8CYB
- Published
- 2018
- Full Text
- View/download PDF
39. The nonpathogenic commensal Neisseria: friends and foes in infectious disease
- Author
-
Robert C. Read, Anastasia A Theodosiou, Christine E. Jones, and Robert B Dorey
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.medical_specialty ,Neisseriaceae Infections ,030106 microbiology ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,Immunocompromised Host ,0302 clinical medicine ,Molecular genetics ,Medicine ,Animals ,Humans ,In patient ,030212 general & internal medicine ,biology ,business.industry ,Neisseria meningitidis ,Biologic therapies ,biology.organism_classification ,Commensalism ,Disease Models, Animal ,Infectious Diseases ,Infectious disease (medical specialty) ,Neisseria lactamica ,Neisseria ,business - Abstract
Purpose of review Nonpathogenic commensal Neisseria are rarely considered in the clinical setting despite evidence that they can cause invasive opportunistic infections. In contrast, they may offer protection against pathogenic Neisseria, and such relationships are being actively explored in experimental studies. Recent findings Recent case reports are presented of invasive infection caused by nonpathogenic Neisseria in patients on novel biologic therapies. On the other hand, Neisseria lactamica, a nonpathogenic commensal, has been shown in human challenge studies to inhibit colonization by Neisseria meningitidis. Experimental mouse models have also explored the inhibitory effects of nonpathogenic Neisseria on Neisseria gonnhoreae infection. Cutting-edge advances in metagenomics and microbiomics are being used to understand the mechanisms underpinning these effects. Summary Clinicians should have increased awareness of nonpathogenic Neisseria. First, as new immunomodulating therapies become licenced, the interactions that maintain balance between commensals and their human hosts may be altered. Second, these bacteria are showing promise in their capacity to exclude pathogenic Neisseria species from their anatomical niches.
- Published
- 2019
40. Protocol for a controlled human infection with genetically modified Neisseria lactamica expressing the meningococcal vaccine antigen NadA: a potent new technique for experimental medicine
- Author
-
Robert C. Read, Muktar Ibrahim, Hans de Graaf, Andrew Gorringe, Diane Gbesemete, Saul N. Faust, Andrew T M Vaughan, and Jay R. Laver
- Subjects
Adult ,Biomedical Research ,Adolescent ,colonisation ,Meningococcal Vaccines ,Meningococcal vaccine ,Neisseria meningitidis ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,genetically modified organism ,Protocol ,Infection control ,Medicine ,Humans ,030212 general & internal medicine ,Prospective Studies ,Antigens ,Adhesins, Bacterial ,Neisseria lactamica ,030304 developmental biology ,0303 health sciences ,biology ,Transmission (medicine) ,business.industry ,Immunogenicity ,General Medicine ,Middle Aged ,biology.organism_classification ,3. Good health ,Genetically modified organism ,Carriage ,Infectious Diseases ,Research Design ,Immunology ,human challenge study ,Bacterial antigen ,Microorganisms, Genetically-Modified ,business - Abstract
IntroductionNeisseria lactamicais a commensal organism found in the human nasopharynx and is closely related to the pathogenN. meningitidis(meningococcus). Carriage ofN. lactamicais associated with reduced meningococcal carriage and disease. We summarise an ethically approved protocol for an experimental human challenge study using a genetically modified strain ofN. lactamicathat expresses the meningococcal antigen NadA. We aim to develop a model to study the role of specific bacterial antigens in nasopharyngeal carriage and immunity, to evaluate vaccines for their efficacy in preventing colonisation and to provide a proof of principle for the development of bacterial medicines.Methods and analysisHealthy adult volunteers aged 18–45 years will receive an intranasal inoculation of either the NadA containing strain ofN. lactamicaor a genetically modified, but wild-type equivalent control strain. These challenge volunteers will be admitted for 4.5 days observation following inoculation and will then be discharged with strict infection control rules. Bedroom contacts of the challenge volunteers will also be enrolled as contact volunteers. Safety, colonisation, shedding, transmission and immunogenicity will be assessed over 90 days after which carriage will be terminated with antibiotic eradication therapy.Ethics and disseminationThis study has been approved by the Department for Environment, Food and Rural Affairs and South Central Oxford A Research Ethics Committee (reference: 18/SC/0133). Findings will be published in peer-reviewed open-access journals as soon as possible.Trial registration numberNCT03630250; Pre-results.
