VAccine deveLopment for complex Intracellular neglecteD pAThogEns (VALIDATE)

Lead Research Organisation: University of Oxford
Department Name: The Jenner Institute

Abstract

For several complex pathogens, we have an incomplete understanding of the kind of immune response we need an effective vaccine to induce. In part this is because these pathogens live inside cells and can hide from the host immune response. This makes it very difficult to develop vaccines against these pathogens. The diseases caused by these pathogens, including tuberculosis (TB), leprosy, leishmaniasis and melioidosis, disproportionately affect people in low and middle-income countries (LMICs) and are often neglected due to the poor predicted commercial return and marginalisation of affected populations. The current approach to vaccine development is to identify a part of the immune response that might be important, develop vaccines that induce that kind of immune response and then test that vaccine to see if it works, first in animal models and then in human clinical trials. This approach does not work for these complex pathogens and is slow and inefficient. In this Network we will adopt an iterative approach and develop more accurate working models of the immune response by cycles of repeated integration of data from human clinical studies and animal models across the different pathogens. The inclusion of vets working in large animal models will mean the results obtained are relevant for the development of animal vaccines as well. The inclusion of partners from LMIC where these diseases are endemic, together with partners from industry will help ensure real world relevance. Furthermore, the smaller animal models will allow us to easily look at single components of the immune system. The insight gained into immune mechanisms will allow us to design and develop vaccines more efficiently and more effectively.

This Network will provide a unique opportunity to bring together individuals working on four exemplar complex intracellular pathogens (M.tb, M.leprae, Leishmania spp. and B.pseudomallei), which share a common lifestyle as pathogens that live in a particular host cell called macrophages, induce similar end-stage pathologies in humans and are known to affect host immune and metabolic responses. The horizontal collaborations established throughout this network, together with the provision of a protected environment for early data sharing, will exploit the biological synergies between these pathogens. Positive and negative results can be disseminated rapidly, and partners working on each of the complex diseases can learn from the successes and failures of those working on the other diseases. By understanding mechanisms that lead from infection to disease, we plan to uncover common approaches to inform vaccine development strategies for these and other complex intracellular pathogens.

Key Network activities and deliverables include annual meetings for knowledge sharing; creation of a virtual network for real-time data sharing; pump-priming funding to fill knowledge gaps, and provide data to enhance the competitiveness of larger grant applications; a website and other communications to enhance collaborations and research dissemination; and career development by funding, training and mentoring of early post-doctoral and new PI researchers. All activities work towards more effective vaccine development for these important diseases, and the improvement of research capabilities in our partner LMICs.

Technical Summary

For several complex intracellular pathogens, we have an incomplete understanding of protective immunity. These diseases, including TB, leprosy, leishmaniasis and melioidosis, disproportionately affect LMICs and are often neglected due to the poor predicted commercial return and marginalisation of affected populations. The current linear approaches, which identify a component of the immune response, induce it by vaccination and evaluate efficacy in animal models and clinical trials, are flawed and have proven inefficient for intracellular pathogens. In this Network we will adopt an iterative approach and develop accurate working models of immunity by repeated integration of data from human experimental studies and animal models across the different pathogens. Veterinary vaccinologists and large animal models will facilitate the One Health agenda, and experimental medicine studies and LMIC and industrial partners will ensure relevance and translational value. The smaller animal models will provide the tools with which to easily dissect mechanisms and immune pathways. The integrated approach incorporates in-silico and in-vitro tools to delineate mechanisms, that will feed directly into vaccine design and development.

This Network will provide a unique opportunity to bring together individuals working on four exemplar complex intracellular pathogens (M.tb, M.leprae, Leishmania spp. and B.pseudomallei), which share a common lifestyle as pathogens of macrophages, induce similar end-stage pathologies (granulomatous inflammation, tissue remodeling) and alter host immune and metabolic responses. The horizontal collaborations established throughout this network, together with the provision of a protected environment for early data sharing, will exploit the biological synergies. By interrogating mechanisms that lead from infection to disease, we plan to uncover common approaches to inform vaccine development strategies for these and other complex intracellular pathogens.

Planned Impact

There are many beneficiaries of the work carried out under this Network.

The first obvious beneficiaries of this work are basic scientists, clinicians, veterinarians, epidemiologists and clinical trial investigators working in the field of vaccinology, particularly vaccines for the exemplar pathogens that are the focus of this Network. The scientific data sharing, pump-priming funding, and support to existing and new collaborations that are all supported by this Network will lead to new scientific insights which will facilitate vaccine research and development. The networking activities are designed to maximise the impact of the Network. Furthermore, the academic community will gain from the training, mentoring and knowledge sharing that will occur as part of this network.

Industrial colleagues in vaccinology will also benefit from this work, both in SMEs and in large pharmaceutical companies. Industrial involvement in these complex pathogens is currently limited, in part because of a predicted poor return on investment. This Network aims to 'de-risk' vaccine development by providing novel insights into host immunity and in sharing across pathogens any lessons learned so the broader applicability of any novel findings can be rapidly evaluated.

Ultimately, this Network will 'de-risk' vaccine development for complex intracellular pathogens and allow the design and development of vaccine candidates that are more likely to succeed. The development of effective vaccines for any of the pathogens outlined here would have a very significant impact on global mortality and morbidity, particularly in LMIC where these diseases are endemic. The health and ultimately wealth of LMIC will improve with the development of effective vaccines against these devastating pathogens.

Policy makers, in the UK and LMIC, together with the funders of scientific research, also have the potential to benefit from the work outlined in this Network. The insights gained will allow better selection and prioritisation of vaccine candidates at an earlier stage, and a greater likelihood of success.

Publications

10 25 50

publication icon
Kronsteiner B (2019) Diabetes alters immune response patterns to acute melioidosis in humans. in European journal of immunology

publication icon
Aguilar-López BA (2020) Mitochondria: An Integrative Hub Coordinating Circadian Rhythms, Metabolism, the Microbiome, and Immunity. in Frontiers in cell and developmental biology

publication icon
Parbhoo T (2022) Persistence of Mycobacterium tuberculosis in response to infection burden and host-induced stressors. in Frontiers in cellular and infection microbiology

 
Description Liaison with MPs
Geographic Reach National 
Policy Influence Type Implementation circular/rapid advice/letter to e.g. Ministry of Health
URL https://www.validate-network.org/un-high-level-tb-meeting-2018
 
Description "Sepsis induced stress and Network Analysis on Pathogen Expression of stress-combat genes: the SNAPE study" (Mukhopadhyay - P033 outcome)
Amount ₹2,149,848 (INR)
Organisation Indian Council of Medical Research (ICMR) 
Sector Public
Country India
Start 02/2023 
End 01/2026
 
Description "Understanding the effect of diabetes mellitus on immunometabolic response to Bps in Indian patients" (Mukhopadhyay - P033 output)
Amount ₹5,977,968 (INR)
Organisation Indian Council of Medical Research (ICMR) 
Sector Public
Country India
Start 02/2023 
End 01/2026
 
Description Additional MRC funding for VALIDATE 2022
Amount £450,000 (GBP)
Organisation Medical Research Council (MRC) 
Sector Public
Country United Kingdom
Start 07/2022 
End 03/2023
 
Description BSI Conference Co-hosting 2019
Amount £5,000 (GBP)
Organisation British Society For Immunology 
Sector Charity/Non Profit
Country United Kingdom
Start 01/2019 
End 12/2019
 
Description BSI Travel Award (Rachel Tanner Fellowship)
Amount £1,000 (GBP)
Organisation British Society For Immunology 
Sector Charity/Non Profit
Country United Kingdom
Start 01/2020 
End 02/2020
 
Description Bactivax (Rajko Reljic/Siobhan McClean/Danny Altman - member collaboration)
Amount € 4,009,373 (EUR)
Funding ID 860325 
Organisation European Commission H2020 
Sector Public
Country Belgium
Start 10/2019 
End 09/2023
 
Description Basque Government Fellowship (Julen Tomas Cortazar, P023)
Amount € 150,000 (EUR)
Organisation Basque Government 
Sector Public
Country Spain
Start 02/2020 
End 01/2023
 
Description Develop novel test for PKDL/leprosy diagnosis
Amount $399,623 (USD)
Funding ID INV-048694 
Organisation Bill and Melinda Gates Foundation 
Sector Charity/Non Profit
Country United States
Start 11/2022 
End 10/2024
 
Description Developing a vaccine to prevent death from melioidosis in people with type 2 diabetes mellitus in low- and middle-income countries
Amount £1,956,321 (GBP)
Funding ID NIHR300791 
Organisation National Institute for Health Research 
Sector Public
Country United Kingdom
Start 02/2021 
End 01/2026
 
Description Diabetes and infection: developing a network for using real-world datasets (Susanne Dunachie P016 outcome)
Amount £26,728 (GBP)
Organisation United Kingdom Research and Innovation 
Department Global Challenges Research Fund
Sector Public
Country United Kingdom
Start 01/2019 
End 12/2020
 
Description Firland Foundation grant (P021)
Amount $50,000 (USD)
Organisation Firland Foundation 
Sector Charity/Non Profit
Country United States
Start 01/2020 
End 01/2022
 
Description Industrial Challenge Strategy Fund
Amount £600,000 (GBP)
Organisation Innovate UK 
Sector Public
Country United Kingdom
Start 03/2018 
End 03/2019
 
Description Interaction of BCG with immune responses of leishmania in a mouse model (Javier Salguero - P003 outcome)
Amount £90,000 (GBP)
Organisation Ministry of Science and Innovation (MICINN) 
Sector Public
Country Spain
Start 01/2019 
End 12/2022
 
Description Internal GCRF funding award to Panjaporn Chaichana (VALIDATE P007 output)
Amount £29,928 (GBP)
Organisation United Kingdom Research and Innovation 
Department Global Challenges Research Fund
Sector Public
Country United Kingdom
Start  
 
Description MRC DPFS 2020 Award 'Developing a vaccine for Burkholderia pseudomallei - a Phase I Clinical Trial' (Dunachie, Brett, McShane, Chantratita; melioidosis workshop output)
Amount £3,238,214 (GBP)
Organisation Medical Research Council (MRC) 
Sector Public
Country United Kingdom
Start  
 
Description NIHR Senior Investigator - Prof Helen McShane (ref NIHR200225)
Amount £60,000 (GBP)
Funding ID NIHR200225 
Organisation National Institute for Health Research 
Sector Public
Country United Kingdom
Start 04/2019 
End 03/2023
 
Description Optimization of lead melioidosis and glanders vaccine formulations (Brett, Dunachie; melioidosis workshop outcome)
Amount $733,976 (USD)
Organisation Defense Threat Reduction Agency 
Sector Public
Country United States
Start 01/2020 
End 12/2022
 
Description Preclinical development of multicomponent vaccines against melioidosis (Charles Gauthier & Siobhan McClean, VALIDATE member collaboration)
Amount $703,800 (CAD)
Organisation Canadian Institutes of Health Research 
Sector Public
Country Canada
Start 01/2022 
End 12/2027
 
Description Public Health England PhD studentship (P038)
Amount £105,100 (GBP)
Organisation Public Health England 
Sector Public
Country United Kingdom
Start 10/2020 
End 09/2023
 
Description VALIDATE Network TB Vaccine Support
Amount $1,538,836 (USD)
Funding ID INV-031830 
Organisation Bill and Melinda Gates Foundation 
Sector Charity/Non Profit
Country United States
Start 07/2021 
End 03/2024
 
Description Wellcome Enriching Engagement - BCG100 Programme
Amount £21,103 (GBP)
Organisation Wellcome Trust 
Sector Charity/Non Profit
Country United Kingdom
Start 03/2021 
End 11/2021
 
Title Sharing of documents with other GCRF networks 
Description Items shared with the other GCRF Vaccine Networks (HIC-VAC, Bactivac, IMPRINT, IVVN) to streamline their processes and help them with their procedural set up: VALIDATE pump-priming call template and guidance documents, training grant template and guidance (all shared with BBSRC Vector Networks as well), Network Management Board confidentiality agreement, tiered membership structure, membership and members' directory templates, member terms & conditions, first annual meeting agenda, as well as being asked for feedback and lessons learned from our various grant calls and procedures. 
Type Of Material Improvements to research infrastructure 
Year Produced 2018 
Provided To Others? No  
Impact The sharing of our documents enabled the other networks to more quickly and easily set up their processes and procedures, and template documents. 
 
Title Oxysterol_LCMS/MS raw data_whole cell/mitochondria 
Description Mass spectrometry data (raw mass spectra files) for whole cell and mitochondrial oxysterols derived from THP-1 monocytes and macrophages, SH-SY5Y and human peripheral blood mononuclear cells 
Type Of Material Database/Collection of data 
Year Produced 2020 
Provided To Others? Yes  
Impact This is an output from VALIDATE Training Grant T022 (Khushboo Borah). 
URL https://data.mendeley.com/datasets/npcc7yrfjk/2
 
Title VALIDATE Data Sharing Portal and VALIDATE Research Data Analyst 
Description In January 2019 we launched the VALIDATE Data Sharing Portal, which aimed to facilitate and encourage the sharing of published and unpublished data amongst our members, to enable cross-comparisons (e.g. of similar assays across pathogens, or across assays for one pathogen, or across species or countries) that will accelerate vaccine development, as well as to avoid duplication of effort where unpublished negative data already exists. Our VALIDATE Research Data Analyst was in charge of the database and facilitated the uploading and analyses of data by our members. This has resulted in two publications to date: 1. Satti I et al 2022. Inflammation and immune activation are associated with risk of Mycobacterium tuberculosis infection in BCG-vaccinated infants | Nature Communications DOI 10.1038/s41467-022-34061-7 2. Tanner R et al 2020. Tools for Assessing the Protective Efficacy of TB Vaccines in Humans: in vitro Mycobacterial Growth Inhibition Predicts Outcome of in vivo Mycobacterial Infection. Frontiers in Immunology DOI: 10.3389/fimmu.2019.02983 Our Research Data Analyst, Dr Deniz Cizmeci, moved to a post-doc position at Harvard University in early May 2019 and we had significant challenges recruiting an appropriate replacement, with no appointable candidates in three rounds of advertisement. Our fourth round resulted in an appointable candidate, who started in post on 1 January 2020. This means we had a gap of eight months in this post that was beyond our control, but moved this area of the Network forwards with our new VALIDATE Bioinformatician/Computational Biologist, Dr Mirvat Surakhy, who was in post from March 2020 to Jan 2021. Sadly, it became clear that neither our Bioinformatician or the VALIDATE Data Portal were being used much by members, so we decided to close both. Before she left, Mirvat Surakhy added a comprehensive list of data resources (including free training options and free databases) to the VALIDATE website to support members (https://www.validate-network.org/bioinformatics). Instead of the data portal, we now have a list of data sets available via VALIDATE members from pump-priming/fellowship projects at https://www.validate-network.org/data-sharing for members. 20 data sets are listed as available for members, ranging from clinical trial data (e.g. microarray, RNAseq) to animal model lab data (e.g. ELISA, MGIA, flow cytometry). 
Type Of Material Database/Collection of data 
Year Produced 2019 
Provided To Others? Yes  
Impact List of data sets available via VALIDATE members from pump-priming/fellowship projects at https://www.validate-network.org/data-sharing for members. 
URL https://www.validate-network.org/data-sharing
 
Description VALIDATE Fellowships - Jomien Mouton 
Organisation University of Stellenbosch
Country South Africa 
Sector Academic/University 
PI Contribution VALIDATE aims to encourage Continuing Professional Development (CPD) and career progression amongst its members. As part of this, two VALIDATE Fellowships were awarded in 2018. These VALIDATE Fellowships were open to Early Career Researcher (Associate) Members to fund two years' independent scientific research with the aim of producing initial/pilot data, and a funding and experience track record, that can be used by the Fellow to go on to obtain independent external funding and launch their career as a Principal Investigator and/or Group Leader. After competitive application to the Network Management Board, and subsequent interviews, the two fellowships were awarded to: 1. Dr Jomien Mouton, Stellenbosch University South Africa, "Identification of latency associated antigens and biosignatures associated with Mycobacterium tuberculosis" 2. Dr Rachel Tanner, University of Oxford, UK "Characterising the BCG-induced antibody response to inform the design of improved vaccines against M.tuberculosis, M.leprae and M.bovis" We are proud that both awardees happen to be female, and one is also LMIC. Both Jomien and Rachel joined our Network Management Board for the duration of their Fellowships, giving them experience at this senior level and in reviewing grant and membership applications made to VALIDATE, and giving VALIDATE valuable ECR input to our NMB.
Collaborator Contribution Fellowship Aims 1. Characterise the transcriptional profile of replicating and non-replicating Mycobacterium tuberculosis populations using a single-cell dual host-pathogen RNA sequencing approach. 2. Identify biosignatures associated with replicating and non-replicating M. tuberculosis in host cells. 3. Identify potential antigen candidates associated with non-replicating persister M. tuberculosis using bioinformatics analyses. 4. Assess the immunogenicity of selected candidate latency antigens in human populations in South Africa to which tuberculosis (TB) is endemic. Fellowship Outcomes To elucidate how macrophages interact with replicating and non-replicating mycobacterium, we employed a dual-fluorescent replication reporter in combination with flow cytometry to identify and isolate host cells containing either non-replicating or replicating mycobacteria for RNA sequencing and fluorophore-conjugated antibody staining. Our results indicate that macrophages containing differentially replicating mycobacteria are distinctly associated with either M1 or M2 macrophage polarisation. The VALIDATE Fellowship has had a tremendous impact on my career development. Specifically, it allowed me to be appointed as a researcher, which afforded me the opportunity to supervise my own students and apply for funding as a primary investigator. In addition, I have had the opportunity to serve on the Network Management Board (NMB) and expand my network by meeting new people at the annual meetings. The funding I have received as part of my fellowship has made high-end research possible that contributed to scarce skills development in RNA sequencing in South Africa. Progress during the course of this funding includes the following: 1. Intracellular M. tuberculosis H37Rv growth inside alveolar macrophages from black versus clear bronchoalveolar lavage samples has been monitored using a combination of the fluorescence dilution (FD) technology and flow cytometry. This data was combined with data from another research group in our department that looked at cytokine responses of black versus clear alveolar macrophages for a publication (Objective 1). We have prepared a manuscript (2nd co-author) entitled, "Inhaled particulate matter affects immune responsiveness of human lung phagocytes to mycobacterial species" that was accepted in July 2021 (objective 1). 2. A material transfer agreement between Stellenbosch University and Helmholtz Institute for RNA-based Infection Research (HIRI) in Wursburg, Germany for shipping of extracted bacterial and host RNA for dual and single-cell RNA sequencing were set up (Objective 1). Although RNA sequencing have been performed at the South African Medical Research Council for the proposed work (to avoid covid-related delays), we maintain our relationship with collaborators at Wursburg University for future projects and to provide support with RNA sequencing analyses. 3. The dual host-bacterial RNA extraction protocol resulted in low bacterial RNA yield. In the interest of limited time, and restrictions due to circumstances related to COVID-19 we focused on RNA extraction of macrophages containing differentially replicating mycobacteria for RNA sequencing. The RNA extraction protocol has been optimised and provides good quality and high yield RNA. RNA samples were prepared for sequencing in May 2021 at the South African Medical Research Council Sequencing Unit in Cape Town, South Africa (objective 1). The results will be included in the PhD thesis of Mrs. Zimvo Obasa (I am her co- supervisor), who is expected to graduate in March 2022. Additionally, the RNA sequencing results will be included in a manuscript that is in preparation. 4. Progress on objectives 2 - 4 have subsequently been delayed due to circumstances related to COVID-19, however related work to the proposed work included investigating the relation between persister formation and clinical outcome in TB patients. This study was undertaken by my MSc student, Mr. Julian Coetzee (who will graduate in December 2021, cum laude). He employed a combination of bioinformatics analyses and experimental research to determine whether M. tuberculosis strains from TB patients who were considered cured, but have relapsed, or failed treatment (based on PET-CT imaging), are more likely to be predisposed to persister formation than those who remained "cured" using phenotypic and genotypic approaches. To achieve this, persister proportions in all clinical isolates (taken at baseline) from cured, recurrent/failed patient groups using fluorescence dilution (FD) and flow cytometry were assessed. Additionally, comparative next generation sequencing analyses of the isolates were performed to determine whether sequence variation predisposes persister formation in clinical isolates (taken at baseline) from patient groups. Results from this work revealed a significantly higher proportion of persisters in clinical isolates from patients who have remaining lesion activity in the lung (post-treatment) based on PET-CT imaging. This result suggests that the inherent tendency to form persister-like cells may have an impact on pulmonary TB treatment outcome. Data suggests that persister-like cell formation may be strain dependent. Results from this research are currently being written up for a manuscript. 5. Ms Lesedi Dikhoba, a MSc student working on aspects of the project, utilised a more comprehensive cytokine panel for flow cytometry to assess macrophage polarisation in response to replicating and non-replicating M. tuberculosis. She has demonstrated that we are able to identify a distinct M1-like macrophage polarisation (inflammatory) associated with non-replicating M. tuberculosis (MHCII and CD274) and a M2-like macrophage polarisation (anti-inflammatory) associated with replicating M. tuberculosis (Arg-1) (objective 2). She submitted this work as part of her MSc thesis for examination in May 2021, she is expected to graduate in December 2021. [As a result of the COVID-19 pandemic and implementation of restrictions in the research environment in South Africa during 2020/2021, experimental work were severely affected. VALIDATE granted Jomien a no-cost extension for this.]
Impact Fellowship Outputs Student supervision: o 2 MSc students (primary supervisor), will graduate in Dec 2021 o 2 Hons students (co-supervision), graduated in Dec 2019/2020 o 1 PhD student (co-supervision), graduated March 2021 o 2 PhD students (co-supervision), expected to graduate in December 2021 Workshops Attended: o AMNIS® IMAGESTREAM®XMARKII Workshop, Cape Town, May 2019 Publications: o Apiyo D, Mouton JM, Louw C, Sampson SL, Louw TM. A rigorous approach to develop, validate and evaluate a dynamic mathematical model predicting Mycobacterium smegmatis growth in physiologically relevant conditions. Journal of Theoretical Biology. (under revision July 2021) o Paarwater B, Mouton JM, Sampson SL, Malherbe S, Kotze L, Walzl G, du Plessis N. Inhaled particulate matter affects immune responsiveness of human lung phagocytes to mycobacteria. American Journal of Physiology-Lung Cellular and Molecular Physiology. Accepted July 2021. Impact factor: 4.037. o Parbhoo T, Sampson SL, Mouton JM. Recent developments in the application of flow cytometry to advance our understanding of Mycobacterium tuberculosis physiology and pathogenesis. Cytometry Part A 2020. 97(7):783. Impact factor: 3.124. o Gallant J, Mouton JM, Ummels R, ten Hagen-Jongman C, Kriel N, Pain A, Warren RW, Bitter W, Heunis T, Sampson SL. Identification of gene fusion events in Mycobacterium tuberculosis that encode chimeric proteins. NAR Genomics and Bioinformatics 2020. 2(2): lqaa033. o Mouton JM, Heunis T, Dippenaar A, Gallant JL, Kleynhans L, Sampson SL. Comprehensive characterisation of the attenuated double auxotroph Mycobacterium tuberculosis ?leuD?panCD as an alternative to H37Rv. Frontiers in Microbiology 2019. 10: 1922. Impact factor: 4.259 Publications in preparation: o Obasa Z, Smith L, Mouton JM and Sampson SL. Different approaches to study Mycobacterium tuberculosis persister formation in vivo. BMC microbiology journal, submission in October 2021. Impact factor 3.19 Honours/Rewards: o Top 20 Post-doc award (Nov 2018) o Promotion to appointed as Researcher (Nov 2018) o NRF Y-rating (Nov 2020) Funding: o NRF Y-rating, incentive funding (Jan 2021) o Involved in a subsequent successful VALIDATE pump-priming grant application as co-investigator, with Samantha Sampson (SU) and Andrea Zelmar (LSHTM; a new collaborator), for £37,031. o As a result of being a SU staff member, Jomien was a co-investigator for a National Research Foundation National Equipment Fund application, to acquire an Amnis Imagestream Imaging Flow Cytometer. This grant was successful, with R12,000,000; the instrument is the first of its kind in Africa and arrived in July 2019, and Jomien subsequently capacity builds by presenting Amnis Imaging Flow Cytometer data analyses workshops in the division, teaching honours students, and leading a Joint Stellenbosch University-University of Cape Town bacterial flow cytometry interest group. o Submitted a R01 funding application to the National Institute of Health as PI in collaboration with VALIDATE fellow, Dr. Rachel Tanner from Oxford University, Prof. Sophie Helaine from Harvard Medical School and Prof. Samantha Sampson from Stellenbosch University entitled, "Evaluation of the interplay between host cells and mycobacteria in drug-resistant tuberculosis (DR-TB) patients from the Eastern Cape, South Africa" in July 2021 (outcome pending). o Jomien submitted a Dorothy Temple Cross Tuberculosis International collaboration grant as co-PI in collaboration with VALIDATE Fellow, Dr Rachel Tanner ( Oxford University) entitled, "Combining expertise in mycobacterial killing assays to evaluate functional immune and treatment responses in drug-resistant tuberculosis (DR-TB) patients" in October 2020. Although this was not funded, Jomien and Rachel aim to incorporate feedback and resubmit in 2021 and/or submit our application to another funding call. o As a member of VALIDATE Jomien also participated in a larger European COST funding application that included 97 VALIDATE members from 49 countries (unsuccessful). o Ms Lesedi Dikhoba, MSc student working on aspects of the project, received a bursary to support her 2nd year of studies from the National Research Foundation. New Collaborations: o Associate Professor Sean Wasserman, University of Cape Town, South Africa (consultant on NIH R01 application submitted in July 2021) o Alex Westermann, Antoine-Emmaneul Saliba, Helmholtz Institute for RNA-based infection research, Wursburg University, Germany o Sophie Helaine, Harvard Medical School, Boston, United States of America (co-investigator on NIH R01 application submitted in July 2021) o Rachel Tanner, University of Oxford, United Kingdom (co-investigator on NIH R01 application submitted in July 2021) Talks/Presentations: o Poster Presentation at the international Meeting 'Bacterial morphogenesis, survival and virulence: regulation in 4D' in Cape Town, Nov 2019 o Presentations at the VALIDATE 2018 and 2019 Annual Meetings Guest Lecturer: o What if I don't want to work in the BSL3? Departmental Year Day, Cape Town, November 2019. o M. tuberculosis in the host: Persisters and PE/PPE proteins, VALIDATE Winter Seminar Series, 24 November 2020. Student presentations: o Maqeda Z, Smith L, Mouton J, Sampson SL. CYTO2020 (International society for advanced cytometry), 04-05 August 2020, "Mycobacterium tuberculosis at the host-pathogen interface", ePoster o Maqeda Z, Smith L, Mouton J, Sampson SL. 63rd Annual Academic Day 2020, Faculty of Medicine and Health Sciences, Stellenbosch University, 26 - 27 August 2020, "Mycobacterium tuberculosis at the host-pathogen interface", ePoster o Coetzee J, Sampson SL, Mouton JM. 63rd Annual Academic Day, Faculty of Medicine and Health Sciences, Stellenbosch University, 26-27 August 2020, "The relation between persisters and clinical outcome in tuberculosis (TB) patients", ePoster o Coetzee JL, Sampson SL, Mouton JM. 6th SATB virtual conference. June 2021, "Investigating the relation between persister formation and clinical outcome in tuberculosis (TB) patients", ePoster o Dikhoba L, Gutschmidt A, Sampson SL, Mouton JM. Departmental Research Day, Faculty of Medicine and Health Sciences, Stellenbosch University, 21 November 2019, "Identification of biosignatures associated latent Mycobacterium tuberculosis.", ePoster o Dikhoba L, Gutschmidt A, Sampson SL, Mouton JM. The Conversation Africa Scientific Article Writing Competition, December 2019, "Identification of biosignatures associated latent Mycobacterium tuberculosis.", ePoster o Dikhoba L, Gutschmidt A, Sampson SL, Mouton JM. 63rd Annual Academic Day, Faculty of Medicine and Health Sciences, Stellenbosch University, 26-27 August 2020, "Identification of biosignatures associated latent Mycobacterium tuberculosis.", ePoster Scientific involvement: o Organizer of Bacterial Flow Cytometry Forum (Joint SU-UCT), March 2014 - March 2020 o VALIDATE Network Board Member, Nov 2018 - 2021 o SA Acid Fast Organizing Committee, August 2019 o Reviewer for PlosOne, Tuberculosis, Epigenetics o Examination of 2 MSc theses (Stellenbosch University, University of KwaZulu-Natal) Public engagement: o Participated in VALIDATE for Schools, by presenting a lecture to school pupils at a UK-based school about career path and research interests in May 2021. o Assisted 2 school pupils from local South Africa schools to "job shadow" in 2019, showing them the department, laboratory and campus and engaging with them about her science interests, experimental procedures that she use in the laboratory and discussing what our careers involve. o Students directly or indirectly involved in the project (2 MSc and 1 PhD students that Jomien either supervised/co-supervised) presented aspects of the project to other scientists at scientific conferences, or the general public at related meetings. For example, Ms Lesedi Dikhoba, the 2nd year MSc student working on aspects of the project presented at The Conversation Africa workshop in Stellenbosch on 3 December 2019. o Presented a lecture on "M. tuberculosis in the host: Persisters and PE/PPE proteins" at the VALIDATE Winter Seminar Series on 24 November 2020 o Jomien has applied for funding from the Faculty of Medicine and Health Science Social Impact Committee for an outreach project, entitled "Vaccines saves lives" that involves radio interviews and visits to local clinics to inform the community about vaccines and vaccine development. Following this, she has prepared a written brief for an interview with local radio stations in the Eastern Cape to communicate the importance of vaccines to the more rural communities in the region. The radio interviews will take place in early 2021. Capacity development During the course of the VALIDATE project funding Jomien contributed to capacity building and skills development in the following ways; o Had 2 final year school pupils visit for a day in the Division and laboratory to communicate her research to and convey what a career in science involves. o Supervised 2 MSc students on work related to the project. o Co-supervised 3 PhD students, 1 MSc student and 2 Honours students. o Presented an Amnis Imaging Flow Cytometer data analyses workshops in the division from 2019 - 2020. o Presented 3 lectures in 2 modules to honours students in 2018 - 2021. o Led a Joint Stellenbosch University - University of Cape Town bacterial flow cytometry interest group in 2018 - 2020. o As a result of moving to the Eastern Cape in June 2020, Jomien offered training to students to use the BD FACSJazz cell sorter in the BSL3 facility for cell sorting of infectious samples for downstream analyses. Other During the contracts process in November 2018, Jomien noticed that VALIDATE's grant conditions require employment for the duration of the award. At Stellenbosch University post-doctoral fellows are not employed and receive tax-free bursaries on a contract basis. She stated in an email to us "As a young researcher, employment by the university would be extremely beneficial e.g., allowing supervision of students as a primary supervisor, applying for funding otherwise not eligible to apply for." VALIDATE agreed to pay the FEC so that she could be made an employee at University of Stellenbosch, a promotion that would not have happened without her Fellowship. As an employed researcher at Stellenbosch University she was able to apply for rating in December 2019 in the Y category (young researchers) from the National Research Foundation that aims to benchmark the quality of South African researchers against the best in the world. NRF ratings are allocated based on a researcher's recent research outputs and impacts perceived by international peer reviewers. Several South African universities require NRF evaluation and rating for appointments, or provide incentives for their staff members to acquire or maintain a rating. The outcome of this application has been successful and she has received a Y-rating for the following 5 years. As a result of this appointment she has supervised two South African MSc students (one female, one male) for work related to her VALIDATE project, as their primary supervisor (prior to this Jomien had co-supervised students only) who aim to submit in April 2021. This has obviously further spread the local capacity building value of Jomien's VALIDATE Fellowship, as well as contributing to the development of important aspects of Jomien's scientific career e.g. mentoring, project management, interpersonal skills and teaching. She published her first senior author publication in March 2020. VALIDATE also supported Jomien through her maternity leave from 14 August to 13 December 2019; without her VALIDATE Fellowship she would have had to take unpaid maternity leave. She states, "Being supported and encouraged in this way has taught me that women in science have the opportunity to take up different roles (e.g. mothers, wives, professionals etc.)." As part of her Fellowship, Jomien has been mentored by Prof Paul Kaye, at the University of York, UK, since November 2018. As a result of this connection, they are working on a review together and Paul has connected Jomien with colleagues in his department at York. Jomien also received mentorship from her SU host, Prof Samantha Sampson, and Prof Helena Kuivaniemi at SU. The VALIDATE Fellows were made VALIDATE Network Management Board (NMB) full members. At the VALIDATE NMB bi-monthly meetings Jomien has participated in discussions about sustainability of the network, recruitment of new members, and management of funds. As part of the NMB, Fellows had the opportunity to review funding applications for training grants and pump-priming projects, which Jomien says has "provided incredible insight into how seasoned researchers that are experts in their field review, critique and score funding applications". Importantly, this improved her knowledge about what fundable grant applications should look like, what reviewers look for and also how to review grant applications. Her exposure to the discussions by internationally recognised scientists during the NMB meetings have been invaluable for her development as a young scientist. Jomien says: 'Receiving the VALIDATE Fellowship has had a tremendous impact on my research and career. Apart from being appointed as a researcher and being able to supervise my own students, I have had the opportunity to serve on the NMB and expand my network by meeting new people at the annual meeting. The funding I have received as part of my fellowship has made research possible (eg. dual RNA sequencing and single cell RNA sequencing) that has not yet been done in South Africa (to our knowledge). This has a large impact on capacity development and knowledge transfer in scarce skills and allows us to be able to use this data to apply for follow-on funding.'
Start Year 2018
 