- Published
- 2019
41. Erratum: Blume, C., et al. Modulation of Human Airway Barrier Functions during Burkholderia thailandensis and Francisella tularensis Infection Running Title: Airway Barrier Functions during Bacterial Infections. Pathogens 2016, 5, 53
- Author
-
Jonathan David, Rachel Bell, Emily J. Swindle, Cornelia Blume, Donna E. Davies, Robert C. Read, Graeme C. Clark, and Jay R. Laver
- Subjects
Microbiology (medical) ,Infectious Diseases ,Francisella tularensis Infection ,General Immunology and Microbiology ,Burkholderia thailandensis ,Immunology and Allergy ,Human airway ,Biology ,Airway ,biology.organism_classification ,Molecular Biology ,Microbiology - Abstract
There is an error in the title [...]
- Published
- 2020
- Full Text
- View/download PDF
42. A qPCR assay for Bordetella pertussis cells that enumerates both live and dead bacteria
- Author
-
H. de Graaf, Andrew Preston, Robert C. Read, Iain MacArthur, Stacy Ramkissoon, and Muktar Ibrahim
- Subjects
Bacterial Diseases ,Bordetella pertussis ,Bordetella ,THP-1 Cells ,Artificial Gene Amplification and Extension ,Pathology and Laboratory Medicine ,Polymerase Chain Reaction ,law.invention ,0302 clinical medicine ,Cell Signaling ,law ,Medicine and Health Sciences ,030212 general & internal medicine ,Polymerase chain reaction ,0303 health sciences ,Vaccines ,Multidisciplinary ,biology ,C700 ,Bacterial Pathogens ,3. Good health ,Vaccination ,Infectious Diseases ,Eukaryotic Cells ,Molecular Diagnostic Techniques ,Immune correlates ,Medical Microbiology ,Medicine ,Pathogens ,Cellular Types ,Acellular vaccines ,Research Article ,Signal Transduction ,Signal Inhibition ,Infectious Disease Control ,Science ,Immunology ,Research and Analysis Methods ,Microbiology ,Sensitivity and Specificity ,03 medical and health sciences ,Pertussis ,030225 pediatrics ,medicine ,Humans ,Molecular Biology Techniques ,Microbial Pathogens ,Molecular Biology ,Whooping cough ,030304 developmental biology ,Bacteria ,030306 microbiology ,business.industry ,Organisms ,Immunity ,Biology and Life Sciences ,Cell Biology ,C400 ,biology.organism_classification ,medicine.disease ,Virology ,Quantitative measure ,Whole cell ,business - Abstract
Bordetella pertussis is the causative agent of whooping cough, commonly referred to as pertussis. Although the incidence of pertussis was reduced through vaccination, during the last thirty years it has returned to high levels in a number of countries. This resurgence has been linked to the switch from the use of whole-cell to acellular vaccines. Protection afforded by acellular vaccines appears to be short-lived compared to that afforded by whole cell vaccines. In order to inform future vaccine improvement by identifying immune correlates of protection, a human challenge model of B. pertussis colonisation has been developed. Accurate measurement of colonisation status in this model has required development of a qPCR-based assay to enumerate B. pertussis in samples that distinguishes between viable and dead bacteria. Here we report the development of this assay and its performance in the quantification of B. pertussis from human challenge model samples. This assay has future utility in diagnostic labs and in research where a quantitative measure of both B. pertussis number and viability is required.
- Published
- 2020
- Full Text
- View/download PDF
43. Microevolution of Neisseria lactamica during nasopharyngeal colonisation induced by controlled human infection
- Author
-
Robert C. Read, Adam P. Dale, Xavier Didelot, Alice M. Deasy, Jay R. Laver, Martin C. J. Maiden, Anish K. Pandey, Paul Morris, Andrew Gorringe, and David W. Cleary
- Subjects
0301 basic medicine ,Neisseriaceae Infections ,Science ,Hypothetical protein ,Colony Count, Microbial ,General Physics and Astronomy ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,03 medical and health sciences ,Mutation Rate ,stomatognathic system ,Nasopharynx ,otorhinolaryngologic diseases ,medicine ,Humans ,Allele ,lcsh:Science ,Gene ,Pathogen ,Neisseria lactamica ,Recombination, Genetic ,Multidisciplinary ,biology ,Neisseria meningitidis ,Microevolution ,General Chemistry ,biology.organism_classification ,3. Good health ,QR ,030104 developmental biology ,Genes, Bacterial ,Carrier State ,Mutation ,lcsh:Q ,Bacterial outer membrane - Abstract
Neisseria lactamica is a harmless coloniser of the infant respiratory tract, and has a mutually-excluding relationship with the pathogen Neisseria meningitidis. Here we report controlled human infection with genomically-defined N. lactamica and subsequent bacterial microevolution during 26 weeks of colonisation. We find that most mutations that occur during nasopharyngeal carriage are transient indels within repetitive tracts of putative phase-variable loci associated with host-microbe interactions (pgl and lgt) and iron acquisition (fetA promotor and hpuA). Recurrent polymorphisms occurred in genes associated with energy metabolism (nuoN, rssA) and the CRISPR-associated cas1. A gene encoding a large hypothetical protein was often mutated in 27% of the subjects. In volunteers who were naturally co-colonised with meningococci, recombination altered allelic identity in N. lactamica to resemble meningococcal alleles, including loci associated with metabolism, outer membrane proteins and immune response activators. Our results suggest that phase variable genes are often mutated during carriage-associated microevolution., Carriage of Neisseria lactamica, a harmless coloniser of the human respiratory tract, is inversely correlated with Neisseria meningitidis infection. Here, Pandey et al. provide insights into micro-evolutionary processes in N. lactamica during controlled infection of healthy volunteers.
- Published
- 2018
44. Malaria systems immunology: Plasmodium vivax induces tolerance during primary infection through dysregulation of neutrophils and dendritic cells
- Author
-
Andres F, Vallejo, Robert C, Read, Myriam, Arevalo-Herrera, Sócrates, Herrera, Tim, Elliott, and Marta E, Polak
- Subjects
Antigen Presentation ,Immunity, Cellular ,Vaccines ,Neutrophils ,Gene Expression Profiling ,Plasmodium falciparum ,Down-Regulation ,High-Throughput Nucleotide Sequencing ,Immunoregulation ,Dendritic Cells ,Lymphocyte Activation ,Article ,Malaria ,Controlled human malaria infection ,parasitic diseases ,Immune Tolerance ,Malaria, Vivax ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Malaria, Falciparum ,Plasmodium vivax ,Transcriptomics - Abstract
Highlights • In silico analysis suggests that primary infection with P. vivax induces potent immunosuppression mediated by dendritic cells. • Antigen presentation is attenuated in malaria-experienced vs malaria-naïve individuals • Malaria induce immunosuppressive effect manifested with strong induction of IDO1., Summary Objectives To dissect the transcriptional networks underpinning immune cells responses during primary Plasmodium vivax infection of healthy human adults. Methods We conducted network co-expression analysis of next-generation RNA sequencing data from whole blood from P. vivax and P. falciparum controlled human malaria infection (CHMI) of healthy naïve and malaria-exposed volunteers. Single cell transcription signatures were used to deconvolute the bulk RNA-Seq data into cell-specific signals. Results Initial exposure to P. vivax induced activation of innate immunity, including efficient antigen presentation and complement activation. However, this effect was accompanied by strong immunosuppression mediated by dendritic cells via the induction of Indoleamine 2,3-Dioxygenase 1(IDO1) and Lymphocyte Activation Gene 3 (LAG3). Additionally, P. vivax induced depletion of neutrophil populations associated with down regulation of 3G-protein coupled receptors, CRXCR1, CXCR2 and CSF3R. Accordingly, in malaria-exposed volunteers the inflammatory response was attenuated, with a decreased class II antigen presentation in dendritic cells. While the immunosuppressive signalling was maintained between plasmodium species, response to P. falciparum was significantly more immunogenic. Conclusions In silico analyses suggest that primary infection with P. vivax induces potent immunosuppression mediated by dendritic cells, conditioning subsequent anti-malarial immune responses. Targeting immune evasion mechanisms could be an effective alternative for improving vaccine efficacy.