Description VALIDATE Fellowships - Rachel Tanner 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE aims to encourage Continuing Professional Development (CPD) and career progression amongst its members. As part of this, two VALIDATE Fellowships were awarded in 2018. These VALIDATE Fellowships were open to Early Career Researcher (Associate) Members to fund two years' independent scientific research with the aim of producing initial/pilot data, and a funding and experience track record, that can be used by the Fellow to go on to obtain independent external funding and launch their career as a Principal Investigator and/or Group Leader. After competitive application to the Network Management Board, and subsequent interviews, the two fellowships were awarded to: 1. Dr Jomien Mouton, Stellenbosch University South Africa, "Identification of latency associated antigens and biosignatures associated with Mycobacterium tuberculosis" 2. Dr Rachel Tanner, University of Oxford, UK "Characterising the BCG-induced antibody response to inform the design of improved vaccines against M.tuberculosis, M.leprae and M.bovis" We are proud that both awardees happen to be female, and one is also LMIC. Both Jomien and Rachel joined our Network Management Board for the duration of their Fellowships, giving them experience at this senior level and in reviewing grant and membership applications made to VALIDATE, and giving VALIDATE valuable ECR input to our NMB.
Collaborator Contribution Fellowship Aims This project aimed to characterise the antibody response to BCG vaccination by identifying mycobacterial proteins that are potent inducers of protective antibodies, and testing these proteins as potential vaccine candidates. The findings could inform the design of new, efficacious vaccines against M.tb, M.leprae and M.bovis that may benefit from targeting humoral as well as cell- mediated immunity. Serum samples were selected from n=10 humans, n=10 non-human primates (NHP) and n=10 cattle at pre- and post-BCG vaccination time-points. In collaboration with Antigen Discovery Inc. (ADI), we employed a comprehensive, high-throughput and unbiased approach to antigen discovery, applying protein microarrays spanning the entire proteome of M.tb to identify targets recognised by BCG vaccine-induced antibodies. A large number of proteins showed increased reactivity following BCG vaccination across species, with a higher magnitude of response to BCG in NHPs compared with humans (possibly associated with the lower baseline reactivity resulting in less blocking/masking effect). Interestingly, many top- ranked proteins differed by species and by route of BCG administration in NHP, with higher reactivity in the intravenous (IV) vaccinated compared with intradermal (ID) vaccinated NHP, and in particular the two most protected animals. This suggests that route of vaccination alters target antigen repertoire and indicates potential associations between IgG antigen specificity and improved protection from in vivo M.tb challenge. 14 proteins of interest were selected based on i) higher array reactivity in animals showing superior protection from mycobacterial challenge in vivo, ii) ranking of reactivity in the array based on magnitude of response increase between pre- and post-vaccination time-points, iii) high ranking across more than one species, iv) high degree of homology across M.tb and one or more of M.bovis and M.leprae, v) low homology to human proteins, and vi) surface or secreted protein available for antibody recognition in vivo. 8 of these were successfully synthesised in collaboration with Deepa Paliwal and Ian Jones at the University of Reading, and 7 were taken forward to testing in mice. Antigens were tested in a protein + adjuvant formulation with 3 homologous vaccinations at 2 weekly intervals. Spleen and serum were collected and the ex vivo mycobacterial growth inhibition assay (MGIA) conducted as a high-throughput surrogate of protective efficacy. 3 of the proteins conferred significantly improved ability to control mycobacterial growth in the MGIA following correction for multiple comparisons and will be progressed to further in vivo efficacy studies. The pandemic, associated lockdown and lack of lab capacity since has had a significant impact on this work in 2020 and Rachel was granted a no-cost extension to compensate for this and enable her to finish her research.
Impact New Collaborations • Collaborated with Antigen Discovery Inc on this project, which is a new industry collaboration, and who have subsequently been involved in her follow-up grant applications • Sally Sharpe (PHE, UK - VALIDATE member) • Martin Vordermeier (APHA, UK - VALIDATE member) • Bernardo Villarreal-Ramos (University of Aberystwyth - VALIDATE member) • Ian Jones and Deepa Paliwal (University of Reading & VALIDATE members) • In discussions to set up new collaborations with researchers at Albert Einstein College of Medicine in New York and Leiden University Medical Centre in the Netherlands to explore the functional and biophysical characteristics of the antibody responses induced by antigens identified during her fellowship. Publications • Review on the humoral immune response to BCG vaccination as a basis to this project: Tanner R, Villarreal-Ramos B, Vordermeier M, McShane H. "The humoral immune response to BCG vaccination." Front Immunol. 2019; 10:1317 • Preprint of a primary data paper on the humoral response to BCG vaccination as senior author: "Induction of specific antibodies, IgG-secreting plasmablasts and memory B cells following BCG vaccination." BioRxiv, 2021. • In writing: main data paper from this work • In writing: a VALIDATE review entitled 'Shared challenges to the control of complex intracellular neglected pathogens'. Funding • Submitted a VALIDATE pump-priming application with VALIDATE member Dr Hao Li who hopes to conduct some of his research with Rachel in Oxford in March 2022 (successful - £57,500). • Submitted an R01 NIH grant as co-applicant with VALIDATE Fellow, Dr Jomien Mouton (Stellenbosch University) entitled, "Evaluation of the interplay between host cells and mycobacteria in drug-resistant tuberculosis (DR-TB) patients from the Eastern Cape, South Africa" in July 2021. • Submitted a Dorothy Temple Cross Tuberculosis International collaboration grant as co-PI in collaboration with VALIDATE Fellow, Dr Jomien Mouton (Stellenbosch University) entitled, "Combining expertise in mycobacterial killing assays to evaluate functional immune and treatment responses in drug-resistant tuberculosis (DR-TB) patients" in October 2020. Although this was not funded, Jomien and Rachel aim to incorporate feedback and resubmit in 2021 and/or submit our application to another funding call. • As a member of VALIDATE Jomien and Rachel also participated in a larger European COST funding application that included 97 VALIDATE members from 49 countries (unsuccessful). • Building on the preliminary data generated and experience gained during this fellowship, Rachel recently submitted an application for a £1.5m UKRI Future Leaders Fellowship, as well as for an ERC Starting Grant and a Wellcome Trust Sir Henry Dale Fellowship. • In January 2021, Rachel was awarded a £5,000 grant from the University of Oxford John Fell Fund to rebuild research momentum after COVID-19 and support the completion of the murine studies. • 2021 received a £21,500 grant from the Health Research Bridging Salary Scheme to support her salary for the period between completing this fellowship and hearing the outcome of her pending follow-on grant applications. • Shortlisted for an Oxford NIHR BRC Senior Fellowship but was unsuccessful following interview. • Co-Investigator on a successful grant application from the Songklanagarind Hospital Research Fund, Thailand. • Received a BSI Travel award (£1000) to present this project at the 'Tuberculosis: Immunity and Immune Evasion' Keystone Symposium in Santa Fe, USA in January 2020 Presentations • 6th Global Forum on TB Vaccines in Toulouse France (this meeting became virtual due to COVID) in February 2022: Project abstract was selected for an oral presentation. • Keystone Symposium in Santa Fe, USA in January 2020: project abstract was selected for an oral presentation and a poster. Presenting at such a large international forum was an excellent opportunity for Rachel to reach a wider audience and make new connections, and she met several new contacts who have relevant sample sets to offer or are otherwise interested in collaborating on antibody-related projects. • Presented the protein array findings at the TBScience2020 meeting as part of the 51st Union World Conference on Lung Health. • Received a Collaboration for TB Vaccine Discovery (CTVD) Early Career Investigator Award to present a poster of this work at the CTVD annual meeting in Seattle in June 2020 (this meeting became virtual due to COVID). • Presented preliminary data at the International BCG Symposium (Lille, Dec 2018) and the TBVAC2020 Annual Meeting (Switzerland, Feb 2019), which also opened up new networks and discussions on the project. • Presented her research project to VALIDATE members at the 2018 and 2019 VALIDATE Annual Meetings, as a poster at the VALIDATE-BSI 2019 Conference, and as a virtual seminar "Developing a better vaccine against TB" in February 2019. Continuing professional development In 2018, Rachel joined the Acid Fast Club committee and the Editorial Board of Scientific Reports, and in 2022 was invited to join the Editorial Board of Frontiers in Immunology. In 2019 she joined the British Society of Immunology Vaccine Affinity Group (BSI-VAG) committee, which came about as a result of networking during the VALIDATE-BSI annual meeting. In Jan. 2020, she was elected a Research Fellow of Wolfson College, Oxford, and presented work from this fellowship as part of the interview process. She is currently primary supervisor to a DPhil student and has examined four transfer/confirmation of status vivas and one final DPhil viva in the past 2 years. She has been asked to be external examiner for a further final DPhil viva. In 2021, she completed training on Mental Health Awareness (MFHA England); Equality, Diversity and Inclusion; Safeguarding; and Supporting Student Mental Health. She holds a Lectureship position at Wadham College and is Access and Outreach Officer for the Institute of Human Sciences. In 2019, she completed the Oxford Learning Institute Enhancing Teaching Program (ETP) with a Special Commendation, and a SEDA-PDF Learning, Teaching and Assessing Award (aligned against the UKPSF). Secondments/placements/internships From April to Nov. 2020 Rachel was partially seconded to the Oxford COVID-19 vaccine trial group, testing the safety and efficacy of the ChAdOx1 nCoV-19 vaccine. This has resulted in co-authorship on 59 publications. Outreach In 2019 Rachel contributed to a VALIDATE blog for World Leprosy Day, was recorded by Film Oxford for a film entitled 'Women in Vaccine Research' for International Women's Day, and volunteered at Oxford Open Doors, discussing her research with the public, as well as the Oxford Science and Ideas Festival and 'Uncultured: Late night at the Museum of Natural History'. She also provides regular outreach talks for the university during term time at Wadham College and as Access Officer for the Institute of Human Sciences, giving school talks to groups from over 20 different secondary schools. She also gave a talk as part of the Oxford University 'Meeting Minds' alumni event. In 2020 Rachel filmed clips from a day in the laboratory for the YouTube 'Life in a Day' movie directed by Ridley Scott and Kevin McDonald which premiered at Sundance Film Festival; her submission was featured as part of the press release. She took part in the BSI 'A day in the life of a vaccine researcher' blog, and was interviewed for a 500 Women Scientists article. Rachel was contacted by a school student in California who interviewed her for a presentation on "Scientists that don't fit the conventional mold", and in 2020 took part in 'I'm a Scientist, Stay at Home!' to help school pupils stay connected with STEM during the lockdown. She gave a 'VALIDATE for Schools' virtual talk in December 2020, discussing her research and career in science, to over 200 school children across the UK, and also contributed a 'Useful Resources' section to the VALIDATE Outreach Guide. She published a paper for children on BCG vaccination in Frontiers for Young Minds as part of the VALIDATE BCG100 celebrations. Rachel was invited as a panellist to talk about her work and experience as a female scientist for the 2020 Women of the Future Awards and the 2021 Oxford Women in Engineering, Science and Technology (OxWEST) Annual Symposium. In 2021, Rachel gave a talk at the UNIQ Summer School and the University of Oxford 'Meeting Minds' alumni event. In 2022, she was invited as a panellist for a Wadham College 'Towards Vaccine Equity' event and to chair the 'Two years of COVID-19 - did we follow the science?' event at UCL. Rachel was featured in the 'Wonderful Women' campaign by Wolfson College in celebration of International Women's Day 2022. Awards Rachel was awarded the 2019 Women of the Future Award for Science, partly in recognition of her VALIDATE Fellowship research. In 2021, she was elected a Research Fellow of Wolfson College, Oxford, was awarded a divisional Teaching Excellence Award, the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC) International Global 3Rs Award, and a Justice, Equality, Diversity and Inclusion (JEDI) award from the Life Sciences Editors Foundation. Career Development In 2022 Rachel gained an Assistant Professor position at University of Oxford, to start her own research group.
Start Year 2018
 
Description VALIDATE Pump-priming project P003 
Organisation Federal University of Bahia
Country Brazil 
Sector Academic/University 
PI Contribution VALIDATE funded Pump-priming project P003
Collaborator Contribution The effect of BCG vaccination in immune responses against visceral leishmaniasis in a natural (canine) model of infection Led by Dr Javier Salguero Bodes (Public Heath England), with Dr Isadora dos Santos Lima (FIOCRUZ), Assoc Prof Daniela Farias Larangeira (UFBA), Dr Deborah Fraga (FIOCRUZ), Dr Geraldo Sá Oliveira (FIOCRUZ), Dr Washington dos-Santos (FIOCRUZ), and Prof Luiz Freitas (FIOCRUZ). Project Aims Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis due to frequent complications and the risk of developing into untreated death. It is a zoonosis with high prevalence and wide distribution by the world. In urban areas, the dog is considered the main reservoir of VL due to its close relationship with humans. Canine disease is also considered valuable for the understanding of human disease, since the clinical presentation in both species show similarities. Several drugs have been used in the treatment of VL; however, some of them are not recommended by the world health organization for use in veterinary medicine, in order to avoid the parasite's resistance to active principles. Immunotherapy involves the use of biological substances or molecules to modulate immune responses in order to achieve prophylactic and / or therapeutic success. Immunotherapy with or without chemotherapy has been used for the treatment of leishmaniasis. Several studies have described that the use of immunotherapy helps to reduce the clinical signs, the dose of drugs and the time of the treatment. The BCG vaccine is widely used to prevent tuberculosis and originates from attenuated strains of Mycobacterium bovis. The main mechanism of action of BCG induced protection has been described as mediated by Th-1 cells. The BCG vaccine has been studied as a possible immunotherapeutic for the control of leishmaniasis. Studies with murine models have shown that the use of BCG associated with conventional treatment helps to reduce the parasite burden, the severity of clinical manifestations, and helps increasing the resistance of macrophages to infection, also increasing the capacity of these cells to kill the parasite. The present study aims to evaluate the effects of BCG vaccination administration on the clinical presentation and parasite load of naturally infected dogs from an area endemic for visceral leishmaniasis. Project Outcomes Clinical signs associated with canine visceral leishmaniasis were present in all of the dogs during the follow up process. There is a change in the clinical course after tratement, but with no statistically significant difference between the groups. Similarly, clinical pathology outcomes are different between groups but with no statistical significance. Regarding the histological changes observed in the liver, the most frequent alterations were inflammation in the portal spaces, Kupffer cells hyperplasia and hypertrophy, inflammatory infiltrate of lymphocytic mononuclear cells within the sinusoids and the formation of small aggregates. The inflammation in the portal spaces was composed of predominantly lymphoplasmocytic infiltrate. The granulomas in the sinusoids were composed of macrophagic cells, sometimes parasitized, lymphocytes and, in some cases, plasma cells. Subsequent analysis of the collected samples include immunohistochemical staining for cell phenotyping, in situ hybridization to assess cytokine production, and determination of the humoral immune profile of anti-Leishmania antibodies.
Impact New Collaborations: We started a partnership with LAIPHE - Parasite-Host Interaction and Epidemiology Laboratory (at FIOCRUZ) to carry out real time PCR tests. In addition, we started a collaboration with Dr Alexandre Reis, the Federal University of Ouro Preto to determine the immunoglobulin profile in the serum of dogs included in the project. Publications: At least two scientific publications will be produced using the results obtained in this project, in relation to a) the local immune response within the granulomas in VL during the L. infantum infection and after BCG vaccination, and b) the changes in the peripheral immune responses (both in preparation). Grants/follow-on funding: The project lead was awarded a grant (Spanish Ministry of Science and Technology; value of £90k; 2019-2022) with collaborators in ISCIII Madrid (lead: Dr Mercedes Dominguez), to study the interaction of BCG with the immune responses of Leishmania in a mouse model, to expand his results from the natural model of disease (canine) in a more controlled, but less natural environment, including Highly susceptible Balb/c mice, partially resistant C57Bl/6 and KO for C3. Capacity Building: The ECR involved in this project (Dr Isadora Lima) received training in new techniques, such as Laser Capture Microdissection, RNAscope (placement at PHE and UoSurrey) and clinical techniques (at the School of Veterinary Medicine, UFBA) such as needle liver biopsy in dogs. She also presented the partial results of this project for the VALIDATE members at the 2018 VALIDATE Annual Meeting, raising her profile in this community, and presented a talk "Canine visceral leishmaniasis: the role of tissue-specific immunity in the progression of disease" at the VALIDATE-BSI 2019 Conference. This project served as a transition from recent PhD graduate to postdoc and pathology specialist trainee to the Person holding the PDRA contract. Career Development: Dr Isadora Lima is now a University Lecturer at FIOCRUZ. Data sets: The data set obtained so far has been forwarded to the VALIDATE team for sharing with VALIDATE members, and as new data is collected, it will be forwarded to the VALIDATE team. Standardised protocols/SOPs: During the project, we improved the liver biopsy technique using the Menghini type needle, with the aid of ultrasound equipment. We also improved the treatment protocol used in the project, with which we obtained positive results in the clinical improvement of dogs. All protocols /SOPs developed during the project will be made available. Other: Results from this project were also presented to VALIDATE members at the 2019 Annual Meeting.
Start Year 2017
 
Description VALIDATE Pump-priming project P003 
Organisation Oswaldo Cruz Foundation (Fiocruz)
Country Brazil 
Sector Public 
PI Contribution VALIDATE funded Pump-priming project P003
Collaborator Contribution The effect of BCG vaccination in immune responses against visceral leishmaniasis in a natural (canine) model of infection Led by Dr Javier Salguero Bodes (Public Heath England), with Dr Isadora dos Santos Lima (FIOCRUZ), Assoc Prof Daniela Farias Larangeira (UFBA), Dr Deborah Fraga (FIOCRUZ), Dr Geraldo Sá Oliveira (FIOCRUZ), Dr Washington dos-Santos (FIOCRUZ), and Prof Luiz Freitas (FIOCRUZ). Project Aims Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis due to frequent complications and the risk of developing into untreated death. It is a zoonosis with high prevalence and wide distribution by the world. In urban areas, the dog is considered the main reservoir of VL due to its close relationship with humans. Canine disease is also considered valuable for the understanding of human disease, since the clinical presentation in both species show similarities. Several drugs have been used in the treatment of VL; however, some of them are not recommended by the world health organization for use in veterinary medicine, in order to avoid the parasite's resistance to active principles. Immunotherapy involves the use of biological substances or molecules to modulate immune responses in order to achieve prophylactic and / or therapeutic success. Immunotherapy with or without chemotherapy has been used for the treatment of leishmaniasis. Several studies have described that the use of immunotherapy helps to reduce the clinical signs, the dose of drugs and the time of the treatment. The BCG vaccine is widely used to prevent tuberculosis and originates from attenuated strains of Mycobacterium bovis. The main mechanism of action of BCG induced protection has been described as mediated by Th-1 cells. The BCG vaccine has been studied as a possible immunotherapeutic for the control of leishmaniasis. Studies with murine models have shown that the use of BCG associated with conventional treatment helps to reduce the parasite burden, the severity of clinical manifestations, and helps increasing the resistance of macrophages to infection, also increasing the capacity of these cells to kill the parasite. The present study aims to evaluate the effects of BCG vaccination administration on the clinical presentation and parasite load of naturally infected dogs from an area endemic for visceral leishmaniasis. Project Outcomes Clinical signs associated with canine visceral leishmaniasis were present in all of the dogs during the follow up process. There is a change in the clinical course after tratement, but with no statistically significant difference between the groups. Similarly, clinical pathology outcomes are different between groups but with no statistical significance. Regarding the histological changes observed in the liver, the most frequent alterations were inflammation in the portal spaces, Kupffer cells hyperplasia and hypertrophy, inflammatory infiltrate of lymphocytic mononuclear cells within the sinusoids and the formation of small aggregates. The inflammation in the portal spaces was composed of predominantly lymphoplasmocytic infiltrate. The granulomas in the sinusoids were composed of macrophagic cells, sometimes parasitized, lymphocytes and, in some cases, plasma cells. Subsequent analysis of the collected samples include immunohistochemical staining for cell phenotyping, in situ hybridization to assess cytokine production, and determination of the humoral immune profile of anti-Leishmania antibodies.
Impact New Collaborations: We started a partnership with LAIPHE - Parasite-Host Interaction and Epidemiology Laboratory (at FIOCRUZ) to carry out real time PCR tests. In addition, we started a collaboration with Dr Alexandre Reis, the Federal University of Ouro Preto to determine the immunoglobulin profile in the serum of dogs included in the project. Publications: At least two scientific publications will be produced using the results obtained in this project, in relation to a) the local immune response within the granulomas in VL during the L. infantum infection and after BCG vaccination, and b) the changes in the peripheral immune responses (both in preparation). Grants/follow-on funding: The project lead was awarded a grant (Spanish Ministry of Science and Technology; value of £90k; 2019-2022) with collaborators in ISCIII Madrid (lead: Dr Mercedes Dominguez), to study the interaction of BCG with the immune responses of Leishmania in a mouse model, to expand his results from the natural model of disease (canine) in a more controlled, but less natural environment, including Highly susceptible Balb/c mice, partially resistant C57Bl/6 and KO for C3. Capacity Building: The ECR involved in this project (Dr Isadora Lima) received training in new techniques, such as Laser Capture Microdissection, RNAscope (placement at PHE and UoSurrey) and clinical techniques (at the School of Veterinary Medicine, UFBA) such as needle liver biopsy in dogs. She also presented the partial results of this project for the VALIDATE members at the 2018 VALIDATE Annual Meeting, raising her profile in this community, and presented a talk "Canine visceral leishmaniasis: the role of tissue-specific immunity in the progression of disease" at the VALIDATE-BSI 2019 Conference. This project served as a transition from recent PhD graduate to postdoc and pathology specialist trainee to the Person holding the PDRA contract. Career Development: Dr Isadora Lima is now a University Lecturer at FIOCRUZ. Data sets: The data set obtained so far has been forwarded to the VALIDATE team for sharing with VALIDATE members, and as new data is collected, it will be forwarded to the VALIDATE team. Standardised protocols/SOPs: During the project, we improved the liver biopsy technique using the Menghini type needle, with the aid of ultrasound equipment. We also improved the treatment protocol used in the project, with which we obtained positive results in the clinical improvement of dogs. All protocols /SOPs developed during the project will be made available. Other: Results from this project were also presented to VALIDATE members at the 2019 Annual Meeting.
Start Year 2017
 
Description VALIDATE Pump-priming project P003 
Organisation UK Health Security Agency
Country United Kingdom 
Sector Public 
PI Contribution VALIDATE funded Pump-priming project P003
Collaborator Contribution The effect of BCG vaccination in immune responses against visceral leishmaniasis in a natural (canine) model of infection Led by Dr Javier Salguero Bodes (Public Heath England), with Dr Isadora dos Santos Lima (FIOCRUZ), Assoc Prof Daniela Farias Larangeira (UFBA), Dr Deborah Fraga (FIOCRUZ), Dr Geraldo Sá Oliveira (FIOCRUZ), Dr Washington dos-Santos (FIOCRUZ), and Prof Luiz Freitas (FIOCRUZ). Project Aims Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis due to frequent complications and the risk of developing into untreated death. It is a zoonosis with high prevalence and wide distribution by the world. In urban areas, the dog is considered the main reservoir of VL due to its close relationship with humans. Canine disease is also considered valuable for the understanding of human disease, since the clinical presentation in both species show similarities. Several drugs have been used in the treatment of VL; however, some of them are not recommended by the world health organization for use in veterinary medicine, in order to avoid the parasite's resistance to active principles. Immunotherapy involves the use of biological substances or molecules to modulate immune responses in order to achieve prophylactic and / or therapeutic success. Immunotherapy with or without chemotherapy has been used for the treatment of leishmaniasis. Several studies have described that the use of immunotherapy helps to reduce the clinical signs, the dose of drugs and the time of the treatment. The BCG vaccine is widely used to prevent tuberculosis and originates from attenuated strains of Mycobacterium bovis. The main mechanism of action of BCG induced protection has been described as mediated by Th-1 cells. The BCG vaccine has been studied as a possible immunotherapeutic for the control of leishmaniasis. Studies with murine models have shown that the use of BCG associated with conventional treatment helps to reduce the parasite burden, the severity of clinical manifestations, and helps increasing the resistance of macrophages to infection, also increasing the capacity of these cells to kill the parasite. The present study aims to evaluate the effects of BCG vaccination administration on the clinical presentation and parasite load of naturally infected dogs from an area endemic for visceral leishmaniasis. Project Outcomes Clinical signs associated with canine visceral leishmaniasis were present in all of the dogs during the follow up process. There is a change in the clinical course after tratement, but with no statistically significant difference between the groups. Similarly, clinical pathology outcomes are different between groups but with no statistical significance. Regarding the histological changes observed in the liver, the most frequent alterations were inflammation in the portal spaces, Kupffer cells hyperplasia and hypertrophy, inflammatory infiltrate of lymphocytic mononuclear cells within the sinusoids and the formation of small aggregates. The inflammation in the portal spaces was composed of predominantly lymphoplasmocytic infiltrate. The granulomas in the sinusoids were composed of macrophagic cells, sometimes parasitized, lymphocytes and, in some cases, plasma cells. Subsequent analysis of the collected samples include immunohistochemical staining for cell phenotyping, in situ hybridization to assess cytokine production, and determination of the humoral immune profile of anti-Leishmania antibodies.
Impact New Collaborations: We started a partnership with LAIPHE - Parasite-Host Interaction and Epidemiology Laboratory (at FIOCRUZ) to carry out real time PCR tests. In addition, we started a collaboration with Dr Alexandre Reis, the Federal University of Ouro Preto to determine the immunoglobulin profile in the serum of dogs included in the project. Publications: At least two scientific publications will be produced using the results obtained in this project, in relation to a) the local immune response within the granulomas in VL during the L. infantum infection and after BCG vaccination, and b) the changes in the peripheral immune responses (both in preparation). Grants/follow-on funding: The project lead was awarded a grant (Spanish Ministry of Science and Technology; value of £90k; 2019-2022) with collaborators in ISCIII Madrid (lead: Dr Mercedes Dominguez), to study the interaction of BCG with the immune responses of Leishmania in a mouse model, to expand his results from the natural model of disease (canine) in a more controlled, but less natural environment, including Highly susceptible Balb/c mice, partially resistant C57Bl/6 and KO for C3. Capacity Building: The ECR involved in this project (Dr Isadora Lima) received training in new techniques, such as Laser Capture Microdissection, RNAscope (placement at PHE and UoSurrey) and clinical techniques (at the School of Veterinary Medicine, UFBA) such as needle liver biopsy in dogs. She also presented the partial results of this project for the VALIDATE members at the 2018 VALIDATE Annual Meeting, raising her profile in this community, and presented a talk "Canine visceral leishmaniasis: the role of tissue-specific immunity in the progression of disease" at the VALIDATE-BSI 2019 Conference. This project served as a transition from recent PhD graduate to postdoc and pathology specialist trainee to the Person holding the PDRA contract. Career Development: Dr Isadora Lima is now a University Lecturer at FIOCRUZ. Data sets: The data set obtained so far has been forwarded to the VALIDATE team for sharing with VALIDATE members, and as new data is collected, it will be forwarded to the VALIDATE team. Standardised protocols/SOPs: During the project, we improved the liver biopsy technique using the Menghini type needle, with the aid of ultrasound equipment. We also improved the treatment protocol used in the project, with which we obtained positive results in the clinical improvement of dogs. All protocols /SOPs developed during the project will be made available. Other: Results from this project were also presented to VALIDATE members at the 2019 Annual Meeting.
Start Year 2017
 
Description VALIDATE Pump-priming project P004 
Organisation Brunel University London
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution How do functional and metabolic characteristics of trained monocytes affect their anti-bacterial activity? Led by Asst Prof Steven Smith (LSHTM (now Brunel University)), with Dr Javier Sanchez-Garcia (Instituto Politécnico Nacional), Prof Jo Prior (dstl), and Prof Gregory Bancroft (LSHTM) Project Aims The human immune response has two components, the innate and the adaptive responses. This project will investigate the potential of the innate response to contribute to protection against tuberculosis (TB) and melioidosis. TB remains a major global problem being one of the world's leading causes of death from infectious disease. In addition to better drugs and better means of diagnosing TB, we also need a better vaccine. The current vaccine, BCG, is only partially effective and an improved version is needed. However, many novel TB vaccine candidates focus on the adaptive immune response involving T-lymphocytes as this is where immune memory is found which is essential for vaccine-induced protection. We think that an optimal innate response is also essential. It has recently been discovered that the innate response can be "trained" by some vaccines to respond better to later infections. In this project, we aim to investigate this training effect on the innate response to a) determine how best to train innate cells to prevent the growth of the causative organisms of TB and melioidosis; b) characterise these trained innate cells to find which immune molecules are important for their protective effect and how the metabolism (or biochemical energy production pathways) of these cells affects the trained response and c) to investigate why innate cells from different people do not respond in the same way to training - to do this we will look at differences in the DNA of immune cells (termed epigenetic marks) which cause these cells to produce different immune molecules when exposed to the same microorganism. The data produced by this project should help us understand better how different components of the immune response contribute to protection against TB and melioidosis and should aid the design of better vaccination regimes for bacterial diseases. Project Outcomes Tuberculosis remains the deadliest infectious disease worldwide and thus, any effort to understand the biological basis of disease progression as well as to provide the means to boost anti-Mycobacterium tuberculosis immune responses should be considered, as it might have a positive impact on overcoming this human health burden. Innate immune training offers an interesting theoretical framework for the understanding of tuberculosis, since monocyte/macrophages are the main target cells for infection by M. tuberculosis. Innate immune training allow these cells to respond faster and better to a second antigenic challenge, even if the first and second challenges are qualitatively different. An in vitro model of trained immunity in human monocytes was established in the laboratory based on a previous publication. This involved the isolation of CD14+ monocytes from peripheral blood and co-culture with training stimuli including live BCG, heat-killed BCG or beta-glucan followed by a rest period of 6 days. Training of monocytes was demonstrated by their enhanced ability to produce cytokines (TNF and IL-6) in response to heterologous ligands. Trained monocytes were also cocultured with BCG to determine their capacity to inhibit the growth of mycobacteria which was observed for some, but not all donor monocytes. Monocyte aliquots were stored for analysis of DNA methylation patterns. Recent developments in the field show that, in some instances, innate immune training correlates with intracellular fumarate accumulation. Of note, fumarate-stimulation of monocyte/macrophages induces the functional and epigenetic signatures of innate immune training. As part of this project we aimed to characterize the ''mitochondrial signature'' of this training, as well as to explore some other monocyte/macrophage functions relevant for the control of mycobacterial infection. Human peripheral blood monocytes were first stimulated with monomethyl fumarate and then with LPS or BCG, and mitochondrial dynamics; mitochondrial membrane potential was followed for 3 h post-stimulation and mitochondrial calcium uptake followed for 15min after the addition of BCG. These three functional activities of mitochondria were increased in fumarate-treated monocytes as compared to un-treated cells. Fumarate-stimulated monocyte/macrophages (plus the appropriate controls) were subjected to transmission electron microscopy in order to analyse the mitochondrial cristae. In addition, RAW 264.7 cells (a murine macrophage cell line) were stimulated with monomethyl fumarate in order to study mitochondria and then mitochondrial proteins. Anti-phosphoserine Western blot analyses showed a different pattern of serine-phosphorylated mitochondrial proteins in cells stimulated with a fumarate as compared to un-stimulated cells. All these mitochondrial functional parameters contribute to define the ''mitochondrial signature'' of innate training. As for the function of trained versus non-trained monocytes, the ability of these cells to process antigen was explored. Results showed that fumarate-treated cells have an increased capacity for antigen processing. Taken together, these results provide evidence of a distinctive mitochondrial function (''mitochondrial signature'') in trained monocytes and also show that trained monocytes process exogenous antigens more efficiently that un-trained monocytes. Such a ''mitochondrial signature'' may identify one way to boost anti-mycobacterial immune responses, maintain innate immune training, and understand the basis of mycobacterial dormancy and reactivation.
Impact - This was a new collaboration of the partners with dstl. - New collaboration with the group of Prof Michael Duchen (UCL; expert on mitochondrial calcium) and the group of Carina Kern (UCL; working on C.elegans). - Establishment of scientific collaboration between the groups of LSHTM and ENCB-IPN (Mexico), particularly through Prof Sanchez-Garcia's sabbatical visit to LSHTM. - During the 2018 VALIDATE Annual Meeting this team established a new collaboration with Dr Barbara Kronsteiner-Dobramysl (University of Oxford), leading to a successfully funded new pump-priming project "Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy" (P020). - Butkeviciute E, Jones CE, Smith SG. Heterologous effects of infant BCG vaccination: potential mechanisms of immunity. Future Microbiol. 2018 Aug; 13:1193-1208. - C Angélica Pérez-Hernández et al 2020. Mitochondrial signature in human monocytes and resistance to infection in C.elegans during fumarate-induced innate immune training. Frontiers in Immunology 11: 1715 - In 2020, the work and partnerships established during this project led to the submission of a Newton Fund Institutional Links grant application for which Dr Smith and Prof Sanchez-Garcia were co-applicants. The outcome is still pending for this application but if successful would lead to the strengthening of the partnership established during this VALIDATE project, in particular to the benefit of the Division of Biosciences at Brunel University as members of this division are co-applicants. - In 2020 Dr Steven Smith was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, his application for which was strengthen by having received VALIDATE funding for a project in his area of interest, as well as by the VALIDATE network overall and the collaborations it has been possible to develop. - Prof Javier Sanchez-Garcia states 'The VALIDATE impact on my career is not small. This VALIDATE grant allowed me to spend sabbatical leave at LSHTM, hosted by Professor Hazel Dockrell. While in the UK, I had the opportunity to attend the 2nd annual VALIDATE meeting where I met Dr Barbara Kronsteiner-Dobramysl, who shares our research interest on immunometabolism, as a result we all presented a new research proposal, which luckily received VALIDATE funding. In the same meeting I met some other colleagues and it is likely that in the near future we will establish new collaborations. While in professor Dockrell´s lab, carrying on the experiments for this VALIDATE project, I strengthened collaboration with the group of Professor Michael Duchen at UCL and, while doing some experiments at UCL, I met other colleagues from a different UCL department, soon and quite naturally our respective works merged. In addition, a completely unexpected outcome of all this is that a new experimental animal model for the study of immunometabolism is now available in our lab in Mexico.' - 2022: Dr Steven Smith co-applicant on a proposal led by Dr Luciana Balboa (VALIDATE member, CONICET, Argentina) for a VALIDATE Pump-priming grant "Pro-Solving Lipids: New Players that regulate Local Protection against Mycobacterium tuberculosis". This came about as Dr Balboa was aware of the work done on trained immunity responses in macrophages as part of the P004 project.
Start Year 2017
 
Description VALIDATE Pump-priming project P004 
Organisation Defence Science & Technology Laboratory (DSTL)
Country United Kingdom 
Sector Public 
PI Contribution VALIDATE funded project
Collaborator Contribution How do functional and metabolic characteristics of trained monocytes affect their anti-bacterial activity? Led by Asst Prof Steven Smith (LSHTM (now Brunel University)), with Dr Javier Sanchez-Garcia (Instituto Politécnico Nacional), Prof Jo Prior (dstl), and Prof Gregory Bancroft (LSHTM) Project Aims The human immune response has two components, the innate and the adaptive responses. This project will investigate the potential of the innate response to contribute to protection against tuberculosis (TB) and melioidosis. TB remains a major global problem being one of the world's leading causes of death from infectious disease. In addition to better drugs and better means of diagnosing TB, we also need a better vaccine. The current vaccine, BCG, is only partially effective and an improved version is needed. However, many novel TB vaccine candidates focus on the adaptive immune response involving T-lymphocytes as this is where immune memory is found which is essential for vaccine-induced protection. We think that an optimal innate response is also essential. It has recently been discovered that the innate response can be "trained" by some vaccines to respond better to later infections. In this project, we aim to investigate this training effect on the innate response to a) determine how best to train innate cells to prevent the growth of the causative organisms of TB and melioidosis; b) characterise these trained innate cells to find which immune molecules are important for their protective effect and how the metabolism (or biochemical energy production pathways) of these cells affects the trained response and c) to investigate why innate cells from different people do not respond in the same way to training - to do this we will look at differences in the DNA of immune cells (termed epigenetic marks) which cause these cells to produce different immune molecules when exposed to the same microorganism. The data produced by this project should help us understand better how different components of the immune response contribute to protection against TB and melioidosis and should aid the design of better vaccination regimes for bacterial diseases. Project Outcomes Tuberculosis remains the deadliest infectious disease worldwide and thus, any effort to understand the biological basis of disease progression as well as to provide the means to boost anti-Mycobacterium tuberculosis immune responses should be considered, as it might have a positive impact on overcoming this human health burden. Innate immune training offers an interesting theoretical framework for the understanding of tuberculosis, since monocyte/macrophages are the main target cells for infection by M. tuberculosis. Innate immune training allow these cells to respond faster and better to a second antigenic challenge, even if the first and second challenges are qualitatively different. An in vitro model of trained immunity in human monocytes was established in the laboratory based on a previous publication. This involved the isolation of CD14+ monocytes from peripheral blood and co-culture with training stimuli including live BCG, heat-killed BCG or beta-glucan followed by a rest period of 6 days. Training of monocytes was demonstrated by their enhanced ability to produce cytokines (TNF and IL-6) in response to heterologous ligands. Trained monocytes were also cocultured with BCG to determine their capacity to inhibit the growth of mycobacteria which was observed for some, but not all donor monocytes. Monocyte aliquots were stored for analysis of DNA methylation patterns. Recent developments in the field show that, in some instances, innate immune training correlates with intracellular fumarate accumulation. Of note, fumarate-stimulation of monocyte/macrophages induces the functional and epigenetic signatures of innate immune training. As part of this project we aimed to characterize the ''mitochondrial signature'' of this training, as well as to explore some other monocyte/macrophage functions relevant for the control of mycobacterial infection. Human peripheral blood monocytes were first stimulated with monomethyl fumarate and then with LPS or BCG, and mitochondrial dynamics; mitochondrial membrane potential was followed for 3 h post-stimulation and mitochondrial calcium uptake followed for 15min after the addition of BCG. These three functional activities of mitochondria were increased in fumarate-treated monocytes as compared to un-treated cells. Fumarate-stimulated monocyte/macrophages (plus the appropriate controls) were subjected to transmission electron microscopy in order to analyse the mitochondrial cristae. In addition, RAW 264.7 cells (a murine macrophage cell line) were stimulated with monomethyl fumarate in order to study mitochondria and then mitochondrial proteins. Anti-phosphoserine Western blot analyses showed a different pattern of serine-phosphorylated mitochondrial proteins in cells stimulated with a fumarate as compared to un-stimulated cells. All these mitochondrial functional parameters contribute to define the ''mitochondrial signature'' of innate training. As for the function of trained versus non-trained monocytes, the ability of these cells to process antigen was explored. Results showed that fumarate-treated cells have an increased capacity for antigen processing. Taken together, these results provide evidence of a distinctive mitochondrial function (''mitochondrial signature'') in trained monocytes and also show that trained monocytes process exogenous antigens more efficiently that un-trained monocytes. Such a ''mitochondrial signature'' may identify one way to boost anti-mycobacterial immune responses, maintain innate immune training, and understand the basis of mycobacterial dormancy and reactivation.
Impact - This was a new collaboration of the partners with dstl. - New collaboration with the group of Prof Michael Duchen (UCL; expert on mitochondrial calcium) and the group of Carina Kern (UCL; working on C.elegans). - Establishment of scientific collaboration between the groups of LSHTM and ENCB-IPN (Mexico), particularly through Prof Sanchez-Garcia's sabbatical visit to LSHTM. - During the 2018 VALIDATE Annual Meeting this team established a new collaboration with Dr Barbara Kronsteiner-Dobramysl (University of Oxford), leading to a successfully funded new pump-priming project "Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy" (P020). - Butkeviciute E, Jones CE, Smith SG. Heterologous effects of infant BCG vaccination: potential mechanisms of immunity. Future Microbiol. 2018 Aug; 13:1193-1208. - C Angélica Pérez-Hernández et al 2020. Mitochondrial signature in human monocytes and resistance to infection in C.elegans during fumarate-induced innate immune training. Frontiers in Immunology 11: 1715 - In 2020, the work and partnerships established during this project led to the submission of a Newton Fund Institutional Links grant application for which Dr Smith and Prof Sanchez-Garcia were co-applicants. The outcome is still pending for this application but if successful would lead to the strengthening of the partnership established during this VALIDATE project, in particular to the benefit of the Division of Biosciences at Brunel University as members of this division are co-applicants. - In 2020 Dr Steven Smith was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, his application for which was strengthen by having received VALIDATE funding for a project in his area of interest, as well as by the VALIDATE network overall and the collaborations it has been possible to develop. - Prof Javier Sanchez-Garcia states 'The VALIDATE impact on my career is not small. This VALIDATE grant allowed me to spend sabbatical leave at LSHTM, hosted by Professor Hazel Dockrell. While in the UK, I had the opportunity to attend the 2nd annual VALIDATE meeting where I met Dr Barbara Kronsteiner-Dobramysl, who shares our research interest on immunometabolism, as a result we all presented a new research proposal, which luckily received VALIDATE funding. In the same meeting I met some other colleagues and it is likely that in the near future we will establish new collaborations. While in professor Dockrell´s lab, carrying on the experiments for this VALIDATE project, I strengthened collaboration with the group of Professor Michael Duchen at UCL and, while doing some experiments at UCL, I met other colleagues from a different UCL department, soon and quite naturally our respective works merged. In addition, a completely unexpected outcome of all this is that a new experimental animal model for the study of immunometabolism is now available in our lab in Mexico.' - 2022: Dr Steven Smith co-applicant on a proposal led by Dr Luciana Balboa (VALIDATE member, CONICET, Argentina) for a VALIDATE Pump-priming grant "Pro-Solving Lipids: New Players that regulate Local Protection against Mycobacterium tuberculosis". This came about as Dr Balboa was aware of the work done on trained immunity responses in macrophages as part of the P004 project.
Start Year 2017
 