- Published
- 2018
45. Summary of the international clinical guidelines for the management of hospital-acquired and ventilator-acquired pneumonia
- Author
-
Carlos M. Luna, Jean Chastre, Santiago Ewig, Marin H. Kollef, Gianluigi Li Bassi, Michael S. Niederman, Robert C. Read, J. Artur Paiva, Richard G. Wunderink, Patricia Fernandez-Vandellos, Jean-François Timsit, Tobias Welte, Håkan Hanberger, David Rigau, Ignacio Martin-Loeches, and Antoni Torres
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,business.industry ,lcsh:R ,MEDLINE ,Klinisk medicin ,lcsh:Medicine ,Pneumonia ventilator associated ,medicine.disease ,3. Good health ,respiratory tract diseases ,03 medical and health sciences ,Pneumonia ,Clinical Signpost ,0302 clinical medicine ,030228 respiratory system ,medicine ,030212 general & internal medicine ,Clinical Medicine ,Intensive care medicine ,business - Abstract
The most recent European guidelines and task force reports on hospital-acquired pneumonia (HAP) and ventilator-associated pneumonia (VAP) were published almost 10 years ago. Since then, further randomised clinical trials of HAP and VAP have been conducted and new information has become available. Studies of epidemiology, diagnosis, empirical treatment, response to treatment, new antibiotics or new forms of antibiotic administration, and disease prevention have changed old paradigms. In addition, important differences between approaches in Europe and the USA have become apparent. The European Respiratory Society (ERS) launched a project to develop new international guidelines for HAP and VAP. Other European societies, including the European Society of Intensive Care Medicine (ESICM) and the European Society of Clinical Microbiology and Infectious Diseases (ESCMID), were invited to participate and appointed their representatives. The Latin American Society of Thoracic Diseases (ALAT) was also invited to participate. This manuscript summarises the evidence and recommendations of these international guidelines on HAP and VAP., A summary of the evidence and recommendations made in the ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia http://ow.ly/S3zA30iZfLa
- Published
- 2018
46. Inability to sustain intraphagolysosomal killing of Staphylococcus aureus predisposes to bacterial persistence in macrophages
- Author
-
Simon A. Johnston, David H. Dockrell, Helen M. Marriott, Simon Stoneham, Robert C. Read, Simon J. Foster, Andrew A. Peden, Jamil Jubrail, Paul Morris, and Martin A. Bewley
- Subjects
0301 basic medicine ,Staphylococcus aureus ,Cell Survival ,Phagocytosis ,medicine.disease_cause ,Microbiology ,Phagolysosome ,immunology ,03 medical and health sciences ,Mice ,Virology ,Pneumonia, Staphylococcal ,medicine ,Animals ,Pathogen ,Staphylococci ,Microbial Viability ,biology ,Intracellular parasite ,Macrophages ,microbial‐cell interaction ,Original Articles ,biology.organism_classification ,Disease Models, Animal ,030104 developmental biology ,antimicrobial ,Original Article ,Bacteria ,Intracellular - Abstract
Summary Macrophages are critical effectors of the early innate response to bacteria in tissues. Phagocytosis and killing of bacteria are interrelated functions essential for bacterial clearance but the rate‐limiting step when macrophages are challenged with large numbers of the major medical pathogen Staphylococcus aureus is unknown. We show that macrophages have a finite capacity for intracellular killing and fail to match sustained phagocytosis with sustained microbial killing when exposed to large inocula of S. aureus (Newman, SH1000 and USA300 strains). S. aureus ingestion by macrophages is associated with a rapid decline in bacterial viability immediately after phagocytosis. However, not all bacteria are killed in the phagolysosome, and we demonstrate reduced acidification of the phagolysosome, associated with failure of phagolysosomal maturation and reduced activation of cathepsin D. This results in accumulation of viable intracellular bacteria in macrophages. We show macrophages fail to engage apoptosis‐associated bacterial killing. Ultittop mately macrophages with viable bacteria undergo cell lysis, and viable bacteria are released and can be internalized by other macrophages. We show that cycles of lysis and reuptake maintain a pool of viable intracellular bacteria over time when killing is overwhelmed and demonstrate intracellular persistence in alveolar macrophages in the lungs in a murine model.