Description VALIDATE Pump-priming project P004 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution How do functional and metabolic characteristics of trained monocytes affect their anti-bacterial activity? Led by Asst Prof Steven Smith (LSHTM (now Brunel University)), with Dr Javier Sanchez-Garcia (Instituto Politécnico Nacional), Prof Jo Prior (dstl), and Prof Gregory Bancroft (LSHTM) Project Aims The human immune response has two components, the innate and the adaptive responses. This project will investigate the potential of the innate response to contribute to protection against tuberculosis (TB) and melioidosis. TB remains a major global problem being one of the world's leading causes of death from infectious disease. In addition to better drugs and better means of diagnosing TB, we also need a better vaccine. The current vaccine, BCG, is only partially effective and an improved version is needed. However, many novel TB vaccine candidates focus on the adaptive immune response involving T-lymphocytes as this is where immune memory is found which is essential for vaccine-induced protection. We think that an optimal innate response is also essential. It has recently been discovered that the innate response can be "trained" by some vaccines to respond better to later infections. In this project, we aim to investigate this training effect on the innate response to a) determine how best to train innate cells to prevent the growth of the causative organisms of TB and melioidosis; b) characterise these trained innate cells to find which immune molecules are important for their protective effect and how the metabolism (or biochemical energy production pathways) of these cells affects the trained response and c) to investigate why innate cells from different people do not respond in the same way to training - to do this we will look at differences in the DNA of immune cells (termed epigenetic marks) which cause these cells to produce different immune molecules when exposed to the same microorganism. The data produced by this project should help us understand better how different components of the immune response contribute to protection against TB and melioidosis and should aid the design of better vaccination regimes for bacterial diseases. Project Outcomes Tuberculosis remains the deadliest infectious disease worldwide and thus, any effort to understand the biological basis of disease progression as well as to provide the means to boost anti-Mycobacterium tuberculosis immune responses should be considered, as it might have a positive impact on overcoming this human health burden. Innate immune training offers an interesting theoretical framework for the understanding of tuberculosis, since monocyte/macrophages are the main target cells for infection by M. tuberculosis. Innate immune training allow these cells to respond faster and better to a second antigenic challenge, even if the first and second challenges are qualitatively different. An in vitro model of trained immunity in human monocytes was established in the laboratory based on a previous publication. This involved the isolation of CD14+ monocytes from peripheral blood and co-culture with training stimuli including live BCG, heat-killed BCG or beta-glucan followed by a rest period of 6 days. Training of monocytes was demonstrated by their enhanced ability to produce cytokines (TNF and IL-6) in response to heterologous ligands. Trained monocytes were also cocultured with BCG to determine their capacity to inhibit the growth of mycobacteria which was observed for some, but not all donor monocytes. Monocyte aliquots were stored for analysis of DNA methylation patterns. Recent developments in the field show that, in some instances, innate immune training correlates with intracellular fumarate accumulation. Of note, fumarate-stimulation of monocyte/macrophages induces the functional and epigenetic signatures of innate immune training. As part of this project we aimed to characterize the ''mitochondrial signature'' of this training, as well as to explore some other monocyte/macrophage functions relevant for the control of mycobacterial infection. Human peripheral blood monocytes were first stimulated with monomethyl fumarate and then with LPS or BCG, and mitochondrial dynamics; mitochondrial membrane potential was followed for 3 h post-stimulation and mitochondrial calcium uptake followed for 15min after the addition of BCG. These three functional activities of mitochondria were increased in fumarate-treated monocytes as compared to un-treated cells. Fumarate-stimulated monocyte/macrophages (plus the appropriate controls) were subjected to transmission electron microscopy in order to analyse the mitochondrial cristae. In addition, RAW 264.7 cells (a murine macrophage cell line) were stimulated with monomethyl fumarate in order to study mitochondria and then mitochondrial proteins. Anti-phosphoserine Western blot analyses showed a different pattern of serine-phosphorylated mitochondrial proteins in cells stimulated with a fumarate as compared to un-stimulated cells. All these mitochondrial functional parameters contribute to define the ''mitochondrial signature'' of innate training. As for the function of trained versus non-trained monocytes, the ability of these cells to process antigen was explored. Results showed that fumarate-treated cells have an increased capacity for antigen processing. Taken together, these results provide evidence of a distinctive mitochondrial function (''mitochondrial signature'') in trained monocytes and also show that trained monocytes process exogenous antigens more efficiently that un-trained monocytes. Such a ''mitochondrial signature'' may identify one way to boost anti-mycobacterial immune responses, maintain innate immune training, and understand the basis of mycobacterial dormancy and reactivation.
Impact - This was a new collaboration of the partners with dstl. - New collaboration with the group of Prof Michael Duchen (UCL; expert on mitochondrial calcium) and the group of Carina Kern (UCL; working on C.elegans). - Establishment of scientific collaboration between the groups of LSHTM and ENCB-IPN (Mexico), particularly through Prof Sanchez-Garcia's sabbatical visit to LSHTM. - During the 2018 VALIDATE Annual Meeting this team established a new collaboration with Dr Barbara Kronsteiner-Dobramysl (University of Oxford), leading to a successfully funded new pump-priming project "Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy" (P020). - Butkeviciute E, Jones CE, Smith SG. Heterologous effects of infant BCG vaccination: potential mechanisms of immunity. Future Microbiol. 2018 Aug; 13:1193-1208. - C Angélica Pérez-Hernández et al 2020. Mitochondrial signature in human monocytes and resistance to infection in C.elegans during fumarate-induced innate immune training. Frontiers in Immunology 11: 1715 - In 2020, the work and partnerships established during this project led to the submission of a Newton Fund Institutional Links grant application for which Dr Smith and Prof Sanchez-Garcia were co-applicants. The outcome is still pending for this application but if successful would lead to the strengthening of the partnership established during this VALIDATE project, in particular to the benefit of the Division of Biosciences at Brunel University as members of this division are co-applicants. - In 2020 Dr Steven Smith was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, his application for which was strengthen by having received VALIDATE funding for a project in his area of interest, as well as by the VALIDATE network overall and the collaborations it has been possible to develop. - Prof Javier Sanchez-Garcia states 'The VALIDATE impact on my career is not small. This VALIDATE grant allowed me to spend sabbatical leave at LSHTM, hosted by Professor Hazel Dockrell. While in the UK, I had the opportunity to attend the 2nd annual VALIDATE meeting where I met Dr Barbara Kronsteiner-Dobramysl, who shares our research interest on immunometabolism, as a result we all presented a new research proposal, which luckily received VALIDATE funding. In the same meeting I met some other colleagues and it is likely that in the near future we will establish new collaborations. While in professor Dockrell´s lab, carrying on the experiments for this VALIDATE project, I strengthened collaboration with the group of Professor Michael Duchen at UCL and, while doing some experiments at UCL, I met other colleagues from a different UCL department, soon and quite naturally our respective works merged. In addition, a completely unexpected outcome of all this is that a new experimental animal model for the study of immunometabolism is now available in our lab in Mexico.' - 2022: Dr Steven Smith co-applicant on a proposal led by Dr Luciana Balboa (VALIDATE member, CONICET, Argentina) for a VALIDATE Pump-priming grant "Pro-Solving Lipids: New Players that regulate Local Protection against Mycobacterium tuberculosis". This came about as Dr Balboa was aware of the work done on trained immunity responses in macrophages as part of the P004 project.
Start Year 2017
 
Description VALIDATE Pump-priming project P016 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Vaccines to target people with diabetes: characterising the pathways of immune response to M. tuberculosis and B. pseudomallei in people with diabetes compared to non-diabetics Led by Prof Susanna Dunachie (University of Oxford), with Assistant Prof Jacqueline Cliff (LSHTM), and Prof Gregory Bancroft (LSHTM) Project Aims A better vaccine is urgently required for tuberculosis (TB), and there is no vaccine at all for the neglected tropical disease melioidosis. Both diseases are caused by bacteria that live inside cells, and there are shared defence mechanisms. People with diabetes are at increased risk of getting ill with TB (three-fold increased risk) and are twelve time more likely to develop melioidosis than non-diabetics. We therefore need to understand why people with diabetes get ill with these bacteria, and how diabetes prevents the body from clearing the infection. We will combine expertise at the London School of Hygiene and Tropical Medicine, Mahidol- Oxford Tropical Medicine Unit in Bangkok and the University of Oxford, to study the pattern of human immune response to TB and melioidosis in people with and without diabetes. To do this, we will use "transcriptomics" - the big-scale study of the gene readouts from the body's DNA ("transcripts") which are instruction messages for protein manufacture carried as ribonucleic acid (RNA). By measuring the pattern of RNA in a person's blood, we can compare which immune pathways are active in different groups of people. We will identify differences in the pattern of responses in TB patients with and without diabetes, see how they relate to cure from TB, and then compare the results with responses in melioidosis patients with and without diabetes. We will detect the most important protective immune response pathways which are seen in nondiabetic patients but are lacking in people with diabetes. This will allow us to design better vaccines for TB and melioidosis that work well in people with diabetes, and can also help us develop ways to use medicines at the time of vaccination to boost immune responses in people with diabetes. Project Outcomes Diabetes mellitus (DM) currently affects over 425 million people worldwide, of which almost 80% live in low- and middle-income countries (LMICs). The majority of people with diabetes in LMICs are thought to have Type 2 diabetes. Diabetes increases susceptibility to many global infections including Escherichia coli, Staphylococcus aureus and dengue, but the most established susceptibility relationships are for melioidosis and tuberculosis (TB). Melioidosis is a deadly infectious disease caused by the Gram-negative bacterium, Burkholderia pseudomallei (BP). The current estimation of melioidosis cases is 165,000 cases worldwide with 89,000 deaths. Mortality in severe melioidosis cases presenting with sepsis can reach 50% in Thailand. Diabetes is a major risk factor for melioidosis with a 12-fold increased susceptibility to the infection. Tuberculosis (TB) is a major global killer. In 2018, an estimated 10 million people were infected with Mycobacterium tuberculosis and over a million people died from TB. Moreover, DM confers a 3-fold increased susceptibility to TB. Tuberculosis and melioidosis share many key features including an overlap of clinical manifestations, immunopathology, and host immune responses. A previous study in Thailand demonstrated that transcriptomic profiles between tuberculosis and melioidosis were indistinguishable. Host-immune responses to the infections are dysregulated in people with diabetes including killing mechanisms, cell-cell communication, and migration. However, mechanisms involved in the increased susceptibility in DM to intracellular pathogens are to be elucidated. In this project we used RNA sequencing to study transcriptomics in melioidosis and tuberculosis patients in order to characterise the difference in host response to infection for those with and without DM. Differential gene expression analysis between melioidosis patients with (n = 49) and without DM (n = 32) identified two genes CD163L1 (scavenger-receptor cytokine-rich protein) and FAM1932 (a member of the CC chemokine family) were differentially expressed with a false discovery rate < 0.05. However, gene set enrichment analysis using hallmark gene sets showed distinctive profiles in several comparisons. A comparison between melioidosis cases with and without DM showed a number of enriched gene sets dominated by immune, proliferation, and signalling in the DM group only. We identified gene signatures in people with diabetes indicative of endoplasmic reticulum stress and are now confirming this finding in validation experiments in peripheral blood mononuclear cells. Further analysis comparing the impact of diabetes in acute melioidosis with tuberculosis revealed shared transcriptomic signatures of increased inflammatory responses, increased neutrophil degranulation and increased platelet degranulation. However, looking at the whole blood transcriptome we saw opposite effects of diabetes on interferon-gamma signalling, with an increase in acute melioidosis (which may represent non-specific neutrophil activation) and a decrease in tuberculosis. Using an additional sample set from a collaboration with Eoin West at University of Washington, USA, we compared patients who went on to survive melioidosis in Ubon Ratchathani (n = 80) with fatal cases (n = 84) and found dominance in fatal cases for non-specific pro-inflammatory pathways but down-regulation of T cell signalling pathways, especially in those with diabetes. Overall, our data shows that excessive pro-inflammatory immune responses with reduced T cell signalling are associated with poor clinical outcomes, and people with diabetes have an excessive non-specific pro-inflammatory response to both melioidosis and tuberculosis, with increased neutrophil and platelet activation. The massive neutrophil activation in sepsis from acute melioidosis is associated with increased interferon-gamma pathway activation, in contrast to tuberculosis where overall interferon-gamma activity is reduced in diabetes - most likely reflecting the less acute nature of disease from M. tuberculosis. Vaccines targeting T cells for these intracellular pathogens may be especially important to correct the increased susceptibility seen in diabetes.
Impact Collaborations: • This project allowed the building of a collaboration between S. Dunachie and J. Cliff, which we aim to be a foundation for further work. • Further collaborations that this project was a springboard for: 1. Paul Brett and Mary Burtnick, University of Nevada in Reno, USA - planning a Phase 1 vaccine trial of a public health melioidosis vaccine to target people with diabetes 2. Helen McShane and Elena Stylianou, U. Oxford, UK - establishing a mouse model of diabetes for vaccine testing 3. Claire Chewapreecha, Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand - "Dissecting the genetic basis of melioidosis infection" 4. Eoin West, University of Washington, Seattle, USA (extension from previous collaboration on sepsis and Toll-like receptors) - transcriptomic signatures of melioidosis and sepsis 5. Chiranjay Mukhopadhyay, Manipal Academy of Higher Education, India - cellular immunity in TB and melioidosis (which has been awarded a subsequent VALIDATE pump-priming grant, P033) 6. Mohammad Ali, Dhaka Medical College, Bangladesh - metabolism, DM and TB 7. Vivek Jha, George Institute, New Delhi, India - diabetes and infection database research 8. Prof. Chang-Hwa Song, Chungman National University, South Korea - macrophage immunology in TB +/- DM As a result of the VALIDATE networking session in 2018, a team assembled with an interest in developing a diabetic mouse model for testing of immune response to disease and vaccination, and this was a successful pump-priming award from VALIDATE in early 2019 (PI Elena Stylianou, P022 - further details below). Papers: This project influenced the following manuscripts: • Dunachie, S. and P. Chamnan, The double burden of diabetes and global infection in low and middle-income countries. Trans R Soc Trop Med Hyg, 2019. 113(2): p. 56-64. https://doi.org/10.1093/trstmh/try124 • Kronsteiner, B., et al., Diabetes alters immune response patterns to acute melioidosis in humans. Eur J Immunol, 2019. 49(7): p. 1092-1106 https://doi.org/10.1002/eji.201848037 • Eckold, C. et al., Impact of intermediate hyperglycaemia as well as diabetes on immune dysfunction in tuberculosis. Clinical Infectious Diseases 2021 https://doi.org/10.1093/cid/ciaa751 • Eckold et al., Impaired resolution of blood transcriptomes through tuberculosis treatment with diabetes comorbidity MedRxiv 2022 https://doi.org/10.1101/2022.02.07.22269422 • Tomás-Cortázar, J., Bossi, L., Quinn, C., Reynolds, C.J., Butler, D.K., Corcoran, N., . . . McClean, S. BpOmpW Antigen Stimulates the Necessary Protective T-Cell Responses Against Melioidosis. Front. Immunol. 12, 767359-767359 (2021). 10.3389/fimmu.2021.767359. https://pubmed.ncbi.nlm.nih.gov/34966388 • Chewapreecha, C., Pensar, J., Chattagul, S., Pesonen, M., Sangphukieo, A., Boonklang, P., . . . Corander, J. Co-evolutionary Signals Identify Burkholderia pseudomallei Survival Strategies in a Hostile Environment. Mol. Biol. Evol. 39, msab306 (2022). 10.1093/molbev/msab306. https://pubmed.ncbi.nlm.nih.gov/34662416 Two manuscripts are in preparation directly from this work: 1. Rongkard, P., Kronsteiner B., Hantrakunb, V., Teparrukkul, P., Wongsuvanb, G., Limmathurotsakul, D., Lovelace-Macon, L., Day, N.P., Klenerman, P., Chantratita, N., Gharib, S., Dunachie S., West, T.E. Transcriptomic signatures in fatal melioidosis. Manuscript in preparation 2. Rongkard, P., Hantrakunb, V., Teparrukkul, P., Wongsuvanb, G., Limmathurotsakul, D., Lovelace-Macon, L., Day, N.P., Gharib, S., West, T.E, Cliff, J., Klenerman, P., Kronsteiner B., Dunachie S. The impact of type 2 Diabetes on the immune response to melioidosis and tuberculosis. Manuscript in preparation Related funding: 1. 2021-2026 NIHR Professorship in Global Health "Developing a vaccine to prevent death from melioidosis in people with type 2 diabetes mellitus in low- and middle-income countries" (Dunachie PI), £1,951,296 2. 2020-2022 US Defense Threat Reduction Agency award "Optimization of lead melioidosis and glanders vaccine formulations" Prof Paul Brett University of Nevada, Reno PI with sub-contract award to Dunachie, Oxford PI, $733,976.61 USD (£524,269) to Dunachie lab. 3. 2022-2025 MRC Biomedical Catalyst Developmental Pathway Funding Scheme (DPFS) / US Defense Threat reduction Agency award, "Developing a vaccine for Burkholderia pseudomallei - a Phase I Clinical Trial" (Dunachie PI) £3,238,214. 4. 2019-2020 VALIDATE pump-priming project: "Characterising the cellular immunity and metabolic response to Mycobacterium tuberculosis and Burkholderia pseudomallei in Indian patients for vaccine design". (Mukhopadhyay, Manipal Academy of Higher Education, India PI, Dunachie Co-PI) £48,993 (£7,000 to Dunachie lab). 5. 2019-2020 Global Challenges Research Fund (GCRF) "Diabetes and Infection: Developing a network for using real-world datasets to evaluate the relationship between diabetes and infection in low and middle income countries?" (Dunachie PI) £?26,728.50. 6. 2019-2022 Bangladesh Prime Minister's PhD Scholarship for DPhil study at U. Oxford for "Metabolic changes in immune cells of diabetic patients and their impact on immune responses to tuberculosis"(Mohammad Ali, Dhaka Medical College, Bangladesh applicant, Dunachie supervisor) $258,823 USD (£184,874). 7. 2019-2020 MRC VALIDATE "Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis' (Stylianou PI, Dunachie Co-PI) £49,733.91 - £5,500 to Dunachie lab). In addition, S.Dunachie has been PI or Co-Investigator for grants totalling £5.7 million addressing the immune response to SARS-CoV-2 from infection and vaccination. The immunology expertise developed in her melioidosis work was rapidly applicable to SARS-CoV-2 due to similarity of vulnerable patient group (increased age, diabetes, immunocompromise). Capacity building: • This was Prof Susanna Dunachie's first formal funding to look at the relationship between diabetes and infection (although it was already an interest), and has sparked a whole programme of work (see collaborations and funding). She is now leading the world's first clinical trial of a vaccine for melioidosis. • Mr Patpong Rongkard, Mahidol-Oxford Tropical Medicine Research Unit (MORU), Bangkok - came to Oxford to undertake this project, and was able to register for a DPhil "Understanding how type II diabetes increases susceptibility to intracellular infections". This VALIDATE project has enabled him to receive extensive training in bioinformatics for analysis of RNAseq data, all using open-source software in R. He passed his DPhil viva in November 2022 and has now been awarded the DPhil. • Susanna Dunachie is supporting development of immunology research in India and Bangladesh, by supporting a new collaboration with Dr Mukhopadhyay at Manipal Academy of Higher Education, India, and supported the writing of a successful PhD scholarship application by Dr Ali of Dhaka Medical College, who is now undertaking a DPhil at U Oxford. • Dr Lee (LSHTM) was instrumental in writing her first grant application "Building investigator links for studies on the roles of complement C1q and endoplasmicreticulum (ER) stress in macrophage polarisation during Mycobacterium tuberculosis infection for a successfully funded MRC-KHIDI UK-Korea Partnering Award. • Secondments: Mr Rongkard was unable to undertake the planned secondment at U. Washington, Seattle, USA, in May 2020 due to the pandemic. However, using Microsoft Teams we have been able to build a close collaboration and he has used the skills acquired in this project to analyse the raw RNAseq data generated in an independent cohort of melioidosis patients as part of a sepsis study in the same region of Northeast Thailand as our study (see results). • Susanna organised the VALIDATE Melioidosis Vaccine Symposium: Moving from animal models to man on February 26th 2019, with support from the VALIDATE team (see separate section on this Symposium). Outreach: March 2022. Talk to local 6th form college about careers in tropical medicine. 2022 Dunachie S. Vaccines against melioidosis: How far are we? European Melioidosis Congress, Gratz, Austria, May 2022 2022 Rongkard P. T-cell dysfunction is associated with fatality in community-acquired melioidosis 2019 Dunachie S. Diabetes and melioidosis: What do we know, and what can we do? Invited plenary speaker at World Melioidosis Congress, Hanoi, October 2019 This project was presented at the 2018, 2019 and 2021 VALIDATE Annual Meetings. As a result of the VALIDATE networking session in 2018, a team assembled with an interest in developing a diabetic mouse model for testing of immune response to disease and vaccination, and this was a successful pump-priming award from VALIDATE in early 2019 (PI Elena Stylianou, P022). New datasets of RNAseq data in acute melioidosis have been generated from this project and will be available to VALIDATE members. Awards/Recognition: Susanna Dunachie: - Professor of Infectious Diseases - NIHR Global Research Professorship - Medical Fellow, Corpus Christi College, Oxford Product Development We organised a VALIDATE workshop on vaccines for melioidosis in February 2019, and as a result submitted a successful application to MRC's DPFS grant call to undertake the first human Phase I Vaccine Trial of a melioidosis vaccine, in Oxford using Paul Brett and Mary Burtnick's melioidosis vaccine candidate. The subunit vaccine has been developed at University of Nevada, Reno (UNR) and combines the bacterial capsule polysaccharide (CPS) conjugated to the immunogenic diphtheria toxin mutant CRM197, with recombinant B. pseudomallei Hcp1 protein in an Alhydrogel-CpG adjuvant. In a mouse model, the combined vaccine generated high levels of opsonising anti-CPS antibodies, alongside high T-cell responses to Hcp1, and protected 100% of mice against a robust lethal inhalational B. pseudomallei challenge. We plan to develop the vaccine as a public health vaccine for people with diabetes in low- and middle-income countries (LMICs). The group of investigators in UK (Dunachie, McShane, Kronsteiner), USA (Brett, Burtnick) and Thailand (Chantratita), brought together by the MRC VALIDATE Network, are planning a first-in-man study of 36 healthy human volunteer subjects in Oxford, UK, randomised to receive either the CPS-CRM197 alone, Hcp1 alone, or the CPS-CRM197 / Hcp1 combination. This will be followed by 12 subjects aged 40-60 years with Type II diabetes in Oxford receiving the CPS-CRM197 / Hcp1 combination. Endpoints will be safety and immunogenicity (antibody and T cell responses). The team seeks to define the key immune correlates of protection for monitoring vaccine efficacy in subsequent field trials, using functional growth inhibition assays and transcriptomics in the vaccinated population, existing melioidosis patient cohort studies, and animal models in collaboration with UNR. The vaccine is currently completing manufacture at Resilience Corp, Florida, USA (funded by US Defense Threat Reduction Agency) and we will commence the Phase 1 clinical trial in Oxford, UK in early 2023. Next we plan to undertake a Phase 1b study in Thailand (will require future funding). Ultimately we seek to accelerate production of a vaccine for the 280 million people with diabetes and the many with other risk factors for melioidosis including older age, chronic renal disease and immunosuppression who live in melioidosis-endemic regions in 83 countries across the globe.
Start Year 2018
 