- Published
- 2015
47. Investigating Bordetella pertussis colonisation and immunity: protocol for an inpatient controlled human infection model
- Author
-
Diane Gbesemete, Andrew Gorringe, Saul N. Faust, Dimitri A. Diavatopoulos, Kent E. Kester, Robert C. Read, and Hans de Graaf
- Subjects
0301 basic medicine ,Bordetella pertussis ,medicine.medical_specialty ,colonisation ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Dose-Response Relationship, Immunologic ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Vaccines, Acellular ,Clinical Protocols ,Immunity ,Internal medicine ,Protocol ,Medicine ,Humans ,whooping cough ,030212 general & internal medicine ,Whooping cough ,Pertussis Vaccine ,biology ,business.industry ,Transmission (medicine) ,Approved Protocol ,Vaccination ,General Medicine ,bordetella pertussis ,biology.organism_classification ,medicine.disease ,3. Good health ,Colonisation ,030104 developmental biology ,Infectious Diseases ,vaccine development ,Research Design ,Immunology ,human challenge study ,business ,Developed country - Abstract
IntroductionWe summarise an ethically approved protocol for the development of an experimental human challenge colonisation model. GloballyBordetella pertussisis one of the leading causes of vaccine-preventable death. Many countries have replaced whole cell vaccines with acellular vaccines over the last 20 years during which pertussis appears to be resurgent in a number of countries in the developed world that boast high immunisation coverage. The acellular vaccine provides relatively short-lived immunity and, in contrast to whole cell vaccines, may be less effective against colonisation and subsequent transmission. To improve vaccine strategies, a greater understanding of humanB. pertussiscolonisation is required. This article summarises a protocol and does not contain any results.Methods and analysisA controlled human colonisation model will be developed over two phases. In phase A, a low dose of the inoculum will be given intranasally to healthy participants. This dose will be escalated or de-escalated until colonisation is achieved in approximately 70% (95% CI 47% to 93%) of the exposed volunteers without causing disease. The colonisation period, shedding and exploratory immunology will be assessed during a 17-day inpatient stay and follow-up over 1 year. The dose of inoculum that achieves 70% colonisation will then be confirmed in phase B, comparing healthy participants exposed toB. pertussiswith a control group receiving a sham inoculum.Ethics and disseminationThis study has been approved by the ethical committee reference: 17/SC/0006, 24 February 2017. Findings will be published in peer-reviewed open access journals as soon as possible.
- Published
- 2017
48. Neisseria lactamica Y92-1009 complete genome sequence
- Author
-
David W. Cleary, Jay R. Laver, Andrew Gorringe, Robert C. Read, Martin C. J. Maiden, Xavier Didelot, Anish K. Pandey, and Medical Research Council (MRC)
- Subjects
0301 basic medicine ,GENES ,lcsh:QH426-470 ,030106 microbiology ,ASSEMBLIES ,Sequence assembly ,Nasopharyngeal microflora ,Commensal ,CHILDREN ,Meningococcal disease ,medicine.disease_cause ,Genome ,Microbiology ,DISEASE ,COLONIZATION ,03 medical and health sciences ,Short read sequencing ,MENINGITIDIS ,Genetics ,medicine ,Gene ,Whole genome sequencing ,Genetics & Heredity ,Science & Technology ,Genome assembly ,biology ,SMRT cell sequencing ,Bacteria ,IDENTIFICATION ,Neisseria meningitidis ,biology.organism_classification ,medicine.disease ,EVOLUTION ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,CARRIAGE ,Neisseria lactamica ,Neisseria ,Life Sciences & Biomedicine - Abstract
We present the high quality, complete genome assembly of Neisseria lactamica Y92–1009 used to manufacture an outer membrane vesicle (OMV)-based vaccine, and a member of the Neisseria genus. The strain is available on request from the Public Health England Meningococcal Reference Unit. This Gram negative, dipplococcoid bacterium is an organism of worldwide clinical interest because human nasopharyngeal carriage is related inversely to the incidence of meningococcal disease, caused by Neisseria meningitidis. The organism sequenced was isolated during a school carriage survey in Northern Ireland in 1992 and has been the subject of a variety of laboratory and clinical studies. Four SMRT cells on a RSII machine by Pacific Biosystems were used to produce a complete, closed genome assembly. Sequence data were obtained for a total of 30,180,391 bases from 2621 reads and assembled using the HGAP algorithm. The assembly was corrected using short reads obtained from an Illumina HiSeq 2000instrument. This resulted in a 2,146,723 bp assembly with approximately 460 fold mean coverage depth and a GC ratio of 52.3%.