Description VALIDATE Pump-priming project P016 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Vaccines to target people with diabetes: characterising the pathways of immune response to M. tuberculosis and B. pseudomallei in people with diabetes compared to non-diabetics Led by Prof Susanna Dunachie (University of Oxford), with Assistant Prof Jacqueline Cliff (LSHTM), and Prof Gregory Bancroft (LSHTM) Project Aims A better vaccine is urgently required for tuberculosis (TB), and there is no vaccine at all for the neglected tropical disease melioidosis. Both diseases are caused by bacteria that live inside cells, and there are shared defence mechanisms. People with diabetes are at increased risk of getting ill with TB (three-fold increased risk) and are twelve time more likely to develop melioidosis than non-diabetics. We therefore need to understand why people with diabetes get ill with these bacteria, and how diabetes prevents the body from clearing the infection. We will combine expertise at the London School of Hygiene and Tropical Medicine, Mahidol- Oxford Tropical Medicine Unit in Bangkok and the University of Oxford, to study the pattern of human immune response to TB and melioidosis in people with and without diabetes. To do this, we will use "transcriptomics" - the big-scale study of the gene readouts from the body's DNA ("transcripts") which are instruction messages for protein manufacture carried as ribonucleic acid (RNA). By measuring the pattern of RNA in a person's blood, we can compare which immune pathways are active in different groups of people. We will identify differences in the pattern of responses in TB patients with and without diabetes, see how they relate to cure from TB, and then compare the results with responses in melioidosis patients with and without diabetes. We will detect the most important protective immune response pathways which are seen in nondiabetic patients but are lacking in people with diabetes. This will allow us to design better vaccines for TB and melioidosis that work well in people with diabetes, and can also help us develop ways to use medicines at the time of vaccination to boost immune responses in people with diabetes. Project Outcomes Diabetes mellitus (DM) currently affects over 425 million people worldwide, of which almost 80% live in low- and middle-income countries (LMICs). The majority of people with diabetes in LMICs are thought to have Type 2 diabetes. Diabetes increases susceptibility to many global infections including Escherichia coli, Staphylococcus aureus and dengue, but the most established susceptibility relationships are for melioidosis and tuberculosis (TB). Melioidosis is a deadly infectious disease caused by the Gram-negative bacterium, Burkholderia pseudomallei (BP). The current estimation of melioidosis cases is 165,000 cases worldwide with 89,000 deaths. Mortality in severe melioidosis cases presenting with sepsis can reach 50% in Thailand. Diabetes is a major risk factor for melioidosis with a 12-fold increased susceptibility to the infection. Tuberculosis (TB) is a major global killer. In 2018, an estimated 10 million people were infected with Mycobacterium tuberculosis and over a million people died from TB. Moreover, DM confers a 3-fold increased susceptibility to TB. Tuberculosis and melioidosis share many key features including an overlap of clinical manifestations, immunopathology, and host immune responses. A previous study in Thailand demonstrated that transcriptomic profiles between tuberculosis and melioidosis were indistinguishable. Host-immune responses to the infections are dysregulated in people with diabetes including killing mechanisms, cell-cell communication, and migration. However, mechanisms involved in the increased susceptibility in DM to intracellular pathogens are to be elucidated. In this project we used RNA sequencing to study transcriptomics in melioidosis and tuberculosis patients in order to characterise the difference in host response to infection for those with and without DM. Differential gene expression analysis between melioidosis patients with (n = 49) and without DM (n = 32) identified two genes CD163L1 (scavenger-receptor cytokine-rich protein) and FAM1932 (a member of the CC chemokine family) were differentially expressed with a false discovery rate < 0.05. However, gene set enrichment analysis using hallmark gene sets showed distinctive profiles in several comparisons. A comparison between melioidosis cases with and without DM showed a number of enriched gene sets dominated by immune, proliferation, and signalling in the DM group only. We identified gene signatures in people with diabetes indicative of endoplasmic reticulum stress and are now confirming this finding in validation experiments in peripheral blood mononuclear cells. Further analysis comparing the impact of diabetes in acute melioidosis with tuberculosis revealed shared transcriptomic signatures of increased inflammatory responses, increased neutrophil degranulation and increased platelet degranulation. However, looking at the whole blood transcriptome we saw opposite effects of diabetes on interferon-gamma signalling, with an increase in acute melioidosis (which may represent non-specific neutrophil activation) and a decrease in tuberculosis. Using an additional sample set from a collaboration with Eoin West at University of Washington, USA, we compared patients who went on to survive melioidosis in Ubon Ratchathani (n = 80) with fatal cases (n = 84) and found dominance in fatal cases for non-specific pro-inflammatory pathways but down-regulation of T cell signalling pathways, especially in those with diabetes. Overall, our data shows that excessive pro-inflammatory immune responses with reduced T cell signalling are associated with poor clinical outcomes, and people with diabetes have an excessive non-specific pro-inflammatory response to both melioidosis and tuberculosis, with increased neutrophil and platelet activation. The massive neutrophil activation in sepsis from acute melioidosis is associated with increased interferon-gamma pathway activation, in contrast to tuberculosis where overall interferon-gamma activity is reduced in diabetes - most likely reflecting the less acute nature of disease from M. tuberculosis. Vaccines targeting T cells for these intracellular pathogens may be especially important to correct the increased susceptibility seen in diabetes.
Impact Collaborations: • This project allowed the building of a collaboration between S. Dunachie and J. Cliff, which we aim to be a foundation for further work. • Further collaborations that this project was a springboard for: 1. Paul Brett and Mary Burtnick, University of Nevada in Reno, USA - planning a Phase 1 vaccine trial of a public health melioidosis vaccine to target people with diabetes 2. Helen McShane and Elena Stylianou, U. Oxford, UK - establishing a mouse model of diabetes for vaccine testing 3. Claire Chewapreecha, Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand - "Dissecting the genetic basis of melioidosis infection" 4. Eoin West, University of Washington, Seattle, USA (extension from previous collaboration on sepsis and Toll-like receptors) - transcriptomic signatures of melioidosis and sepsis 5. Chiranjay Mukhopadhyay, Manipal Academy of Higher Education, India - cellular immunity in TB and melioidosis (which has been awarded a subsequent VALIDATE pump-priming grant, P033) 6. Mohammad Ali, Dhaka Medical College, Bangladesh - metabolism, DM and TB 7. Vivek Jha, George Institute, New Delhi, India - diabetes and infection database research 8. Prof. Chang-Hwa Song, Chungman National University, South Korea - macrophage immunology in TB +/- DM As a result of the VALIDATE networking session in 2018, a team assembled with an interest in developing a diabetic mouse model for testing of immune response to disease and vaccination, and this was a successful pump-priming award from VALIDATE in early 2019 (PI Elena Stylianou, P022 - further details below). Papers: This project influenced the following manuscripts: • Dunachie, S. and P. Chamnan, The double burden of diabetes and global infection in low and middle-income countries. Trans R Soc Trop Med Hyg, 2019. 113(2): p. 56-64. https://doi.org/10.1093/trstmh/try124 • Kronsteiner, B., et al., Diabetes alters immune response patterns to acute melioidosis in humans. Eur J Immunol, 2019. 49(7): p. 1092-1106 https://doi.org/10.1002/eji.201848037 • Eckold, C. et al., Impact of intermediate hyperglycaemia as well as diabetes on immune dysfunction in tuberculosis. Clinical Infectious Diseases 2021 https://doi.org/10.1093/cid/ciaa751 • Eckold et al., Impaired resolution of blood transcriptomes through tuberculosis treatment with diabetes comorbidity MedRxiv 2022 https://doi.org/10.1101/2022.02.07.22269422 • Tomás-Cortázar, J., Bossi, L., Quinn, C., Reynolds, C.J., Butler, D.K., Corcoran, N., . . . McClean, S. BpOmpW Antigen Stimulates the Necessary Protective T-Cell Responses Against Melioidosis. Front. Immunol. 12, 767359-767359 (2021). 10.3389/fimmu.2021.767359. https://pubmed.ncbi.nlm.nih.gov/34966388 • Chewapreecha, C., Pensar, J., Chattagul, S., Pesonen, M., Sangphukieo, A., Boonklang, P., . . . Corander, J. Co-evolutionary Signals Identify Burkholderia pseudomallei Survival Strategies in a Hostile Environment. Mol. Biol. Evol. 39, msab306 (2022). 10.1093/molbev/msab306. https://pubmed.ncbi.nlm.nih.gov/34662416 Two manuscripts are in preparation directly from this work: 1. Rongkard, P., Kronsteiner B., Hantrakunb, V., Teparrukkul, P., Wongsuvanb, G., Limmathurotsakul, D., Lovelace-Macon, L., Day, N.P., Klenerman, P., Chantratita, N., Gharib, S., Dunachie S., West, T.E. Transcriptomic signatures in fatal melioidosis. Manuscript in preparation 2. Rongkard, P., Hantrakunb, V., Teparrukkul, P., Wongsuvanb, G., Limmathurotsakul, D., Lovelace-Macon, L., Day, N.P., Gharib, S., West, T.E, Cliff, J., Klenerman, P., Kronsteiner B., Dunachie S. The impact of type 2 Diabetes on the immune response to melioidosis and tuberculosis. Manuscript in preparation Related funding: 1. 2021-2026 NIHR Professorship in Global Health "Developing a vaccine to prevent death from melioidosis in people with type 2 diabetes mellitus in low- and middle-income countries" (Dunachie PI), £1,951,296 2. 2020-2022 US Defense Threat Reduction Agency award "Optimization of lead melioidosis and glanders vaccine formulations" Prof Paul Brett University of Nevada, Reno PI with sub-contract award to Dunachie, Oxford PI, $733,976.61 USD (£524,269) to Dunachie lab. 3. 2022-2025 MRC Biomedical Catalyst Developmental Pathway Funding Scheme (DPFS) / US Defense Threat reduction Agency award, "Developing a vaccine for Burkholderia pseudomallei - a Phase I Clinical Trial" (Dunachie PI) £3,238,214. 4. 2019-2020 VALIDATE pump-priming project: "Characterising the cellular immunity and metabolic response to Mycobacterium tuberculosis and Burkholderia pseudomallei in Indian patients for vaccine design". (Mukhopadhyay, Manipal Academy of Higher Education, India PI, Dunachie Co-PI) £48,993 (£7,000 to Dunachie lab). 5. 2019-2020 Global Challenges Research Fund (GCRF) "Diabetes and Infection: Developing a network for using real-world datasets to evaluate the relationship between diabetes and infection in low and middle income countries?" (Dunachie PI) £?26,728.50. 6. 2019-2022 Bangladesh Prime Minister's PhD Scholarship for DPhil study at U. Oxford for "Metabolic changes in immune cells of diabetic patients and their impact on immune responses to tuberculosis"(Mohammad Ali, Dhaka Medical College, Bangladesh applicant, Dunachie supervisor) $258,823 USD (£184,874). 7. 2019-2020 MRC VALIDATE "Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis' (Stylianou PI, Dunachie Co-PI) £49,733.91 - £5,500 to Dunachie lab). In addition, S.Dunachie has been PI or Co-Investigator for grants totalling £5.7 million addressing the immune response to SARS-CoV-2 from infection and vaccination. The immunology expertise developed in her melioidosis work was rapidly applicable to SARS-CoV-2 due to similarity of vulnerable patient group (increased age, diabetes, immunocompromise). Capacity building: • This was Prof Susanna Dunachie's first formal funding to look at the relationship between diabetes and infection (although it was already an interest), and has sparked a whole programme of work (see collaborations and funding). She is now leading the world's first clinical trial of a vaccine for melioidosis. • Mr Patpong Rongkard, Mahidol-Oxford Tropical Medicine Research Unit (MORU), Bangkok - came to Oxford to undertake this project, and was able to register for a DPhil "Understanding how type II diabetes increases susceptibility to intracellular infections". This VALIDATE project has enabled him to receive extensive training in bioinformatics for analysis of RNAseq data, all using open-source software in R. He passed his DPhil viva in November 2022 and has now been awarded the DPhil. • Susanna Dunachie is supporting development of immunology research in India and Bangladesh, by supporting a new collaboration with Dr Mukhopadhyay at Manipal Academy of Higher Education, India, and supported the writing of a successful PhD scholarship application by Dr Ali of Dhaka Medical College, who is now undertaking a DPhil at U Oxford. • Dr Lee (LSHTM) was instrumental in writing her first grant application "Building investigator links for studies on the roles of complement C1q and endoplasmicreticulum (ER) stress in macrophage polarisation during Mycobacterium tuberculosis infection for a successfully funded MRC-KHIDI UK-Korea Partnering Award. • Secondments: Mr Rongkard was unable to undertake the planned secondment at U. Washington, Seattle, USA, in May 2020 due to the pandemic. However, using Microsoft Teams we have been able to build a close collaboration and he has used the skills acquired in this project to analyse the raw RNAseq data generated in an independent cohort of melioidosis patients as part of a sepsis study in the same region of Northeast Thailand as our study (see results). • Susanna organised the VALIDATE Melioidosis Vaccine Symposium: Moving from animal models to man on February 26th 2019, with support from the VALIDATE team (see separate section on this Symposium). Outreach: March 2022. Talk to local 6th form college about careers in tropical medicine. 2022 Dunachie S. Vaccines against melioidosis: How far are we? European Melioidosis Congress, Gratz, Austria, May 2022 2022 Rongkard P. T-cell dysfunction is associated with fatality in community-acquired melioidosis 2019 Dunachie S. Diabetes and melioidosis: What do we know, and what can we do? Invited plenary speaker at World Melioidosis Congress, Hanoi, October 2019 This project was presented at the 2018, 2019 and 2021 VALIDATE Annual Meetings. As a result of the VALIDATE networking session in 2018, a team assembled with an interest in developing a diabetic mouse model for testing of immune response to disease and vaccination, and this was a successful pump-priming award from VALIDATE in early 2019 (PI Elena Stylianou, P022). New datasets of RNAseq data in acute melioidosis have been generated from this project and will be available to VALIDATE members. Awards/Recognition: Susanna Dunachie: - Professor of Infectious Diseases - NIHR Global Research Professorship - Medical Fellow, Corpus Christi College, Oxford Product Development We organised a VALIDATE workshop on vaccines for melioidosis in February 2019, and as a result submitted a successful application to MRC's DPFS grant call to undertake the first human Phase I Vaccine Trial of a melioidosis vaccine, in Oxford using Paul Brett and Mary Burtnick's melioidosis vaccine candidate. The subunit vaccine has been developed at University of Nevada, Reno (UNR) and combines the bacterial capsule polysaccharide (CPS) conjugated to the immunogenic diphtheria toxin mutant CRM197, with recombinant B. pseudomallei Hcp1 protein in an Alhydrogel-CpG adjuvant. In a mouse model, the combined vaccine generated high levels of opsonising anti-CPS antibodies, alongside high T-cell responses to Hcp1, and protected 100% of mice against a robust lethal inhalational B. pseudomallei challenge. We plan to develop the vaccine as a public health vaccine for people with diabetes in low- and middle-income countries (LMICs). The group of investigators in UK (Dunachie, McShane, Kronsteiner), USA (Brett, Burtnick) and Thailand (Chantratita), brought together by the MRC VALIDATE Network, are planning a first-in-man study of 36 healthy human volunteer subjects in Oxford, UK, randomised to receive either the CPS-CRM197 alone, Hcp1 alone, or the CPS-CRM197 / Hcp1 combination. This will be followed by 12 subjects aged 40-60 years with Type II diabetes in Oxford receiving the CPS-CRM197 / Hcp1 combination. Endpoints will be safety and immunogenicity (antibody and T cell responses). The team seeks to define the key immune correlates of protection for monitoring vaccine efficacy in subsequent field trials, using functional growth inhibition assays and transcriptomics in the vaccinated population, existing melioidosis patient cohort studies, and animal models in collaboration with UNR. The vaccine is currently completing manufacture at Resilience Corp, Florida, USA (funded by US Defense Threat Reduction Agency) and we will commence the Phase 1 clinical trial in Oxford, UK in early 2023. Next we plan to undertake a Phase 1b study in Thailand (will require future funding). Ultimately we seek to accelerate production of a vaccine for the 280 million people with diabetes and the many with other risk factors for melioidosis including older age, chronic renal disease and immunosuppression who live in melioidosis-endemic regions in 83 countries across the globe.
Start Year 2018
 
Description VALIDATE Pump-priming project P019 
Organisation Federal University of Minas Gerais
Country Brazil 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Protective efficacy of conserved Leishmania hypothetical proteins against visceral leishmaniasis Led by Prof Myron Christodoulides (University of Southampton, UK), with Prof Eduardo Coelho (Federal Unversity of Minas Gerais, Brazil) Project Aims The parasite Leishmania infantum causes the disease Visceral Leishmaniasis (VL) with ~700,000 - 1 million new cases and 50,000 deaths reported annually. The parasite is transmitted by a sandfly vector and the highest rates of leishmaniasis are reported in Brazil, India and from other low-middle-income countries on the Indian/African continents. In Brazil, VL disproportionately affects the poorest in society, especially those who live in remote areas with low incomes and poor access to health programs. Dogs act as a parasite reservoir in Brazil for transmission to humans and thus, VL is of veterinary importance also, as the dog population in Brazil alone numbers ~20-30 million. There are NO vaccines for human VL and this project is collaboration between Brazilian and UK research groups with expertise in parasitology, vaccine development and immunology, with the aim to develop a novel experimental VL vaccine. We have already identified four components (proteins) of the parasite that could potentially go into a vaccine, and for this application we propose to discover the important parts of these proteins (the 'epitopes') that can stimulate human blood cells in the laboratory to produce the specific type of response that is needed for protection against infection. We will then combine these 'epitopes' to produce a chimera vaccine, which we will then produce in the laboratory by genetic engineering. We will also produce large amounts of this chimera vaccine for studies aimed at defining its structure. Importantly, we will test the chimera vaccine to see if it protects mice from L. infantum infection. Finally, the desired measures of success for our project of i) chimera vaccine development and ii) demonstrable protection in laboratory animals against parasite infection, could then be used as a springboard for eventual phase I testing in humans and dogs living in areas with endemic disease. Project Outcomes The parasite Leishmania infantum causes the disease Visceral Leishmaniasis (VL) with ~700,000 - 1 million new cases and 50,000 deaths reported annually, globally. The parasite is transmitted by a sandfly vector and the highest rates of leishmaniasis are reported in many of the least developed and low-middle income countries of Asia (e.g. India and Bangladesh), Africa (Sudan and Ethiopia), the Middle East and Latin America (e.g. Brazil). In Brazil, VL disproportionately affects the poorest in society, especially those who live in favelas and remote areas with low incomes and poor access to health programs. Dogs act as a parasite reservoir in Brazil for transmission to humans and thus, VL is of veterinary importance also, as the dog population in Brazil alone numbers ~20-30 million. There are no vaccines for human VL and this project was a collaboration between Brazilian and UK research groups with expertise in parasitology, vaccine development and immunology. Their aim was to develop a novel experimental VL vaccine. The team had already identified four components (proteins) of the parasite that could potentially go into a vaccine, and for this application they went further and discovered the important parts of these proteins (the 'epitopes') that could stimulate human blood cells in the laboratory to produce the specific type of response that is needed for protection against infection. This response is believed to be mediated by human T cells, and thus the team identified the important T cell epitopes from these proteins. The team then combined these 'T cell epitopes' to produce a vaccine called ChimeraT, which they then synthesised in the laboratory by genetic engineering. They then mixed ChimeraT with a detergent saponin and also incorporated it into artificial membranes called liposomes, to make two vaccines - ChimeraT/Saponin and ChimeraT/Liposome. These vaccines were injected into mice and the team studied the immune response following vaccination and also following infection. The key outcomes of their animal studies was that the vaccines stimulated a specific T cell response that protected the mice from infection with live parasites. Furthermore, ChimeraT/Liposome vaccine was the preferred choice of the two vaccines, as liposomes are non-toxic and safe for human vaccines. Thus, the project led to the development of ChimeraT/Liposome vaccine, and the outputs from the project will form the basis for further investigations into the potential of this vaccine for preventing leishmaniasis in both human and dogs.
Impact VALIDATE has been instrumental in joining together researchers in leishmaniasis in Brazil and microbiologists/vaccinologists in the UK to develop a candidate human chimera vaccine. The funding has allowed the Brazilian partner to do this valuable work, which might not otherwise have been possible given the current funding situation in Brazil. Direct Papers 1. A candidate vaccine for human visceral leishmaniasis based on a specific T cell epitope-containing chimeric protein protects mice against Leishmania infantum infection. Lage DP, Ribeiro PAF, Dias DS, Mendonça DVC, Ramos FF, Carvalho LM, de Oliveira D, Steiner BT, Martins VT, Perin L, Machado AS, Santos TTO, Tavares GSV, Oliveira-da-Silva JA, Oliveira JS, Roatt BM, Machado-de-Ávila RA, Teixeira AL, Humbert MV, Coelho EAF*, Christodoulides M*. NPJ Vaccines. 2020 Aug 13;5:75. doi: 10.1038/s41541-020-00224-0. eCollection 2020. PMID: 32821440 Free PMC article. 2. Liposomal Formulation of ChimeraT, a Multiple T-Cell Epitope-Containing Recombinant Protein, Is a Candidate Vaccine for Human Visceral Leishmaniasis. Lage DP, Ribeiro PAF, Dias DS, Mendonça DVC, Ramos FF, Carvalho LM, Steiner BT, Tavares GSV, Martins VT, Machado AS, Oliveira-da-Silva JA, Santos TTO, Freitas CS, Oliveira JS, Roatt BM, Machado-de-Ávila RA, Humbert MV, Christodoulides M*, Coelho EAF*. Vaccines (Basel). 2020 Jun 9;8(2):289. doi: 10.3390/vaccines8020289. PMID: 32526867 Free PMC article. * Equal co-investigators. Book: "Vaccines for Neglected Pathogens: Strategies, Achievements and Challenges - Focus on Leprosy, Leishmaniasis, Melioidosis and Tuberculosis". Editors: Myron Christodoulides. ISBN: 978-3-031-24354-7. Due for publication March/April 2023. This book showcases VALIDATE, research by VALIDATE members, and highlights the importance of VALIDATE's pathogens. Papers also citing VALIDATE as the supported post-docs contributed to these studies: 1. Preclinical Assessment of the Immunogenicity of Experimental Leishmania Vaccines. Martins VT, Machado AS, Humbert MV, Christodoulides M, Coelho EAF. Methods Mol Biol. 2022;2410:481-502. doi: 10.1007/978-1-0716-1884-4_25. PMID: 34914064 2. In Silico Design of Recombinant Chimera T Cell Peptide Epitope Vaccines for Visceral Leishmaniasis. Machado AS, Martins VT, Humbert MV, Christodoulides M, Coelho EAF. Methods Mol Biol. 2022;2410:463-480. doi: 10.1007/978-1-0716-1884-4_24. PMID: 34914063 3. Sensitive and specific serodiagnosis of tegumentary leishmaniasis using a new chimeric protein based on specific B-cell epitopes of Leishmania antigenic proteins. Galvani NC, Machado AS, Lage DP, Martins VT, de Oliveira D, Freitas CS, Vale DL, Fernandes BB, Oliveira-da-Silva JA, Reis TAR, Santos TTO, Ramos FF, Bandeira RS, Ludolf F, Tavares GSV, Guimarães NS, Tupinambás U, Chávez-Fumagalli MA, Humbert MV, Gonçalves DU, Christodoulides M, Machado-de-Ávila RA, Coelho EAF. Microb Pathog. 2022 Jan;162:105341. doi: 10.1016/j.micpath.2021.105341. Epub 2021 Dec 6. PMID: 34883228 4. ChimLeish, a new recombinant chimeric protein evaluated as a diagnostic and prognostic marker for visceral leishmaniasis and human immunodeficiency virus coinfection. Galvani NC, Machado AS, Lage DP, Freitas CS, Vale DL, de Oliveira D, Ludolf F, Ramos FF, Fernandes BB, Luiz GP, Mendonça DVC, Oliveira-da-Silva JA, Reis TAR, Tavares GSV, Chaves AT, Guimarães NS, Tupinambás U, Cota GF, Humbert MV, Martins VT, Christodoulides M, Coelho EAF, Machado-de-Ávila RA. Parasitol Res. 2021 Dec;120(12):4037-4047. doi: 10.1007/s00436-021-07342-1. Epub 2021 Oct 19. PMID: 34664113 Free PMC article. 5. Potential of recombinant LiHyQ, a novel Leishmania infantum protein, for the diagnosis of canine visceral leishmaniasis and as a diagnostic and prognostic marker for human leishmaniasis and human immunodeficiency virus co-infection: A preliminary study. Santos TTO, Ramos FF, Gonçalves IAP, Tavares GSV, Ludolf F, Bandeira RS, Silva AM, Oliveira-da-Silva JA, Reis TAR, Machado AS, Lage DP, Freitas CS, Vale DL, Martins VT, Alves LA, Guimarães NS, Chaves AT, Chávez-Fumagalli MA, Cota GF, Silveira JAG, Tupinambás U, Gonçalves DU, Christodoulides M, Coelho EAF. Acta Trop. 2021 Dec;224:106126. doi: 10.1016/j.actatropica.2021.106126. Epub 2021 Sep 16. PMID: 34537185 Grants Myron and Eduardo submitted a BBSCR (responsive mode in animal health) grant application in April 2021 to propose hamster vaccination trials with ChimeraT/Liposome vaccine (unsuccessful). Early Career Researcher Dr Victoria Humbert applied for, and was awarded, a VALIDATE training grant of £2,490 to visit Prof Eduardo Coelho and his research group in Brazil. Capacity building Three early career postdoctoral researchers ECR have been involved in the project (Dr Humbert (UK), Drs Tamietti and Lage (Brazil)) and they are all named authors on the published papers. In addition, Dr Humbert from the UK won a separate VALIDATE training grant and has visited the laboratories in Brazil to learn Leishmania biology. This is important for developing her portfolio of research techniques and for forward planning (for further details see training grant section). In addition, some of the data have been presented by the Brazilian ECR at their 5th and 6th Annual Meeting of the Post-Graduation Program in Healthy Sciences: Infectious Diseases and Tropical Medicine of Faculty of Medicine (UFMG, Brazil) (November, 2019 and 2020, online). The application to BBSRC has Drs Lage and Tamietti as named early career researchers, as well. Engagement Project lead Prof Myron Christodoulides (Southampton) was invited to a workshop in Brasilia in March 2020 (British Academy) that included discussion of health issues in Brazil (infectious diseases) and the contribution of factors such as climate change, city infrastructure, pollution. In fact, he was awarded a £10,000 pump-priming grant from the British Academy in collaboration with Professors Seabra and Paes, ABC University and Fiocruz, Sao Paolo State, to kick-start a project on 'Diagnostics and Vaccines for Leishmaniasis'. This project was presented to VALIDATE members at the 2019 VALIDATE Annual Meeting.
Start Year 2021
 