- Published
- 2017
49. List of Contributors
- Author
-
Fredrick M. Abrahamian, Michael J. Aldape, Edelweiss Aldasoro, Upton D. Allen, Hythem Al-Sum, Milan J. Anadkat, Katherine Anders, Emmanouil Angelakis, Brian John Angus, Anastasia Antoniadou, Fabio Arena, Joop E. Arends, Jose R. Arribas, Andrew W. Artenstein, John C. Atherton, John N. Aucott, Tar-Ching Aw, Hilary M. Babcock, Robin Bailey, Thomas C. Bailey, Adam Z. Banks, David J. Barillo, Ernie-Paul Barrette, Martijn P. Bauer, Roger Bayston, C. Ben Beard, Justin Beardsley, Nick J. Beeching, Rodolfo E. Bégué, Guido Beldi, Constance A. Benson, Elie F. Berbari, Jean-Michel Berenger, Christoph Berger, Jose I. Bernardino, Jacques Bille, Alexander C. Billioux, Ari Bitnun, Iain Blair, Stéphane Blanche, Thomas P. Bleck, Chantal P. Bleeker-Rovers, Gijs Bleijenberg, Karen C. Bloch, Johannes Blum, Emily A. Blumberg, Robert A. Bonomo, Marc J.M. Bonten, Rafik Bourayou, Emilio Bouza, K. Ashley Brandt, Florence Bretelle, Sylvain Brisse, Warwick J. Britton, Itzhak Brook, Matthijs C. Brouwer, Sarah K. Browne, Amy E. Bryant, Silja Bühler, Eileen M. Bulger, R. Mark L. Buller, Leah A. Burke, Christian Burri, Marcus W. Butler, Thierry Calandra, David P. Calfee, Antonia Calvo-Cano, D. William Cameron, Joseph A. Carcillo, Gail Carson, Stephen T. Chambers, Remi N. Charrel, Vinh Chau Van Nguyen, Stéphane Chevaliez, Tom M. Chiller, Eirini Christaki, Kevin K. Chung, David B. Clifford, Nathan Clumeck, Jonathan Cohen, John Collinge, Christopher P. Conlon, Curdin Conrad, Fiona J. Cooke, Jennifer Rittenhouse Cope, G. Ralph Corey, John H. Cross, Burke A. Cunha, Cheston B. Cunha, Benoit D'Journo, George L. Daikos, Johannes M.A. Daniels, Robert N. Davidson, Nicholas P.J. Day, Kevin M. De Cock, Thushan I. de Silva, Henry J.C. de Vries, Stéphane de Wit, Julie Delaloye, David W. Denning, David T. Dennis, Shireesha Dhanireddy, Elodi J. Dielubanza, David J. Diemert, Mehmet Doganay, Tom Doherty, Christiane Dolecek, Arjen M. Dondorp, Abby Douglas, Michel Drancourt, Grégory Dubourg, Michael N. Dudley, Guillaume Durand, Benjamin J. Eckhardt, Androulla Efstratiou, Miquel B. Ekkelenkamp, Ambika Eranki, Hakan Erdem, Gerome V. Escota, Heather L. Evans, Alice Chijioke Eziefula, Florence Fenollar, Alan Fenwick, Joshua Fierer, Roger G. Finch, James M. Fleckenstein, Christina Forstner, Federico Foschi, Pierre-Edouard Fournier, Martyn A. French, Kenneth L. Gage, Lynne S. Garcia, Joaquim Gascon, Arturo S. Gastañaduy, Philippe Gautret, William M. Geisler, Khalil G. Ghanem, Tommaso Giani, Maddalena Giannella, Bruce L. Gilliam, Michel Gilliet, Carol A. Glaser, Youri Glupczynski, John W. Gnann, Ellie J.C. Goldstein, Bruno Gottstein, Frederique Gouriet, Patti E. Gravitt, Michael D. Green, Stephen T. Green, Andreas H. Groll, Roy M. Gulick, Arjun Gupta, Gilbert Habib, Stephan Harbarth, Marianne Harris, Frederick G. Hayden, David J. Hetem, Philip C. Hill, Bernard Hirschel, Aimee C. Hodowanec, Louis Hoffart, Christian Hoffmann, Steven M. Holland, Peter W. Horby, David J. Horne, Sami Hraiech, Mark W. Hull, Angela Huttner, Richard J.M. Ingram, Jasmin Islam, Michael G. Ison, Scott H. James, Claire Jenkins, Stephen G. Jenkins, Jørgen Skov Jensen, Christine