Description VALIDATE Pump-priming project P019 
Organisation University of Southampton
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Protective efficacy of conserved Leishmania hypothetical proteins against visceral leishmaniasis Led by Prof Myron Christodoulides (University of Southampton, UK), with Prof Eduardo Coelho (Federal Unversity of Minas Gerais, Brazil) Project Aims The parasite Leishmania infantum causes the disease Visceral Leishmaniasis (VL) with ~700,000 - 1 million new cases and 50,000 deaths reported annually. The parasite is transmitted by a sandfly vector and the highest rates of leishmaniasis are reported in Brazil, India and from other low-middle-income countries on the Indian/African continents. In Brazil, VL disproportionately affects the poorest in society, especially those who live in remote areas with low incomes and poor access to health programs. Dogs act as a parasite reservoir in Brazil for transmission to humans and thus, VL is of veterinary importance also, as the dog population in Brazil alone numbers ~20-30 million. There are NO vaccines for human VL and this project is collaboration between Brazilian and UK research groups with expertise in parasitology, vaccine development and immunology, with the aim to develop a novel experimental VL vaccine. We have already identified four components (proteins) of the parasite that could potentially go into a vaccine, and for this application we propose to discover the important parts of these proteins (the 'epitopes') that can stimulate human blood cells in the laboratory to produce the specific type of response that is needed for protection against infection. We will then combine these 'epitopes' to produce a chimera vaccine, which we will then produce in the laboratory by genetic engineering. We will also produce large amounts of this chimera vaccine for studies aimed at defining its structure. Importantly, we will test the chimera vaccine to see if it protects mice from L. infantum infection. Finally, the desired measures of success for our project of i) chimera vaccine development and ii) demonstrable protection in laboratory animals against parasite infection, could then be used as a springboard for eventual phase I testing in humans and dogs living in areas with endemic disease. Project Outcomes The parasite Leishmania infantum causes the disease Visceral Leishmaniasis (VL) with ~700,000 - 1 million new cases and 50,000 deaths reported annually, globally. The parasite is transmitted by a sandfly vector and the highest rates of leishmaniasis are reported in many of the least developed and low-middle income countries of Asia (e.g. India and Bangladesh), Africa (Sudan and Ethiopia), the Middle East and Latin America (e.g. Brazil). In Brazil, VL disproportionately affects the poorest in society, especially those who live in favelas and remote areas with low incomes and poor access to health programs. Dogs act as a parasite reservoir in Brazil for transmission to humans and thus, VL is of veterinary importance also, as the dog population in Brazil alone numbers ~20-30 million. There are no vaccines for human VL and this project was a collaboration between Brazilian and UK research groups with expertise in parasitology, vaccine development and immunology. Their aim was to develop a novel experimental VL vaccine. The team had already identified four components (proteins) of the parasite that could potentially go into a vaccine, and for this application they went further and discovered the important parts of these proteins (the 'epitopes') that could stimulate human blood cells in the laboratory to produce the specific type of response that is needed for protection against infection. This response is believed to be mediated by human T cells, and thus the team identified the important T cell epitopes from these proteins. The team then combined these 'T cell epitopes' to produce a vaccine called ChimeraT, which they then synthesised in the laboratory by genetic engineering. They then mixed ChimeraT with a detergent saponin and also incorporated it into artificial membranes called liposomes, to make two vaccines - ChimeraT/Saponin and ChimeraT/Liposome. These vaccines were injected into mice and the team studied the immune response following vaccination and also following infection. The key outcomes of their animal studies was that the vaccines stimulated a specific T cell response that protected the mice from infection with live parasites. Furthermore, ChimeraT/Liposome vaccine was the preferred choice of the two vaccines, as liposomes are non-toxic and safe for human vaccines. Thus, the project led to the development of ChimeraT/Liposome vaccine, and the outputs from the project will form the basis for further investigations into the potential of this vaccine for preventing leishmaniasis in both human and dogs.
Impact VALIDATE has been instrumental in joining together researchers in leishmaniasis in Brazil and microbiologists/vaccinologists in the UK to develop a candidate human chimera vaccine. The funding has allowed the Brazilian partner to do this valuable work, which might not otherwise have been possible given the current funding situation in Brazil. Direct Papers 1. A candidate vaccine for human visceral leishmaniasis based on a specific T cell epitope-containing chimeric protein protects mice against Leishmania infantum infection. Lage DP, Ribeiro PAF, Dias DS, Mendonça DVC, Ramos FF, Carvalho LM, de Oliveira D, Steiner BT, Martins VT, Perin L, Machado AS, Santos TTO, Tavares GSV, Oliveira-da-Silva JA, Oliveira JS, Roatt BM, Machado-de-Ávila RA, Teixeira AL, Humbert MV, Coelho EAF*, Christodoulides M*. NPJ Vaccines. 2020 Aug 13;5:75. doi: 10.1038/s41541-020-00224-0. eCollection 2020. PMID: 32821440 Free PMC article. 2. Liposomal Formulation of ChimeraT, a Multiple T-Cell Epitope-Containing Recombinant Protein, Is a Candidate Vaccine for Human Visceral Leishmaniasis. Lage DP, Ribeiro PAF, Dias DS, Mendonça DVC, Ramos FF, Carvalho LM, Steiner BT, Tavares GSV, Martins VT, Machado AS, Oliveira-da-Silva JA, Santos TTO, Freitas CS, Oliveira JS, Roatt BM, Machado-de-Ávila RA, Humbert MV, Christodoulides M*, Coelho EAF*. Vaccines (Basel). 2020 Jun 9;8(2):289. doi: 10.3390/vaccines8020289. PMID: 32526867 Free PMC article. * Equal co-investigators. Book: "Vaccines for Neglected Pathogens: Strategies, Achievements and Challenges - Focus on Leprosy, Leishmaniasis, Melioidosis and Tuberculosis". Editors: Myron Christodoulides. ISBN: 978-3-031-24354-7. Due for publication March/April 2023. This book showcases VALIDATE, research by VALIDATE members, and highlights the importance of VALIDATE's pathogens. Papers also citing VALIDATE as the supported post-docs contributed to these studies: 1. Preclinical Assessment of the Immunogenicity of Experimental Leishmania Vaccines. Martins VT, Machado AS, Humbert MV, Christodoulides M, Coelho EAF. Methods Mol Biol. 2022;2410:481-502. doi: 10.1007/978-1-0716-1884-4_25. PMID: 34914064 2. In Silico Design of Recombinant Chimera T Cell Peptide Epitope Vaccines for Visceral Leishmaniasis. Machado AS, Martins VT, Humbert MV, Christodoulides M, Coelho EAF. Methods Mol Biol. 2022;2410:463-480. doi: 10.1007/978-1-0716-1884-4_24. PMID: 34914063 3. Sensitive and specific serodiagnosis of tegumentary leishmaniasis using a new chimeric protein based on specific B-cell epitopes of Leishmania antigenic proteins. Galvani NC, Machado AS, Lage DP, Martins VT, de Oliveira D, Freitas CS, Vale DL, Fernandes BB, Oliveira-da-Silva JA, Reis TAR, Santos TTO, Ramos FF, Bandeira RS, Ludolf F, Tavares GSV, Guimarães NS, Tupinambás U, Chávez-Fumagalli MA, Humbert MV, Gonçalves DU, Christodoulides M, Machado-de-Ávila RA, Coelho EAF. Microb Pathog. 2022 Jan;162:105341. doi: 10.1016/j.micpath.2021.105341. Epub 2021 Dec 6. PMID: 34883228 4. ChimLeish, a new recombinant chimeric protein evaluated as a diagnostic and prognostic marker for visceral leishmaniasis and human immunodeficiency virus coinfection. Galvani NC, Machado AS, Lage DP, Freitas CS, Vale DL, de Oliveira D, Ludolf F, Ramos FF, Fernandes BB, Luiz GP, Mendonça DVC, Oliveira-da-Silva JA, Reis TAR, Tavares GSV, Chaves AT, Guimarães NS, Tupinambás U, Cota GF, Humbert MV, Martins VT, Christodoulides M, Coelho EAF, Machado-de-Ávila RA. Parasitol Res. 2021 Dec;120(12):4037-4047. doi: 10.1007/s00436-021-07342-1. Epub 2021 Oct 19. PMID: 34664113 Free PMC article. 5. Potential of recombinant LiHyQ, a novel Leishmania infantum protein, for the diagnosis of canine visceral leishmaniasis and as a diagnostic and prognostic marker for human leishmaniasis and human immunodeficiency virus co-infection: A preliminary study. Santos TTO, Ramos FF, Gonçalves IAP, Tavares GSV, Ludolf F, Bandeira RS, Silva AM, Oliveira-da-Silva JA, Reis TAR, Machado AS, Lage DP, Freitas CS, Vale DL, Martins VT, Alves LA, Guimarães NS, Chaves AT, Chávez-Fumagalli MA, Cota GF, Silveira JAG, Tupinambás U, Gonçalves DU, Christodoulides M, Coelho EAF. Acta Trop. 2021 Dec;224:106126. doi: 10.1016/j.actatropica.2021.106126. Epub 2021 Sep 16. PMID: 34537185 Grants Myron and Eduardo submitted a BBSCR (responsive mode in animal health) grant application in April 2021 to propose hamster vaccination trials with ChimeraT/Liposome vaccine (unsuccessful). Early Career Researcher Dr Victoria Humbert applied for, and was awarded, a VALIDATE training grant of £2,490 to visit Prof Eduardo Coelho and his research group in Brazil. Capacity building Three early career postdoctoral researchers ECR have been involved in the project (Dr Humbert (UK), Drs Tamietti and Lage (Brazil)) and they are all named authors on the published papers. In addition, Dr Humbert from the UK won a separate VALIDATE training grant and has visited the laboratories in Brazil to learn Leishmania biology. This is important for developing her portfolio of research techniques and for forward planning (for further details see training grant section). In addition, some of the data have been presented by the Brazilian ECR at their 5th and 6th Annual Meeting of the Post-Graduation Program in Healthy Sciences: Infectious Diseases and Tropical Medicine of Faculty of Medicine (UFMG, Brazil) (November, 2019 and 2020, online). The application to BBSRC has Drs Lage and Tamietti as named early career researchers, as well. Engagement Project lead Prof Myron Christodoulides (Southampton) was invited to a workshop in Brasilia in March 2020 (British Academy) that included discussion of health issues in Brazil (infectious diseases) and the contribution of factors such as climate change, city infrastructure, pollution. In fact, he was awarded a £10,000 pump-priming grant from the British Academy in collaboration with Professors Seabra and Paes, ABC University and Fiocruz, Sao Paolo State, to kick-start a project on 'Diagnostics and Vaccines for Leishmaniasis'. This project was presented to VALIDATE members at the 2019 VALIDATE Annual Meeting.
Start Year 2021
 
Description VALIDATE Pump-priming project P020 
Organisation Brunel University London
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy Led by Prof Francisco Javier Sánchez-García (Instituto Politécnico Nacional, Mexico), with Dr Steven Smith (LSHTM originally, now Brunel University, UK), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Prof Hazel Dockrell (LSHTM, UK) Project Aims In theory, every single infectious disease could be eliminated from the earth if good vaccines (plus appropriate social and public health policies) are developed. In this scenario the best example is the vaccine against smallpox that lead to complete eradication of the disease by the year 1980. Other vaccines, however, have not been so successful, notoriously BCG. This vaccine, first used in humans in 1921, has greatly contributed to reducing TB, but still, TB remains the world's top infectious cause of mortality. In addition, BCG has proved to be protective in some countries but not in others. Why is that so? Several hypotheses have been put forward and, for practical purposes, this has conducted researchers to try to develop "better anti-tuberculosis vaccines". Here, we propose a different approach: "current BCG is an excellent vaccine, it contains most of the potentially protective antigens. Moreover, it contains its own natural adjuvants; the problem is that we have failed to deliver it into the appropriate metabolic tissue microenvironment". Recent research shows that the quality of immune cell effector functions are determined by their particular metabolism at the time (i.e. how they produce energy), and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumors. We propose to reprogram the local cell metabolism at the site (skin) where the BCG vaccine will be delivered, to ensure the best immune cell activation and consequently, the highest protection. Project Outcomes This project originated from the need to stop TB and from the fact that the BCG vaccine, used in humans since 1921, has proved to be protective against TB in some countries but not in others. We asked ourselves: why is that so? While BCG fulfils all the criteria of an excellent vaccine, containing most of the potentially protective antigens and its own natural adjuvants, we hypothesized that so far it has not been delivered into the appropriate metabolic microenvironment. This idea is supported by the recent finding that the quality of immune cell effector functions is largely determined by their particular metabolism at the time, and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumours. We proposed to metabolically pre-condition the local tissue microenvironment to potentiate antigen uptake/processing and trafficking upon the administration of the current BCG vaccine, in order to improve anti-TB protection. We selected five metabolites to be tested: lactate, succinate, fumarate acetate and butyrate, all of which harbour cell signalling properties. Based on our preliminary results, all were used at a final concentration of 100 uM. Firstly, we found structural and functional changes of mitochondria from human monocyte-derived macrophages in response to in vitro treatment with the aforementioned metabolites (100 uM). Fumarate induced mitochondrial fusion, polarization, and changes in mitochondrial cristae; in contrast, succinate, lactate and butyrate caused mitochondrial fission and depolarization. All the metabolites tested triggered an increase in cytoplasmic and mitochondrial calcium concentration within a few seconds, and fumarate promoted the highest calcium influx to the cytoplasm whereas the mitochondrial calcium concentrations, following metabolite treatment, were similar for all the metabolites tested. Furthermore, extracellular flux analysis of human monocytes demonstrated changes in the use of energy pathways upon metabolite treatment. Succinate treatment resulted in an immediate increase of extracellular acidification rate (ECAR), indicative of increased glycolysis, whereas no effect was observed for the other metabolites. Overnight pre- treatment with succinate led to a reduction in mitochondrial ATP production compared to vehicle, whereas fumarate treatment had the opposite effect. We then exposed human peripheral blood mononuclear cells (PBMC) to the different metabolites for 24 h and cultured the cells in the presence of BCG in order to assess mycobacterial growth inhibition by un-stimulated cells and by metabolite-stimulated cells. Results from six donors showed no statistically significant differences between treatments. Metabolite pre-treatment was able to change the expression of co-stimulatory (CD80, CD86) and co-inhibitory (PD-L1) molecules on the monocytic/macrophage cell line (RAW 264.7) upon stimulation with lipopolysaccharide (LPS), with succinate increasing activation as demonstrated by increased cell size and relative amount of CD80 compared to LPS alone. Preliminary data from in vivo mouse experiments demonstrate that combined injection (i.d.) of BCG and metabolite into the back of mice elicits histopathological changes and differences in immune cell composition as compared to BCG alone. Overall, our study highlights benefits of metabolite pre-treatment on activation, metabolic properties and function of monocytes/macrophages and suggests that metabolite-induced tissue metabolic reprogramming is feasible.
Impact Collaborations - Continuation of collaboration between Prof Javier Sanchez-Garcia (IPN, Mexico), Dr Steven Smith (LSHTM; now Brunel) and Prof Hazel Dockrell (LSHTM), that started with VALIDATE grant P004. - Project is a new collaboration, formed at VALIDATE's 2nd Annual Meeting, with Dr Barbara Kronsteiner-Dobramysl (University of Oxford). - Addition of Prof Rogelio Hernandez-Pando to the research team (National Institute of Medical Sciences and Nutrition; 2022) - In the course of the project a new collaboration with Professor Oscar Rojas-Espinosa (ENCB-IPN), a leading expert on murine leprosy, was established. - As a direct result of VALIDATE´s policy to encourage collaborations, Prof. Sánchez-García was invited (through VALIDATE) by Dr. Vijay Kumar Prajapati to write a chapter for a book on Vaccines: María Maximina Bertha Moreno-Altamirano, Jacqueline Oliva-Ramírez y, F. Javier Sánchez-García. "Circadian Rhythmicity and Vaccination" In: Vijay Kumar Prajapati (Ed.) System Vaccinology. (2022) Elsevier, Pp 207-229. - A new Collaboration with Professor Rogelio Hernández-Pando, Institito Nacional de Ciencias Medicas y de la Nutrición "Salvador Zubirán", Mexico City, was established, to evaluate the protective effect of BCG plus metabolites immunization in a murine model of M. tuberculosis infection. Work in progress in this area. Direct publications: Pérez-Hernández CA, Moreno-Altamirano MMB, López-Villegas EO, Butkeviciute E, Ali M, Kronsteiner B, Dunachie SJ, Dockrell HM, Smith SG, Sánchez-García, FJ. (2022). Mitochondrial Ultrastructure and Activity are Differentially Regulated by Glycolysis-, Krebs cycle-, and Microbiota-derived Metabolites in Monocytes. Biology 11: 1132. doi.org/10.3390/ biology11081132. Related publications crediting VALIDATE funding: Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno- Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ (2020) Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate-Induced Innate Immune Training. Front. Immunol. 11:1715. doi: 10.3389/fimmu.2020.01715. Prentice S, Dockrell HM. (2020). Antituberculosis BCG vaccination: more reasons for varying innate and adaptive immune responses. J Clin Invest. 130(10):5121-5123. doi: 10.1172/JCI141317. Prentice S, Dockrell HM. (2021). BCG-specific and non-specific effects: different questions, similar challenges. J Infect Dis. jiab307. doi: 10.1093/infdis/jiab307. Dockrell HM, Butkeviciute E. (2021). Can what have we learnt about BCG vaccination in the last 20 years help us to design a better tuberculosis vaccine? Vaccine. S0264- 410X(21)00109-2. doi: 10.1016/j.vaccine.2021.01.068. Moreno-Altamirano MMB, Oliva-Ramírez J, Sánchez-García FJ. (2021). Chapter 9: Circadian Rhythmicity and Vaccination. In: Systems Vaccination. Prajapati VK (Ed.) (In press). Paper published by two VALIDATE/project members: Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García, FJ. (2020). Mitochondria: an integrative hub coordinating circadian rhythms, metabolism, the microbiome, and immunity. Frontiers in Cell and Developmental Biology 8:51. doi: 10.3389/fcell.2020.00051. Engagement activities: International congress : Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno-Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ. Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate Induced Innate Immune Training. 11th World Congress on Targeting Mitochondria (Germany, virtual congress), October 28-30, 2020. The project was also presented to VALIDATE members at the 2019 & 2020 Annual Meeting, and Dr Barbara Kronsteiner-Dobramysl also presented a talk "Diabetes alters cellular metabolism of T cells: implications for vaccine development" at the 2019 VALIDATE-BSI Conference. Grant applications: - A grant application (Newton Fund-The Ministry of Education, Science, Technology and Innovation of Mexico City) aimed at analysing the metabolic basis and trained immunity potential of Covid-19 has been jointly submitted by Steve Smith and F. Javier Sánchez-García, as PIs, along with other colleagues from UK and Mexico. Outcome pending. - Steve Smith submitted a grant application to the Academy of Medical Sciences Springboard scheme. Capacity Building - Career progression of two earlier stage researchers: • Dr Steve Smith: 'In 2020 I was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, my application for which was strengthen by having received VALIDATE funding for a project in my area of interest, as well as by the VALIDATE network overall and the the collaborations it has been possible to develop.' • Dr Barbara Kronsteiner-Dobramysl: 'Being a co-investigator on a VALIDATE Pump priming grant enables me to expand and demonstrate my expertise in Immunometabolism research, significantly contributing to establishing myself as an expert in this field.' • In addition, Prof Javier Sanchez-Garcia: 'Having two VALIDATE grants approved (one as participant and one as PI) has positively contributed in internal (Instituto Politécnico Nacional) academic evaluations; and this plus the expected publications and academic mobility of one of my students will have a good impact on the academic assessment of our Immunology postgraduate program as a whole.' - Two IPN (Mexico) PhD students participated in this VALIDATE project, one of who has successfully graduated in February 2021 (Angélica Pérez-Hernández) and continued to be employed on the project, funded by the P020 VALIDATE grant (instead of the originally planned 2-3 month lab exchange visit of a Mexican student to LSHTM, which could not take place due to the COVID pandemic). - Egle Butkeviciute, a Lithuanian PhD student at LSHTM, joined the project, and performed some mitochondrial dynamics experiments. - Perla Mariana Gutierréz Cedillo earned a M. Sc degree, working on "metabolic reprogramming to improve BCG vaccination-induced protection" , January 2023 - The acquisition of a small equipment (Countess 3, automated cell counter, Thermo Fisher) will have a positive long-term impact on the Immunoregulation Lab work in Mexico.
Start Year 2019
 
Description VALIDATE Pump-priming project P020 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy Led by Prof Francisco Javier Sánchez-García (Instituto Politécnico Nacional, Mexico), with Dr Steven Smith (LSHTM originally, now Brunel University, UK), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Prof Hazel Dockrell (LSHTM, UK) Project Aims In theory, every single infectious disease could be eliminated from the earth if good vaccines (plus appropriate social and public health policies) are developed. In this scenario the best example is the vaccine against smallpox that lead to complete eradication of the disease by the year 1980. Other vaccines, however, have not been so successful, notoriously BCG. This vaccine, first used in humans in 1921, has greatly contributed to reducing TB, but still, TB remains the world's top infectious cause of mortality. In addition, BCG has proved to be protective in some countries but not in others. Why is that so? Several hypotheses have been put forward and, for practical purposes, this has conducted researchers to try to develop "better anti-tuberculosis vaccines". Here, we propose a different approach: "current BCG is an excellent vaccine, it contains most of the potentially protective antigens. Moreover, it contains its own natural adjuvants; the problem is that we have failed to deliver it into the appropriate metabolic tissue microenvironment". Recent research shows that the quality of immune cell effector functions are determined by their particular metabolism at the time (i.e. how they produce energy), and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumors. We propose to reprogram the local cell metabolism at the site (skin) where the BCG vaccine will be delivered, to ensure the best immune cell activation and consequently, the highest protection. Project Outcomes This project originated from the need to stop TB and from the fact that the BCG vaccine, used in humans since 1921, has proved to be protective against TB in some countries but not in others. We asked ourselves: why is that so? While BCG fulfils all the criteria of an excellent vaccine, containing most of the potentially protective antigens and its own natural adjuvants, we hypothesized that so far it has not been delivered into the appropriate metabolic microenvironment. This idea is supported by the recent finding that the quality of immune cell effector functions is largely determined by their particular metabolism at the time, and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumours. We proposed to metabolically pre-condition the local tissue microenvironment to potentiate antigen uptake/processing and trafficking upon the administration of the current BCG vaccine, in order to improve anti-TB protection. We selected five metabolites to be tested: lactate, succinate, fumarate acetate and butyrate, all of which harbour cell signalling properties. Based on our preliminary results, all were used at a final concentration of 100 uM. Firstly, we found structural and functional changes of mitochondria from human monocyte-derived macrophages in response to in vitro treatment with the aforementioned metabolites (100 uM). Fumarate induced mitochondrial fusion, polarization, and changes in mitochondrial cristae; in contrast, succinate, lactate and butyrate caused mitochondrial fission and depolarization. All the metabolites tested triggered an increase in cytoplasmic and mitochondrial calcium concentration within a few seconds, and fumarate promoted the highest calcium influx to the cytoplasm whereas the mitochondrial calcium concentrations, following metabolite treatment, were similar for all the metabolites tested. Furthermore, extracellular flux analysis of human monocytes demonstrated changes in the use of energy pathways upon metabolite treatment. Succinate treatment resulted in an immediate increase of extracellular acidification rate (ECAR), indicative of increased glycolysis, whereas no effect was observed for the other metabolites. Overnight pre- treatment with succinate led to a reduction in mitochondrial ATP production compared to vehicle, whereas fumarate treatment had the opposite effect. We then exposed human peripheral blood mononuclear cells (PBMC) to the different metabolites for 24 h and cultured the cells in the presence of BCG in order to assess mycobacterial growth inhibition by un-stimulated cells and by metabolite-stimulated cells. Results from six donors showed no statistically significant differences between treatments. Metabolite pre-treatment was able to change the expression of co-stimulatory (CD80, CD86) and co-inhibitory (PD-L1) molecules on the monocytic/macrophage cell line (RAW 264.7) upon stimulation with lipopolysaccharide (LPS), with succinate increasing activation as demonstrated by increased cell size and relative amount of CD80 compared to LPS alone. Preliminary data from in vivo mouse experiments demonstrate that combined injection (i.d.) of BCG and metabolite into the back of mice elicits histopathological changes and differences in immune cell composition as compared to BCG alone. Overall, our study highlights benefits of metabolite pre-treatment on activation, metabolic properties and function of monocytes/macrophages and suggests that metabolite-induced tissue metabolic reprogramming is feasible.
Impact Collaborations - Continuation of collaboration between Prof Javier Sanchez-Garcia (IPN, Mexico), Dr Steven Smith (LSHTM; now Brunel) and Prof Hazel Dockrell (LSHTM), that started with VALIDATE grant P004. - Project is a new collaboration, formed at VALIDATE's 2nd Annual Meeting, with Dr Barbara Kronsteiner-Dobramysl (University of Oxford). - Addition of Prof Rogelio Hernandez-Pando to the research team (National Institute of Medical Sciences and Nutrition; 2022) - In the course of the project a new collaboration with Professor Oscar Rojas-Espinosa (ENCB-IPN), a leading expert on murine leprosy, was established. - As a direct result of VALIDATE´s policy to encourage collaborations, Prof. Sánchez-García was invited (through VALIDATE) by Dr. Vijay Kumar Prajapati to write a chapter for a book on Vaccines: María Maximina Bertha Moreno-Altamirano, Jacqueline Oliva-Ramírez y, F. Javier Sánchez-García. "Circadian Rhythmicity and Vaccination" In: Vijay Kumar Prajapati (Ed.) System Vaccinology. (2022) Elsevier, Pp 207-229. - A new Collaboration with Professor Rogelio Hernández-Pando, Institito Nacional de Ciencias Medicas y de la Nutrición "Salvador Zubirán", Mexico City, was established, to evaluate the protective effect of BCG plus metabolites immunization in a murine model of M. tuberculosis infection. Work in progress in this area. Direct publications: Pérez-Hernández CA, Moreno-Altamirano MMB, López-Villegas EO, Butkeviciute E, Ali M, Kronsteiner B, Dunachie SJ, Dockrell HM, Smith SG, Sánchez-García, FJ. (2022). Mitochondrial Ultrastructure and Activity are Differentially Regulated by Glycolysis-, Krebs cycle-, and Microbiota-derived Metabolites in Monocytes. Biology 11: 1132. doi.org/10.3390/ biology11081132. Related publications crediting VALIDATE funding: Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno- Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ (2020) Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate-Induced Innate Immune Training. Front. Immunol. 11:1715. doi: 10.3389/fimmu.2020.01715. Prentice S, Dockrell HM. (2020). Antituberculosis BCG vaccination: more reasons for varying innate and adaptive immune responses. J Clin Invest. 130(10):5121-5123. doi: 10.1172/JCI141317. Prentice S, Dockrell HM. (2021). BCG-specific and non-specific effects: different questions, similar challenges. J Infect Dis. jiab307. doi: 10.1093/infdis/jiab307. Dockrell HM, Butkeviciute E. (2021). Can what have we learnt about BCG vaccination in the last 20 years help us to design a better tuberculosis vaccine? Vaccine. S0264- 410X(21)00109-2. doi: 10.1016/j.vaccine.2021.01.068. Moreno-Altamirano MMB, Oliva-Ramírez J, Sánchez-García FJ. (2021). Chapter 9: Circadian Rhythmicity and Vaccination. In: Systems Vaccination. Prajapati VK (Ed.) (In press). Paper published by two VALIDATE/project members: Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García, FJ. (2020). Mitochondria: an integrative hub coordinating circadian rhythms, metabolism, the microbiome, and immunity. Frontiers in Cell and Developmental Biology 8:51. doi: 10.3389/fcell.2020.00051. Engagement activities: International congress : Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno-Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ. Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate Induced Innate Immune Training. 11th World Congress on Targeting Mitochondria (Germany, virtual congress), October 28-30, 2020. The project was also presented to VALIDATE members at the 2019 & 2020 Annual Meeting, and Dr Barbara Kronsteiner-Dobramysl also presented a talk "Diabetes alters cellular metabolism of T cells: implications for vaccine development" at the 2019 VALIDATE-BSI Conference. Grant applications: - A grant application (Newton Fund-The Ministry of Education, Science, Technology and Innovation of Mexico City) aimed at analysing the metabolic basis and trained immunity potential of Covid-19 has been jointly submitted by Steve Smith and F. Javier Sánchez-García, as PIs, along with other colleagues from UK and Mexico. Outcome pending. - Steve Smith submitted a grant application to the Academy of Medical Sciences Springboard scheme. Capacity Building - Career progression of two earlier stage researchers: • Dr Steve Smith: 'In 2020 I was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, my application for which was strengthen by having received VALIDATE funding for a project in my area of interest, as well as by the VALIDATE network overall and the the collaborations it has been possible to develop.' • Dr Barbara Kronsteiner-Dobramysl: 'Being a co-investigator on a VALIDATE Pump priming grant enables me to expand and demonstrate my expertise in Immunometabolism research, significantly contributing to establishing myself as an expert in this field.' • In addition, Prof Javier Sanchez-Garcia: 'Having two VALIDATE grants approved (one as participant and one as PI) has positively contributed in internal (Instituto Politécnico Nacional) academic evaluations; and this plus the expected publications and academic mobility of one of my students will have a good impact on the academic assessment of our Immunology postgraduate program as a whole.' - Two IPN (Mexico) PhD students participated in this VALIDATE project, one of who has successfully graduated in February 2021 (Angélica Pérez-Hernández) and continued to be employed on the project, funded by the P020 VALIDATE grant (instead of the originally planned 2-3 month lab exchange visit of a Mexican student to LSHTM, which could not take place due to the COVID pandemic). - Egle Butkeviciute, a Lithuanian PhD student at LSHTM, joined the project, and performed some mitochondrial dynamics experiments. - Perla Mariana Gutierréz Cedillo earned a M. Sc degree, working on "metabolic reprogramming to improve BCG vaccination-induced protection" , January 2023 - The acquisition of a small equipment (Countess 3, automated cell counter, Thermo Fisher) will have a positive long-term impact on the Immunoregulation Lab work in Mexico.
Start Year 2019
 