Johnston, Theodore B. Jones, Stephen J. Jordan, Kathleen G. Julian, Yasuyuki Kato, Carol A. Kauffman, Keith S. Kaye, Michael P. Keane, James Keeney, Paul Kelly, Stephen J. Kent, Winfried V. Kern, Yoav Keynan, Andrea A. Kim, Isabelle Koné-Paut, Chris Kosmidis, Aloys C.M. Kroes, Frank P. Kroon, Thomas G. Ksiazek, F. Matthew Kuhlmann, Ed J. Kuijper, Jennie H. Kwon, George B. Kyei, Karine Lacombe, Philippe Lagacé-Wiens, Jean-Christophe Lagier, Theresa Lamagni, Luce Landraud, Fanny Lanternier, Kerry L. LaPlante, Stephen D. Lawn, Steven J. Lawrence, Hakan Leblebicioglu, Nelson Lee, James E. Leggett, Philippe Lehours, Pierre-Yves Levy, Rainer G. Leyh, Rebecca A. Lillis, Direk Limmathurotsakul, Jennifer Lin, H.D. Alan Lindquist, Benjamin A. Lipsky, Christina Liscynesky, David Looney, Olivier Lortholary, Franklin D. Lowy, Benjamin J. Luft, Philip A. Mackowiak, Paul A. MacPherson, Valérie Maghraoui-Slim, Patrick W. Mallon, Julie E. Mangino, Oriol Manuel, Oscar Marchetti, Kristen M. Marks, Kieren A. Marr, Jeanne Marrazzo, Jonas Marschall, David H. Martin, Frédéric Matonti, Richard S. Matulewicz, Kenneth H. Mayer, Russell J. McCulloh, Rose McGready, Rennatus Mdodo, Simon Mead, Francis Mégraud, Graeme Meintjes, Sarah C. Metcalf, Marian G. Michaels, Giovanni Battista Migliori, Michael A. Miles, Alastair Miller, Matthew J. Mimiaga, Marie-Paule Mingeot-Leclercq, Elizabeth Ann Misch, Makedonka Mitreva, Julio S.G. Montaner, Caroline B. Moore, Patricia Muñoz, Jose Muñoz, Clinton K. Murray, Didier Musso, Mable Mutengo, Misha M. Mutizwa, Kurt G. Naber, Pavithra Natarajan, Santiago Neme, Paul N. Newton, Ronald A. Nichols, Lindsay E. Nicolle, François Nosten, Luigi D. Notarangelo, Thomas B. Nutman, Paul Nyirjesy, P. Ronan O'Connell, Steven M. Opal, L. Peter Ormerod, Douglas R. Osmon, Marie Boulze Pankert, Giuseppe Pantaleo, Laurent Papazian, Diane M. Parente, Philippe Parola, Shadi Parsaei, Manuel A. Pascual, Rupa Patel, Eleni Patrozou, Jean-Michel Pawlotsky, Sharon J. Peacock, Jean-Claude Pechère, Ivan Pelegrin, Barry S. Peters, Edgar J.G. Peters, Jeannine M. Petersen, Lyle R. Petersen, Vidmantas Petraitis, Luu-Ly Pham, Albert Picado, Adrian Pilatz, Benoit Pilmis, María-Jesús Pinazo, Mathias W. Pletz, Jason M. Pogue, Evelyn L. Polgreen, Philip M. Polgreen, Klara M. Posfay-Barbe, William G. Powderly, Rachel Presti, Guy Prod'hom, Mirja Puolakkainen, Thomas C. Quinn, Didier Raoult, Raymund R. Razonable, Robert C. Read, Robert R. Redfield, Rob J. Rentenaar, Steven J. Reynolds, Camillo Ribi, Malcolm D. Richardson, Michele L. Ritter, Antoine Roch, Jürgen Kurt Rockstroh, Amanda Rojek, José R. Romero, Suzan H.M. Rooijakkers, Daniel Rosenbluth, Sergio D. Rosenzweig, Gian Maria Rossolini, Ethan Rubinstein, Greg Ryan, Steven A. Safren, Vikrant V. Sahasrabuddhe, Pekka A.I. Saikku, Mohammad M. Sajadi, Michelle R. Salvaggio, Carlos A.Q. Santos, Michael J. Satlin, Anthony J. Schaeffer, Christoph Schimmer, Robert T. Schooley, Richard F. Schumacher, Beverly E. Sha, Daniel S. Shapiro, Gerard Sheehan, David M. Shlaes, Shmuel Shoham, Cameron P. Simmons, Dennis W. Simon, Matthew S. Simon, Kari A. Simonsen, Mary P.E. Slack, Tyrel T. Smith, Jack D. Sobel, Maria Souli, Shruti Sridhar, James