Description VALIDATE Pump-priming project P020 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Metabolic reprogramming of skin microenvironment for improved BCG vaccine efficacy Led by Prof Francisco Javier Sánchez-García (Instituto Politécnico Nacional, Mexico), with Dr Steven Smith (LSHTM originally, now Brunel University, UK), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Prof Hazel Dockrell (LSHTM, UK) Project Aims In theory, every single infectious disease could be eliminated from the earth if good vaccines (plus appropriate social and public health policies) are developed. In this scenario the best example is the vaccine against smallpox that lead to complete eradication of the disease by the year 1980. Other vaccines, however, have not been so successful, notoriously BCG. This vaccine, first used in humans in 1921, has greatly contributed to reducing TB, but still, TB remains the world's top infectious cause of mortality. In addition, BCG has proved to be protective in some countries but not in others. Why is that so? Several hypotheses have been put forward and, for practical purposes, this has conducted researchers to try to develop "better anti-tuberculosis vaccines". Here, we propose a different approach: "current BCG is an excellent vaccine, it contains most of the potentially protective antigens. Moreover, it contains its own natural adjuvants; the problem is that we have failed to deliver it into the appropriate metabolic tissue microenvironment". Recent research shows that the quality of immune cell effector functions are determined by their particular metabolism at the time (i.e. how they produce energy), and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumors. We propose to reprogram the local cell metabolism at the site (skin) where the BCG vaccine will be delivered, to ensure the best immune cell activation and consequently, the highest protection. Project Outcomes This project originated from the need to stop TB and from the fact that the BCG vaccine, used in humans since 1921, has proved to be protective against TB in some countries but not in others. We asked ourselves: why is that so? While BCG fulfils all the criteria of an excellent vaccine, containing most of the potentially protective antigens and its own natural adjuvants, we hypothesized that so far it has not been delivered into the appropriate metabolic microenvironment. This idea is supported by the recent finding that the quality of immune cell effector functions is largely determined by their particular metabolism at the time, and that the presence of selected metabolites is capable of modifying cell immune responses locally, such as in solid tumours. We proposed to metabolically pre-condition the local tissue microenvironment to potentiate antigen uptake/processing and trafficking upon the administration of the current BCG vaccine, in order to improve anti-TB protection. We selected five metabolites to be tested: lactate, succinate, fumarate acetate and butyrate, all of which harbour cell signalling properties. Based on our preliminary results, all were used at a final concentration of 100 uM. Firstly, we found structural and functional changes of mitochondria from human monocyte-derived macrophages in response to in vitro treatment with the aforementioned metabolites (100 uM). Fumarate induced mitochondrial fusion, polarization, and changes in mitochondrial cristae; in contrast, succinate, lactate and butyrate caused mitochondrial fission and depolarization. All the metabolites tested triggered an increase in cytoplasmic and mitochondrial calcium concentration within a few seconds, and fumarate promoted the highest calcium influx to the cytoplasm whereas the mitochondrial calcium concentrations, following metabolite treatment, were similar for all the metabolites tested. Furthermore, extracellular flux analysis of human monocytes demonstrated changes in the use of energy pathways upon metabolite treatment. Succinate treatment resulted in an immediate increase of extracellular acidification rate (ECAR), indicative of increased glycolysis, whereas no effect was observed for the other metabolites. Overnight pre- treatment with succinate led to a reduction in mitochondrial ATP production compared to vehicle, whereas fumarate treatment had the opposite effect. We then exposed human peripheral blood mononuclear cells (PBMC) to the different metabolites for 24 h and cultured the cells in the presence of BCG in order to assess mycobacterial growth inhibition by un-stimulated cells and by metabolite-stimulated cells. Results from six donors showed no statistically significant differences between treatments. Metabolite pre-treatment was able to change the expression of co-stimulatory (CD80, CD86) and co-inhibitory (PD-L1) molecules on the monocytic/macrophage cell line (RAW 264.7) upon stimulation with lipopolysaccharide (LPS), with succinate increasing activation as demonstrated by increased cell size and relative amount of CD80 compared to LPS alone. Preliminary data from in vivo mouse experiments demonstrate that combined injection (i.d.) of BCG and metabolite into the back of mice elicits histopathological changes and differences in immune cell composition as compared to BCG alone. Overall, our study highlights benefits of metabolite pre-treatment on activation, metabolic properties and function of monocytes/macrophages and suggests that metabolite-induced tissue metabolic reprogramming is feasible.
Impact Collaborations - Continuation of collaboration between Prof Javier Sanchez-Garcia (IPN, Mexico), Dr Steven Smith (LSHTM; now Brunel) and Prof Hazel Dockrell (LSHTM), that started with VALIDATE grant P004. - Project is a new collaboration, formed at VALIDATE's 2nd Annual Meeting, with Dr Barbara Kronsteiner-Dobramysl (University of Oxford). - Addition of Prof Rogelio Hernandez-Pando to the research team (National Institute of Medical Sciences and Nutrition; 2022) - In the course of the project a new collaboration with Professor Oscar Rojas-Espinosa (ENCB-IPN), a leading expert on murine leprosy, was established. - As a direct result of VALIDATE´s policy to encourage collaborations, Prof. Sánchez-García was invited (through VALIDATE) by Dr. Vijay Kumar Prajapati to write a chapter for a book on Vaccines: María Maximina Bertha Moreno-Altamirano, Jacqueline Oliva-Ramírez y, F. Javier Sánchez-García. "Circadian Rhythmicity and Vaccination" In: Vijay Kumar Prajapati (Ed.) System Vaccinology. (2022) Elsevier, Pp 207-229. - A new Collaboration with Professor Rogelio Hernández-Pando, Institito Nacional de Ciencias Medicas y de la Nutrición "Salvador Zubirán", Mexico City, was established, to evaluate the protective effect of BCG plus metabolites immunization in a murine model of M. tuberculosis infection. Work in progress in this area. Direct publications: Pérez-Hernández CA, Moreno-Altamirano MMB, López-Villegas EO, Butkeviciute E, Ali M, Kronsteiner B, Dunachie SJ, Dockrell HM, Smith SG, Sánchez-García, FJ. (2022). Mitochondrial Ultrastructure and Activity are Differentially Regulated by Glycolysis-, Krebs cycle-, and Microbiota-derived Metabolites in Monocytes. Biology 11: 1132. doi.org/10.3390/ biology11081132. Related publications crediting VALIDATE funding: Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno- Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ (2020) Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate-Induced Innate Immune Training. Front. Immunol. 11:1715. doi: 10.3389/fimmu.2020.01715. Prentice S, Dockrell HM. (2020). Antituberculosis BCG vaccination: more reasons for varying innate and adaptive immune responses. J Clin Invest. 130(10):5121-5123. doi: 10.1172/JCI141317. Prentice S, Dockrell HM. (2021). BCG-specific and non-specific effects: different questions, similar challenges. J Infect Dis. jiab307. doi: 10.1093/infdis/jiab307. Dockrell HM, Butkeviciute E. (2021). Can what have we learnt about BCG vaccination in the last 20 years help us to design a better tuberculosis vaccine? Vaccine. S0264- 410X(21)00109-2. doi: 10.1016/j.vaccine.2021.01.068. Moreno-Altamirano MMB, Oliva-Ramírez J, Sánchez-García FJ. (2021). Chapter 9: Circadian Rhythmicity and Vaccination. In: Systems Vaccination. Prajapati VK (Ed.) (In press). Paper published by two VALIDATE/project members: Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García, FJ. (2020). Mitochondria: an integrative hub coordinating circadian rhythms, metabolism, the microbiome, and immunity. Frontiers in Cell and Developmental Biology 8:51. doi: 10.3389/fcell.2020.00051. Engagement activities: International congress : Pérez-Hernández CA, Kern CC, Butkeviciute E, McCarthy E, Dockrell HM, Moreno-Altamirano MMB, Aguilar-López BA, Bhosale G, Wang H, Gems D, Duchen MR, Smith SG and Sánchez-García FJ. Mitochondrial Signature in Human Monocytes and Resistance to Infection in C. elegans During Fumarate Induced Innate Immune Training. 11th World Congress on Targeting Mitochondria (Germany, virtual congress), October 28-30, 2020. The project was also presented to VALIDATE members at the 2019 & 2020 Annual Meeting, and Dr Barbara Kronsteiner-Dobramysl also presented a talk "Diabetes alters cellular metabolism of T cells: implications for vaccine development" at the 2019 VALIDATE-BSI Conference. Grant applications: - A grant application (Newton Fund-The Ministry of Education, Science, Technology and Innovation of Mexico City) aimed at analysing the metabolic basis and trained immunity potential of Covid-19 has been jointly submitted by Steve Smith and F. Javier Sánchez-García, as PIs, along with other colleagues from UK and Mexico. Outcome pending. - Steve Smith submitted a grant application to the Academy of Medical Sciences Springboard scheme. Capacity Building - Career progression of two earlier stage researchers: • Dr Steve Smith: 'In 2020 I was appointed Senior Lecturer in Biomedical Sciences at Brunel University London, my application for which was strengthen by having received VALIDATE funding for a project in my area of interest, as well as by the VALIDATE network overall and the the collaborations it has been possible to develop.' • Dr Barbara Kronsteiner-Dobramysl: 'Being a co-investigator on a VALIDATE Pump priming grant enables me to expand and demonstrate my expertise in Immunometabolism research, significantly contributing to establishing myself as an expert in this field.' • In addition, Prof Javier Sanchez-Garcia: 'Having two VALIDATE grants approved (one as participant and one as PI) has positively contributed in internal (Instituto Politécnico Nacional) academic evaluations; and this plus the expected publications and academic mobility of one of my students will have a good impact on the academic assessment of our Immunology postgraduate program as a whole.' - Two IPN (Mexico) PhD students participated in this VALIDATE project, one of who has successfully graduated in February 2021 (Angélica Pérez-Hernández) and continued to be employed on the project, funded by the P020 VALIDATE grant (instead of the originally planned 2-3 month lab exchange visit of a Mexican student to LSHTM, which could not take place due to the COVID pandemic). - Egle Butkeviciute, a Lithuanian PhD student at LSHTM, joined the project, and performed some mitochondrial dynamics experiments. - Perla Mariana Gutierréz Cedillo earned a M. Sc degree, working on "metabolic reprogramming to improve BCG vaccination-induced protection" , January 2023 - The acquisition of a small equipment (Countess 3, automated cell counter, Thermo Fisher) will have a positive long-term impact on the Immunoregulation Lab work in Mexico.
Start Year 2019
 
Description VALIDATE Pump-priming project P021 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Development of an RNA based vaccine against Mycobacterium tuberculosis Led by Affil Prof Rhea Coler (IDRI; now University of Washington, USA), with Prof Helen Fletcher (LSHTM, UK) Project Aims Tuberculosis (TB) now ranks as the leading infectious killer worldwide, surpassing HIV/AIDS and malaria for the last several years in a row. The bacterium that causes TB disease, Mycobacterium tuberculosis (Mtb), is typically transmitted by inhaling the bacteria from infected people. These bacteria infect one-quarter of the world's population, causing disease in ~10.4 million people and resulting in ~1.6 million deaths each year. Therefore, new TB vaccines represent a critical, unmet global public health need. A new vaccine technology, based on the delivery of self-replicating RNA molecules, has gained attention worldwide but there has been limited exploration of this vaccine technology in protection against TB. IDRI has adapted their ID93 vaccine antigen into a self-replicating RNA molecule formulated in a state-of-the-art nanostructured lipid carrier. This enables the RNA vaccine candidate to be compared head-to-head with the classical ID93 protein with adjuvant vaccine, currently in phase 2b trials. Comparison of the nucleic acid based ID93 with the classical protein adjuvant formulation of ID93 will accelerate the development of the RNA based vaccine and provide insight into the breadth and magnitude of immune response induced by nucleic acid versus protein based vaccines. We will evaluate ability of these vaccine candidates to reduce bacterial burden in the mouse model along with the accompanying immune responses. Recent clinical trial data have uncovered sets of host biomarkers that identify people who are more likely to advance to active disease states. This proposal aims to evaluate these clinically defined host markers (post-Mtb challenge) in a preclinical model of TB and to determine if immunization with two different types of vaccine candidates influences these host risk signatures. Successful completion of this proposal would significantly advance the TB vaccine pipeline and has the potential to streamline efforts and resources for the most promising candidates. Project Outcomes A new vaccine technology, based on the delivery of RNA molecules, has gained attention worldwide but there has been limited exploration of this vaccine technology in protection against Tuberculosis (TB). We have designed TB vaccine candidate antigens into self-replicating RNA molecules with a state of the art delivery formulation. This enabled the RNA vaccine candidates to be compared head-to-head with the classical protein with adjuvant vaccines of the same antigenic composition. In this work we observed the lung-based mycobacterial growth inhibition assay (MGIA) corresponded well with traditional in vivo challenge studies examining efficacy by plating organ homogenates. By both efficacy measures we observed homologous prime-boost regimens afford greater protection from Mycobacterium tuberculosis (Mtb) challenge than heterologous regimens, albeit these studies are preliminary. We did not observe safety signals following prime or boost RNA immunizations, and consider this RNA platform and delivery formulation well tolerated in the preclinical mouse model. Due to challenges outside of our control, we were unable to in tandem assess immunogenicity induced by the two vaccine platforms as homologous or heterologous prime-boost strategies nor evaluate RNA host correlate of risk (COR) signatures. Fortunately, these preliminary studies have led to follow-on funding which we are leveraging to complete the full suite of efficacy, immunology and biomarker endpoints for our down selected protein + adjuvant and RNA-based vaccine candidates. Specifically, we will characterize the safety, immunogenicity and protective efficacy of the RNA vaccine expressing Mtb candidate antigens, complexed with Lipid Inorganic Nanoparticle (LION) formulations in a preclinical animal model of Mtb infection. We will also assess the ability of human COR signatures to predict disease burden in animals challenged with Mtb after these prophylactic vaccine strategies. Successfully continuing this work would significantly advance the TB vaccine pipeline and has the potential to streamline efforts and resources for the most promising candidates.
Impact - This project is a new collaboration between Affil Prof Rhea Coler and Prof Helen Fletcher and generated novel hypotheses. - The collaboration also extended to industry partners HDT Bio (Seattle WA) who have developed a state of the art Lipid InOrganic Nanoparticle (LION) formulation for high-efficiency RNA delivery in vivo. We have engaged these industry partners as a reliable source of formulation for our ongoing and future studies. - Secured an additional small research grant from the Firland Foundation (Seattle, WA), in the sum of US$50,000 to repeat essential preclinical experiments on an accurate timeline with full immunologic outcome analysis. These experiments are currently ongoing. - Submission of an NIH R01-level grant to continue and expand this research. Proposal title 'Modifiers, kinetics and limitations of transcriptional TB disease progression biomarkers (01.08.21-31.07.2026). This application also included leaders in biomarker research from the Fred Hutch Cancer Research Center (Seattle, WA), Dr Andrew Fiore-Gartland, expanding the collaborative efforts and connections of the project. Preliminary data generated in this project are being included in additional grant applications by other members of the Coler lab, as proof of concept for MGIA success. - Partly based on her leadership on this project and follow-on funding applications, Dr Sasha Larsen Akins was promoted to Research Scientist III at SCRI. - Dr Fletcher's LSHTM graduate student, Hannah Painter, completed an abbreviated placement in Seattle WA at SCRI. During this placement she helped complete the technology transfer of the lung-based mycobacterial growth inhibition assay to the Coler research team. Her placement was abbreviated due to the COVID-19 pandemic early in 2020. This resulted in an official SOP adapted for SCRI workflows and research questions. - The Coler lab has also trained other SCRI staff on the use of the BACTEC MGIT workflow and instrument after receiving training from the Fletcher lab, which has furthered an atmosphere of collaboration across the institutes. - Hannah Painter (LSHTM) and Dr Sasha Larsen Akins (SCRI) have continued their working partnership and have an article in preparation describing the human correlate of risk biomarker signature application in preclinical models and reanalysis of these data sets. As ECRs they have had the opportunity to advance their project management and collaboration skills through the execution of this project. - Results from this project were presented to VALIDATE members at the 2019 Annual Meeting.
Start Year 2019
 
Description VALIDATE Pump-priming project P021 
Organisation University of Washington
Country United States 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Development of an RNA based vaccine against Mycobacterium tuberculosis Led by Affil Prof Rhea Coler (IDRI; now University of Washington, USA), with Prof Helen Fletcher (LSHTM, UK) Project Aims Tuberculosis (TB) now ranks as the leading infectious killer worldwide, surpassing HIV/AIDS and malaria for the last several years in a row. The bacterium that causes TB disease, Mycobacterium tuberculosis (Mtb), is typically transmitted by inhaling the bacteria from infected people. These bacteria infect one-quarter of the world's population, causing disease in ~10.4 million people and resulting in ~1.6 million deaths each year. Therefore, new TB vaccines represent a critical, unmet global public health need. A new vaccine technology, based on the delivery of self-replicating RNA molecules, has gained attention worldwide but there has been limited exploration of this vaccine technology in protection against TB. IDRI has adapted their ID93 vaccine antigen into a self-replicating RNA molecule formulated in a state-of-the-art nanostructured lipid carrier. This enables the RNA vaccine candidate to be compared head-to-head with the classical ID93 protein with adjuvant vaccine, currently in phase 2b trials. Comparison of the nucleic acid based ID93 with the classical protein adjuvant formulation of ID93 will accelerate the development of the RNA based vaccine and provide insight into the breadth and magnitude of immune response induced by nucleic acid versus protein based vaccines. We will evaluate ability of these vaccine candidates to reduce bacterial burden in the mouse model along with the accompanying immune responses. Recent clinical trial data have uncovered sets of host biomarkers that identify people who are more likely to advance to active disease states. This proposal aims to evaluate these clinically defined host markers (post-Mtb challenge) in a preclinical model of TB and to determine if immunization with two different types of vaccine candidates influences these host risk signatures. Successful completion of this proposal would significantly advance the TB vaccine pipeline and has the potential to streamline efforts and resources for the most promising candidates. Project Outcomes A new vaccine technology, based on the delivery of RNA molecules, has gained attention worldwide but there has been limited exploration of this vaccine technology in protection against Tuberculosis (TB). We have designed TB vaccine candidate antigens into self-replicating RNA molecules with a state of the art delivery formulation. This enabled the RNA vaccine candidates to be compared head-to-head with the classical protein with adjuvant vaccines of the same antigenic composition. In this work we observed the lung-based mycobacterial growth inhibition assay (MGIA) corresponded well with traditional in vivo challenge studies examining efficacy by plating organ homogenates. By both efficacy measures we observed homologous prime-boost regimens afford greater protection from Mycobacterium tuberculosis (Mtb) challenge than heterologous regimens, albeit these studies are preliminary. We did not observe safety signals following prime or boost RNA immunizations, and consider this RNA platform and delivery formulation well tolerated in the preclinical mouse model. Due to challenges outside of our control, we were unable to in tandem assess immunogenicity induced by the two vaccine platforms as homologous or heterologous prime-boost strategies nor evaluate RNA host correlate of risk (COR) signatures. Fortunately, these preliminary studies have led to follow-on funding which we are leveraging to complete the full suite of efficacy, immunology and biomarker endpoints for our down selected protein + adjuvant and RNA-based vaccine candidates. Specifically, we will characterize the safety, immunogenicity and protective efficacy of the RNA vaccine expressing Mtb candidate antigens, complexed with Lipid Inorganic Nanoparticle (LION) formulations in a preclinical animal model of Mtb infection. We will also assess the ability of human COR signatures to predict disease burden in animals challenged with Mtb after these prophylactic vaccine strategies. Successfully continuing this work would significantly advance the TB vaccine pipeline and has the potential to streamline efforts and resources for the most promising candidates.
Impact - This project is a new collaboration between Affil Prof Rhea Coler and Prof Helen Fletcher and generated novel hypotheses. - The collaboration also extended to industry partners HDT Bio (Seattle WA) who have developed a state of the art Lipid InOrganic Nanoparticle (LION) formulation for high-efficiency RNA delivery in vivo. We have engaged these industry partners as a reliable source of formulation for our ongoing and future studies. - Secured an additional small research grant from the Firland Foundation (Seattle, WA), in the sum of US$50,000 to repeat essential preclinical experiments on an accurate timeline with full immunologic outcome analysis. These experiments are currently ongoing. - Submission of an NIH R01-level grant to continue and expand this research. Proposal title 'Modifiers, kinetics and limitations of transcriptional TB disease progression biomarkers (01.08.21-31.07.2026). This application also included leaders in biomarker research from the Fred Hutch Cancer Research Center (Seattle, WA), Dr Andrew Fiore-Gartland, expanding the collaborative efforts and connections of the project. Preliminary data generated in this project are being included in additional grant applications by other members of the Coler lab, as proof of concept for MGIA success. - Partly based on her leadership on this project and follow-on funding applications, Dr Sasha Larsen Akins was promoted to Research Scientist III at SCRI. - Dr Fletcher's LSHTM graduate student, Hannah Painter, completed an abbreviated placement in Seattle WA at SCRI. During this placement she helped complete the technology transfer of the lung-based mycobacterial growth inhibition assay to the Coler research team. Her placement was abbreviated due to the COVID-19 pandemic early in 2020. This resulted in an official SOP adapted for SCRI workflows and research questions. - The Coler lab has also trained other SCRI staff on the use of the BACTEC MGIT workflow and instrument after receiving training from the Fletcher lab, which has furthered an atmosphere of collaboration across the institutes. - Hannah Painter (LSHTM) and Dr Sasha Larsen Akins (SCRI) have continued their working partnership and have an article in preparation describing the human correlate of risk biomarker signature application in preclinical models and reanalysis of these data sets. As ECRs they have had the opportunity to advance their project management and collaboration skills through the execution of this project. - Results from this project were presented to VALIDATE members at the 2019 Annual Meeting.
Start Year 2019
 
Description VALIDATE Pump-priming project P022 
Organisation Mahidol Oxford Tropical Medicine Research Unit
Country Thailand 
Sector Public 
PI Contribution VALIDATE funded project
Collaborator Contribution Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis Led by Dr Elena Stylianou (University of Oxford, UK), with Prof Helen McShane (University of Oxford, UK), Assoc Prof Susanna Dunachie (University of Oxford, UK), Assoc Prof Paul Brett (University of Nevada, USA), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Dr Panjaporn Chaichana (MORU, Thailand) Project Aims The increased susceptibility of individuals with diabetes mellitus (DM) to tuberculosis (TB) and melioidosis is well established. However there is limited understanding of the immune mechanisms behind this susceptibility, which is mainly due to an incomplete understanding of the protective immunity against both Mycobacterium tuberculosis (M.tb) and Burkholderia pseudomellei (B.p). Similarly, very little is known about vaccine efficacy in this susceptible diabetic population. I plan to establish diabetic mouse models to investigate the effect of DM on BCG, the only licensed vaccine against TB, and two promising vaccine candidates against M.tb and B.p. I will use two complementary diabetic models that will best represent human Type 2 diabetes. First I will transfer these two models, already established in Oxford by other groups, into our laboratory. Then, animals will be vaccinated and vaccine immunogenicity and efficacy will be evaluated in control and hyperglycemic mice. In addition, this work will determine how immune cells from diabetic mice differ from control mice in their ability to generate energy in response to a vaccine. Defining the differences in the induction of immune responses between DM and health is essential for TB and melioidosis vaccines currently in development, as these vaccines need to be effective in people that have, or may develop DM in the future. Project Outcomes Diabetes has been shown to alter host immune responses but there is limited information on how it affects vaccines. I established two complementary mouse models of hyperglycaemia that best represent human type 2 diabetes to study its effects on TB and melioidosis vaccine responses. In one model, animals are fed high fat diet and in the second model, mice become hyperglycaemic upon induction. Data generated showed that animals on higher fat diets were able to better control mycobacteria in vitro, compared to mice on control diets. In addition, both vaccine immune responses and efficacy were improved with higher fat diets. On-going work is investigating whether this is also true for hyperglycaemic transgenic mice. Further work will explore the in vivo efficacy of these models. Elena and Barbara had maternity leave in 2020/2021. Work also had to pause in March 2020 due to the COVID-19 pandemic, so this project was awarded a no-cost extension.
Impact This grant is led by an ECR, Dr Elena Stylianou, progressing her career. This project is a new collaboration between Elena Stylianou and the rest of the researchers (excluding Helen McShane). Various grant applications have been made to continue this work.
Start Year 2019
 
Description VALIDATE Pump-priming project P022 
Organisation University of Nevada, Reno School of Medicine
Country United States 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis Led by Dr Elena Stylianou (University of Oxford, UK), with Prof Helen McShane (University of Oxford, UK), Assoc Prof Susanna Dunachie (University of Oxford, UK), Assoc Prof Paul Brett (University of Nevada, USA), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Dr Panjaporn Chaichana (MORU, Thailand) Project Aims The increased susceptibility of individuals with diabetes mellitus (DM) to tuberculosis (TB) and melioidosis is well established. However there is limited understanding of the immune mechanisms behind this susceptibility, which is mainly due to an incomplete understanding of the protective immunity against both Mycobacterium tuberculosis (M.tb) and Burkholderia pseudomellei (B.p). Similarly, very little is known about vaccine efficacy in this susceptible diabetic population. I plan to establish diabetic mouse models to investigate the effect of DM on BCG, the only licensed vaccine against TB, and two promising vaccine candidates against M.tb and B.p. I will use two complementary diabetic models that will best represent human Type 2 diabetes. First I will transfer these two models, already established in Oxford by other groups, into our laboratory. Then, animals will be vaccinated and vaccine immunogenicity and efficacy will be evaluated in control and hyperglycemic mice. In addition, this work will determine how immune cells from diabetic mice differ from control mice in their ability to generate energy in response to a vaccine. Defining the differences in the induction of immune responses between DM and health is essential for TB and melioidosis vaccines currently in development, as these vaccines need to be effective in people that have, or may develop DM in the future. Project Outcomes Diabetes has been shown to alter host immune responses but there is limited information on how it affects vaccines. I established two complementary mouse models of hyperglycaemia that best represent human type 2 diabetes to study its effects on TB and melioidosis vaccine responses. In one model, animals are fed high fat diet and in the second model, mice become hyperglycaemic upon induction. Data generated showed that animals on higher fat diets were able to better control mycobacteria in vitro, compared to mice on control diets. In addition, both vaccine immune responses and efficacy were improved with higher fat diets. On-going work is investigating whether this is also true for hyperglycaemic transgenic mice. Further work will explore the in vivo efficacy of these models. Elena and Barbara had maternity leave in 2020/2021. Work also had to pause in March 2020 due to the COVID-19 pandemic, so this project was awarded a no-cost extension.
Impact This grant is led by an ECR, Dr Elena Stylianou, progressing her career. This project is a new collaboration between Elena Stylianou and the rest of the researchers (excluding Helen McShane). Various grant applications have been made to continue this work.
Start Year 2019
 