M. Steckelberg, Dennis L. Stevens, Heather Strah, A. Willem Sturm, Somnuek Sungkanuparph, Sarah J. Tabrizi, Evelina Tacconelli, Chen Sabrina Tan, Randy A. Taplitz, Guillemette Thomas, Lora D. Thomas, Franck Thuny, Guy Thwaites, Frederic Tissot, Tone Tønjum, Francesca J. Torriani, Christian Toso, Paul M. Tulkens, Allan R. Tunkel, Claire E. Turner, Andrew P. Ustianowski, Françoise van Bambeke, Reinout van Crevel, Diederik van de Beek, Christian van Delden, Menno M. van der Eerden, Jos W.M. van der Meer, Tom van der Poll, Jakko van Ingen, Jos van Putten, Bernard P. Vaudaux, Sten H. Vermund, Raphael P. Viscidi, Kumar Visvanathan, Govinda S. Visvesvara, Lorenz von Seidlein, Florian M.E. Wagenlehner, Anna Wald, Thomas J. Walsh, David C. Warhurst, David W. Warnock, David A. Warrell, Mary J. Warrell, Adilia Warris, Richard R. Watkins, David J. Weatherall, Rainer Weber, Wolfgang Weidner, Jonathan R. White, Peter J. White, James Whitehorn, Richard J. Whitley, Christopher J.M. Whitty, Willem Joost Wiersinga, Mark H. Wilcox, Thomas N. Williams, Cara C. Wilson, Mary Elizabeth Wilson, Hilmar Wisplinghoff, Robin Wood, Richard G. Wunderink, David Wyles, Zhi-Tao Yang, Jonathan S. Yoder, Najam A. Zaidi, Andrea J. Zimmer, Jane N. Zuckerman, and Alimuddin Zumla
- Published
- 2017
- Full Text
- View/download PDF
50. MBL2 deficiency is associated with higher genomic bacterial loads during meningococcemia in young children
- Author
-
Ray Borrow, Malcolm Guiver, Margaret A. Johnson, Nigel Klein, Edward B. Kaczmarski, Robert C. Read, Dominic L. Jack, Malcolm W. Turner, and Thomas C. Darton
- Subjects
Male ,Microbiology (medical) ,Adolescent ,Genotype ,Genotyping Techniques ,Bacteremia ,Biology ,Neisseria meningitidis ,Meningococcal disease ,medicine.disease_cause ,Microbiology ,Cohort Studies ,meningococcal infection ,medicine ,Humans ,mannose-binding lectin ,Child ,Mannan-binding lectin ,Innate immune system ,Pattern recognition receptor ,Infant ,General Medicine ,Genomic bacterial load ,Gram-negative sepsis ,MBL deficiency ,medicine.disease ,Acquired immune system ,Bacterial Load ,Meningococcal Infections ,Blood ,Infectious Diseases ,Child, Preschool ,Immunology ,Female ,Disease Susceptibility ,Metabolism, Inborn Errors - Abstract
Mannose binding lectin (MBL2) is a soluble pattern recognition receptor that is key to generating innate immune responses to invasive infection, including against the cardinal Gram-negative bacterium Neisseria meningitidis. Individuals homozygous or heterozygous for any of three variant alleles of MBL2 (O/O or A/O genotypes) have deficient concentrations of MBL2 in circulating blood, but previous studies linking MBL deficiency to susceptibility to meningococcal disease have not revealed a consistent association. We genotyped 741 patients with microbiologically – proven meningococcal disease and correlated MBL2 genotype with plasma bacterial load of N. meningitidis with blood samples taken during hospital admission. We show that individuals with genotypes compatible with MBL2 deficiency have higher measurable levels of bacterial plasma genomic load with the greatest effect seen in children
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.