Description VALIDATE Pump-priming project P022 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis Led by Dr Elena Stylianou (University of Oxford, UK), with Prof Helen McShane (University of Oxford, UK), Assoc Prof Susanna Dunachie (University of Oxford, UK), Assoc Prof Paul Brett (University of Nevada, USA), Dr Barbara Kronsteiner-Dobramysl (University of Oxford, UK) and Dr Panjaporn Chaichana (MORU, Thailand) Project Aims The increased susceptibility of individuals with diabetes mellitus (DM) to tuberculosis (TB) and melioidosis is well established. However there is limited understanding of the immune mechanisms behind this susceptibility, which is mainly due to an incomplete understanding of the protective immunity against both Mycobacterium tuberculosis (M.tb) and Burkholderia pseudomellei (B.p). Similarly, very little is known about vaccine efficacy in this susceptible diabetic population. I plan to establish diabetic mouse models to investigate the effect of DM on BCG, the only licensed vaccine against TB, and two promising vaccine candidates against M.tb and B.p. I will use two complementary diabetic models that will best represent human Type 2 diabetes. First I will transfer these two models, already established in Oxford by other groups, into our laboratory. Then, animals will be vaccinated and vaccine immunogenicity and efficacy will be evaluated in control and hyperglycemic mice. In addition, this work will determine how immune cells from diabetic mice differ from control mice in their ability to generate energy in response to a vaccine. Defining the differences in the induction of immune responses between DM and health is essential for TB and melioidosis vaccines currently in development, as these vaccines need to be effective in people that have, or may develop DM in the future. Project Outcomes Diabetes has been shown to alter host immune responses but there is limited information on how it affects vaccines. I established two complementary mouse models of hyperglycaemia that best represent human type 2 diabetes to study its effects on TB and melioidosis vaccine responses. In one model, animals are fed high fat diet and in the second model, mice become hyperglycaemic upon induction. Data generated showed that animals on higher fat diets were able to better control mycobacteria in vitro, compared to mice on control diets. In addition, both vaccine immune responses and efficacy were improved with higher fat diets. On-going work is investigating whether this is also true for hyperglycaemic transgenic mice. Further work will explore the in vivo efficacy of these models. Elena and Barbara had maternity leave in 2020/2021. Work also had to pause in March 2020 due to the COVID-19 pandemic, so this project was awarded a no-cost extension.
Impact This grant is led by an ECR, Dr Elena Stylianou, progressing her career. This project is a new collaboration between Elena Stylianou and the rest of the researchers (excluding Helen McShane). Various grant applications have been made to continue this work.
Start Year 2019
 
Description VALIDATE Pump-priming project P023 
Organisation University College Dublin
Country Ireland 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Elucidating the T-cell epitopes and T-cells responses of two B. pseudomallei vaccine antigens Led by Dr Julen Tomás Cortázar (University College Dublin, Ireland), with Prof Susanna Dunachie (University of Oxford, UK), Asst Prof Louise Gourlay (University of Milan, Italy), Prof Giorgio Colombo (University of Pavia, Italy), and Prof Siobhán McClean (University College Dublin, Ireland) Project Aims Melioidosis is a potential lethal infection that is quite common in South East Asia and is on the increase in other tropical countries such as Brazil. It can kill up to 40% of people and is particularly dangerous for people with diabetes. The bacteria that cause melioidosis are extremely resistant to antibiotics and there is no vaccine available to protect people from this infection. We want to develop a vaccine that will be safe, effective and cheap to produce. As a result of several years studying how bacteria attach to human cells, we have discovered some molecules (called antigens) that are likely to be good vaccines as they protect mice from this infection. More recently we have discovered two more antigens that we would like to study in this proposal to see if they would be effective in protecting people from melioidosis. There are two arms to the immune system: one requiring antibodies for effect and the other relying on T cells for effect. In the case of melioidosis the T-cell response is essential in successfully fighting this infection. In this proposal we will try to pinpoint the parts of the antigen molecules that are most likely to stimulate the arm of immune system that is most important to protect people from melioidosis infection. We will identify the parts of the antigen that stimulate the T-cell response. We will also look at the level of T cell response in mice and compare that to the T-cell responses in human blood. Project Outcomes The in silico prediction of T cell immunogenic peptides from the PDB structure of PA26 antigen revealed four different epitopes, located on the surface of the protein and thus, also accessible to antibodies. Unfortunately, one predicted epitope was highly hydrophobic and could not be synthesized in free peptide form. Immunization of mice with PA26 resulted in a weaker antibody response relative to that induced by the protective BpOmpW antigen. T cell responses against PA26 were moderate, and lower in comparison with protective antigen BpOmpW. Nevertheless, we observed elevated protective IFN-y responses relative to the control group. Insulin resistance altered the immune response against PA26, impairing Th2 response and regulatory T cells. Although high-resolution X-ray data were collected for the LP20 antigen, the crystal structure could not be solved, therefore we could not carry out epitope predictions on this antigen. Immunization of mice with LP20 antigen produced a robust antibody response, but poor T cell responses. In contrast, it elicited different Foxp3+ regulatory cell types. Human sera from melioidosis survivors recognised BpOmpW but did not recognize any of the antigens, indicating that they do not participate in the immune response to melioidosis. [This project was impacted by COVID-19 lab closures during lockdown and granted a no-cost extension].
Impact This project is a new collaboration between UCD and Dr Louise Gourlay and Prof Susanna Dunachie, formed for this pump priming grant. The UCD team are also collaborating with Dr Paul Brett and Dr Mary Burtnick (Nevada) since meeting at the 2019 VALIDATE Annual Meeting. Recent studies performed at U. Reno Nevada showed that BpOmpW showed very strong immunogenicity when immunized with CPS-CRM197. Our objective is to publish one or two more papers with PA26 and LP20 antigens. Also, we plan to publish or patent the immunophenotyping method of 15-colours panel antibody for Flow Cytometry in collaboration with Alfonso Blanco, the coordinator of Flow Cytometry Core in UCD Conway Institute. To get the VALIDATE pump priming grant as a PI improved Dr Julen Tomas Cortazar's CV and as a result he was awarded a Basque Government Fellowship in December 2019 enabling him to continue his research in Dublin for two more years. Furthermore, leading this project as the principal investigator allowed Julen to set up his own collaborative network and to interact directly with Drs Louise Gourlay, Giorgio Colombo and Susanna Dunachie. Invention disclosures for these new vaccine antigens have been written, while the BpOmpW vaccine has been patented. This project was presented at the VALIDATE 2019 & 2020 Annual Meeting, raising the profile of the project and PI within the Network. Julen and Siobhan also presented some of the project's results as an online VALIDATE Summer Seminar in 2020 to the VALIDATE community. As outreach, Julen created videos in three different languages (English, Spanish, Basque) talking about his research in vaccine development as part of the VALIDATE Outreach Week in December.
Start Year 2019
 
Description VALIDATE Pump-priming project P023 
Organisation University of Milan
Country Italy 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Elucidating the T-cell epitopes and T-cells responses of two B. pseudomallei vaccine antigens Led by Dr Julen Tomás Cortázar (University College Dublin, Ireland), with Prof Susanna Dunachie (University of Oxford, UK), Asst Prof Louise Gourlay (University of Milan, Italy), Prof Giorgio Colombo (University of Pavia, Italy), and Prof Siobhán McClean (University College Dublin, Ireland) Project Aims Melioidosis is a potential lethal infection that is quite common in South East Asia and is on the increase in other tropical countries such as Brazil. It can kill up to 40% of people and is particularly dangerous for people with diabetes. The bacteria that cause melioidosis are extremely resistant to antibiotics and there is no vaccine available to protect people from this infection. We want to develop a vaccine that will be safe, effective and cheap to produce. As a result of several years studying how bacteria attach to human cells, we have discovered some molecules (called antigens) that are likely to be good vaccines as they protect mice from this infection. More recently we have discovered two more antigens that we would like to study in this proposal to see if they would be effective in protecting people from melioidosis. There are two arms to the immune system: one requiring antibodies for effect and the other relying on T cells for effect. In the case of melioidosis the T-cell response is essential in successfully fighting this infection. In this proposal we will try to pinpoint the parts of the antigen molecules that are most likely to stimulate the arm of immune system that is most important to protect people from melioidosis infection. We will identify the parts of the antigen that stimulate the T-cell response. We will also look at the level of T cell response in mice and compare that to the T-cell responses in human blood. Project Outcomes The in silico prediction of T cell immunogenic peptides from the PDB structure of PA26 antigen revealed four different epitopes, located on the surface of the protein and thus, also accessible to antibodies. Unfortunately, one predicted epitope was highly hydrophobic and could not be synthesized in free peptide form. Immunization of mice with PA26 resulted in a weaker antibody response relative to that induced by the protective BpOmpW antigen. T cell responses against PA26 were moderate, and lower in comparison with protective antigen BpOmpW. Nevertheless, we observed elevated protective IFN-y responses relative to the control group. Insulin resistance altered the immune response against PA26, impairing Th2 response and regulatory T cells. Although high-resolution X-ray data were collected for the LP20 antigen, the crystal structure could not be solved, therefore we could not carry out epitope predictions on this antigen. Immunization of mice with LP20 antigen produced a robust antibody response, but poor T cell responses. In contrast, it elicited different Foxp3+ regulatory cell types. Human sera from melioidosis survivors recognised BpOmpW but did not recognize any of the antigens, indicating that they do not participate in the immune response to melioidosis. [This project was impacted by COVID-19 lab closures during lockdown and granted a no-cost extension].
Impact This project is a new collaboration between UCD and Dr Louise Gourlay and Prof Susanna Dunachie, formed for this pump priming grant. The UCD team are also collaborating with Dr Paul Brett and Dr Mary Burtnick (Nevada) since meeting at the 2019 VALIDATE Annual Meeting. Recent studies performed at U. Reno Nevada showed that BpOmpW showed very strong immunogenicity when immunized with CPS-CRM197. Our objective is to publish one or two more papers with PA26 and LP20 antigens. Also, we plan to publish or patent the immunophenotyping method of 15-colours panel antibody for Flow Cytometry in collaboration with Alfonso Blanco, the coordinator of Flow Cytometry Core in UCD Conway Institute. To get the VALIDATE pump priming grant as a PI improved Dr Julen Tomas Cortazar's CV and as a result he was awarded a Basque Government Fellowship in December 2019 enabling him to continue his research in Dublin for two more years. Furthermore, leading this project as the principal investigator allowed Julen to set up his own collaborative network and to interact directly with Drs Louise Gourlay, Giorgio Colombo and Susanna Dunachie. Invention disclosures for these new vaccine antigens have been written, while the BpOmpW vaccine has been patented. This project was presented at the VALIDATE 2019 & 2020 Annual Meeting, raising the profile of the project and PI within the Network. Julen and Siobhan also presented some of the project's results as an online VALIDATE Summer Seminar in 2020 to the VALIDATE community. As outreach, Julen created videos in three different languages (English, Spanish, Basque) talking about his research in vaccine development as part of the VALIDATE Outreach Week in December.
Start Year 2019
 
Description VALIDATE Pump-priming project P023 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Elucidating the T-cell epitopes and T-cells responses of two B. pseudomallei vaccine antigens Led by Dr Julen Tomás Cortázar (University College Dublin, Ireland), with Prof Susanna Dunachie (University of Oxford, UK), Asst Prof Louise Gourlay (University of Milan, Italy), Prof Giorgio Colombo (University of Pavia, Italy), and Prof Siobhán McClean (University College Dublin, Ireland) Project Aims Melioidosis is a potential lethal infection that is quite common in South East Asia and is on the increase in other tropical countries such as Brazil. It can kill up to 40% of people and is particularly dangerous for people with diabetes. The bacteria that cause melioidosis are extremely resistant to antibiotics and there is no vaccine available to protect people from this infection. We want to develop a vaccine that will be safe, effective and cheap to produce. As a result of several years studying how bacteria attach to human cells, we have discovered some molecules (called antigens) that are likely to be good vaccines as they protect mice from this infection. More recently we have discovered two more antigens that we would like to study in this proposal to see if they would be effective in protecting people from melioidosis. There are two arms to the immune system: one requiring antibodies for effect and the other relying on T cells for effect. In the case of melioidosis the T-cell response is essential in successfully fighting this infection. In this proposal we will try to pinpoint the parts of the antigen molecules that are most likely to stimulate the arm of immune system that is most important to protect people from melioidosis infection. We will identify the parts of the antigen that stimulate the T-cell response. We will also look at the level of T cell response in mice and compare that to the T-cell responses in human blood. Project Outcomes The in silico prediction of T cell immunogenic peptides from the PDB structure of PA26 antigen revealed four different epitopes, located on the surface of the protein and thus, also accessible to antibodies. Unfortunately, one predicted epitope was highly hydrophobic and could not be synthesized in free peptide form. Immunization of mice with PA26 resulted in a weaker antibody response relative to that induced by the protective BpOmpW antigen. T cell responses against PA26 were moderate, and lower in comparison with protective antigen BpOmpW. Nevertheless, we observed elevated protective IFN-y responses relative to the control group. Insulin resistance altered the immune response against PA26, impairing Th2 response and regulatory T cells. Although high-resolution X-ray data were collected for the LP20 antigen, the crystal structure could not be solved, therefore we could not carry out epitope predictions on this antigen. Immunization of mice with LP20 antigen produced a robust antibody response, but poor T cell responses. In contrast, it elicited different Foxp3+ regulatory cell types. Human sera from melioidosis survivors recognised BpOmpW but did not recognize any of the antigens, indicating that they do not participate in the immune response to melioidosis. [This project was impacted by COVID-19 lab closures during lockdown and granted a no-cost extension].
Impact This project is a new collaboration between UCD and Dr Louise Gourlay and Prof Susanna Dunachie, formed for this pump priming grant. The UCD team are also collaborating with Dr Paul Brett and Dr Mary Burtnick (Nevada) since meeting at the 2019 VALIDATE Annual Meeting. Recent studies performed at U. Reno Nevada showed that BpOmpW showed very strong immunogenicity when immunized with CPS-CRM197. Our objective is to publish one or two more papers with PA26 and LP20 antigens. Also, we plan to publish or patent the immunophenotyping method of 15-colours panel antibody for Flow Cytometry in collaboration with Alfonso Blanco, the coordinator of Flow Cytometry Core in UCD Conway Institute. To get the VALIDATE pump priming grant as a PI improved Dr Julen Tomas Cortazar's CV and as a result he was awarded a Basque Government Fellowship in December 2019 enabling him to continue his research in Dublin for two more years. Furthermore, leading this project as the principal investigator allowed Julen to set up his own collaborative network and to interact directly with Drs Louise Gourlay, Giorgio Colombo and Susanna Dunachie. Invention disclosures for these new vaccine antigens have been written, while the BpOmpW vaccine has been patented. This project was presented at the VALIDATE 2019 & 2020 Annual Meeting, raising the profile of the project and PI within the Network. Julen and Siobhan also presented some of the project's results as an online VALIDATE Summer Seminar in 2020 to the VALIDATE community. As outreach, Julen created videos in three different languages (English, Spanish, Basque) talking about his research in vaccine development as part of the VALIDATE Outreach Week in December.
Start Year 2019
 
Description VALIDATE Pump-priming project P023 
Organisation University of Pavia
Country Italy 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Elucidating the T-cell epitopes and T-cells responses of two B. pseudomallei vaccine antigens Led by Dr Julen Tomás Cortázar (University College Dublin, Ireland), with Prof Susanna Dunachie (University of Oxford, UK), Asst Prof Louise Gourlay (University of Milan, Italy), Prof Giorgio Colombo (University of Pavia, Italy), and Prof Siobhán McClean (University College Dublin, Ireland) Project Aims Melioidosis is a potential lethal infection that is quite common in South East Asia and is on the increase in other tropical countries such as Brazil. It can kill up to 40% of people and is particularly dangerous for people with diabetes. The bacteria that cause melioidosis are extremely resistant to antibiotics and there is no vaccine available to protect people from this infection. We want to develop a vaccine that will be safe, effective and cheap to produce. As a result of several years studying how bacteria attach to human cells, we have discovered some molecules (called antigens) that are likely to be good vaccines as they protect mice from this infection. More recently we have discovered two more antigens that we would like to study in this proposal to see if they would be effective in protecting people from melioidosis. There are two arms to the immune system: one requiring antibodies for effect and the other relying on T cells for effect. In the case of melioidosis the T-cell response is essential in successfully fighting this infection. In this proposal we will try to pinpoint the parts of the antigen molecules that are most likely to stimulate the arm of immune system that is most important to protect people from melioidosis infection. We will identify the parts of the antigen that stimulate the T-cell response. We will also look at the level of T cell response in mice and compare that to the T-cell responses in human blood. Project Outcomes The in silico prediction of T cell immunogenic peptides from the PDB structure of PA26 antigen revealed four different epitopes, located on the surface of the protein and thus, also accessible to antibodies. Unfortunately, one predicted epitope was highly hydrophobic and could not be synthesized in free peptide form. Immunization of mice with PA26 resulted in a weaker antibody response relative to that induced by the protective BpOmpW antigen. T cell responses against PA26 were moderate, and lower in comparison with protective antigen BpOmpW. Nevertheless, we observed elevated protective IFN-y responses relative to the control group. Insulin resistance altered the immune response against PA26, impairing Th2 response and regulatory T cells. Although high-resolution X-ray data were collected for the LP20 antigen, the crystal structure could not be solved, therefore we could not carry out epitope predictions on this antigen. Immunization of mice with LP20 antigen produced a robust antibody response, but poor T cell responses. In contrast, it elicited different Foxp3+ regulatory cell types. Human sera from melioidosis survivors recognised BpOmpW but did not recognize any of the antigens, indicating that they do not participate in the immune response to melioidosis. [This project was impacted by COVID-19 lab closures during lockdown and granted a no-cost extension].
Impact This project is a new collaboration between UCD and Dr Louise Gourlay and Prof Susanna Dunachie, formed for this pump priming grant. The UCD team are also collaborating with Dr Paul Brett and Dr Mary Burtnick (Nevada) since meeting at the 2019 VALIDATE Annual Meeting. Recent studies performed at U. Reno Nevada showed that BpOmpW showed very strong immunogenicity when immunized with CPS-CRM197. Our objective is to publish one or two more papers with PA26 and LP20 antigens. Also, we plan to publish or patent the immunophenotyping method of 15-colours panel antibody for Flow Cytometry in collaboration with Alfonso Blanco, the coordinator of Flow Cytometry Core in UCD Conway Institute. To get the VALIDATE pump priming grant as a PI improved Dr Julen Tomas Cortazar's CV and as a result he was awarded a Basque Government Fellowship in December 2019 enabling him to continue his research in Dublin for two more years. Furthermore, leading this project as the principal investigator allowed Julen to set up his own collaborative network and to interact directly with Drs Louise Gourlay, Giorgio Colombo and Susanna Dunachie. Invention disclosures for these new vaccine antigens have been written, while the BpOmpW vaccine has been patented. This project was presented at the VALIDATE 2019 & 2020 Annual Meeting, raising the profile of the project and PI within the Network. Julen and Siobhan also presented some of the project's results as an online VALIDATE Summer Seminar in 2020 to the VALIDATE community. As outreach, Julen created videos in three different languages (English, Spanish, Basque) talking about his research in vaccine development as part of the VALIDATE Outreach Week in December.
Start Year 2019
 
Description VALIDATE Pump-priming project P028 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Modelling the impact of a transmission blocking vaccine against leishmaniasis on sand fly vectorial capacity Led by Dr Matthew Rogers (LSHTM, UK), with Dr Laith Yakob (LSHTM, UK), and Dr Godwin Kwayke-Nuako (University of Cape Coast, Ghana) Project Aims Transmission blocking vaccines could offer a new way of controlling Leishmania infection by preventing the parasite from establishing themselves in their sand fly vector. However, before such targets are sought the feasibility of such a strategy must first be explored using a combination of basic knowledge of the transmission dynamics between hosts and vectors, the biology of the interaction between the parasite and its vector host and mathematical modelling. By applying modelling to a limited data set from the literature we have found that a hypothetical transmision blocking vaccine (TBV), which prevents the parasites from attaching to the gut of the sand fly, could result in effective protection or, worryingly, more transmission. These drastically different scenarios were dependent on whether Leishmania influence the survival of the sand fly in the early or later stages of infection. In this study we aim to clarify this and develop an improved model of sand fly transmission to better assess the impact of TBVs in the future. To do this we propose to conduct a series of sand fly infections using variable infecting doses of parasites, under stressed and unstressed conditions to extract data to parameterize our model. In addition, we will take advantage of mutant Leishmania which fail to attach or survive beyond the infecting bloodmeal, to test which parasites stages are responsible for sand fly mortality. We will also apply our model to an emerging Leishmania-vector relationship involving a new group of pathogenic Leishmania in Ghana and midges, which can host these parasites to a mature, transmissible stage. Data from this study will enable us to develop better TBVs against leishmaniasis. Project Outcomes Transmission blocking vaccines could offer a new way of controlling Leishmania infection by preventing the parasite from establishing themselves in their sand fly vector. The aim of the project was to improve our understanding of this parasite-vector interaction to assess transmission blockade as a means of leishmanaisis control. Our initial modelling found that a hypothetical transmission blocking vaccine (TBV), which prevents the parasites from attaching to the gut of the sand fly, could result in effective protection or, worryingly, more transmission, depending on whether Leishmania influences the survival of the sand fly in the early or later stages of infection. Our results, over a series of sand fly infections using variable infecting doses of parasites, under stressed and unstressed conditions, demonstrated that parasite-induced mortality was determined by the density of infecting parasites. Furthermore, this was only manifest if the infected vector was subjected to other environmental stresses, including sugarmeal deprivation, induced flight and low temperatures. Experiments using mutant Leishmania which fail to attach or survive beyond the infecting bloodmeal, highlighted that sand fly mortality was associated with the early, bloodmeal, stages of the parasite. This was confirmed with wild type Leishmania infections stressed at specific times, which showed that parasite-induced mortality was restricted to the bloodmeal phase of development (days 1-4), not after, when the sand fly became infectious. These data indicate that a TBV which targets midgut attachment should work and have a proportional effect on transmission. Data acquired during these experiments are currently being used to parameterize our TBV model to confirm this. [This project was affected by COVID and received a no-cost extension]
Impact Publications: Hall A. R., Blakeman J. T., Eissa A. M., Chapman P., Morales-García A. L., Stennett L., Martin O., Giraud E., Dockrell D. H., Cameron N. R., Wiese M., Yakob L., Rogers M. E.* and Geoghegan M. (2020). Glycan-glycan interactions determine Leishmania attachment to the midgut of permissive sand fly vectors. Chemical Science. DOI:10.1039/D0SC03298K. (* corresponding author) Grants Global Health Innovative Technology fund (GHIT) (2020-2022): Testing the practicality of transmission blocking vaccines against leishmaniasis through the development of a novel screening technology platform combined with mathematical modelling. £571,000. Unsuccessful New collaborations and partnerships: Prof Mark Geoghegan, Newcastle University - Biophysics of Leishmania attachment. Dr Marion England, The Pirbright Institute - Culicoides-Leishmania infections in Ghana. Staff promotions or continuing professional development: - Promotion of Dr Mojca Kristan from Research Fellow to Assistant Professor - LMIC ECR career development for Dr Kwakye-Nuako, including knowledge transfer and training in terms of Leishmania vaccine development, validation and Leishmania transmission modelling in a TBV context, mentorship, and strengthening of UK-West Africa research collaboration Engagement activities (talks, presentations, workshops, posters, websites, radio/TV): - Presentation to Online VALIDATE Winter seminar series, 2021 - Instituto Nacional de Ciencia e Technologia em Entomologia - "New approaches and New Insights into Leishmania Transmission" Online seminar https://www.youtube.com/watch?v=oC5mKOt52lU - Bug Bitten Blog for Parasite and Vectors: New insight into the interaction between Leishmania and the sand fly midgut, May 2021. https://blogs.biomedcentral.com/bugbitten/2021/05/14/new-insight-into-the-interaction-between-leishmania-and-the-sand-fly-midgut/ - An article for the London Centre for Neglected Tropical Disease Research. Research booklet, 'An Innovative Research Collaboration: Selected Research Highlights 2021': "Atomic force microscopy identified a novel glycan mode of attachment between Leishmania and the sand fly midgut" Authors: Matthew Rogers, Laith Yakob, Mark Geoghegan. https://www.londonntd.org/news/lcntdr-publishes-2021-research-highlights Research databases or models, research tools/methods or software created: New experimental model for trabsmission blockade evaluation New experimental model for testing Leishmania vectorial capacity. New mathematical model for trabsmission blockade evaluation New mathematical model for testing Leishmania vectorial capacity.
Start Year 2019
 
Description VALIDATE Pump-priming project P028 
Organisation University of Cape Coast
Country Ghana 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Modelling the impact of a transmission blocking vaccine against leishmaniasis on sand fly vectorial capacity Led by Dr Matthew Rogers (LSHTM, UK), with Dr Laith Yakob (LSHTM, UK), and Dr Godwin Kwayke-Nuako (University of Cape Coast, Ghana) Project Aims Transmission blocking vaccines could offer a new way of controlling Leishmania infection by preventing the parasite from establishing themselves in their sand fly vector. However, before such targets are sought the feasibility of such a strategy must first be explored using a combination of basic knowledge of the transmission dynamics between hosts and vectors, the biology of the interaction between the parasite and its vector host and mathematical modelling. By applying modelling to a limited data set from the literature we have found that a hypothetical transmision blocking vaccine (TBV), which prevents the parasites from attaching to the gut of the sand fly, could result in effective protection or, worryingly, more transmission. These drastically different scenarios were dependent on whether Leishmania influence the survival of the sand fly in the early or later stages of infection. In this study we aim to clarify this and develop an improved model of sand fly transmission to better assess the impact of TBVs in the future. To do this we propose to conduct a series of sand fly infections using variable infecting doses of parasites, under stressed and unstressed conditions to extract data to parameterize our model. In addition, we will take advantage of mutant Leishmania which fail to attach or survive beyond the infecting bloodmeal, to test which parasites stages are responsible for sand fly mortality. We will also apply our model to an emerging Leishmania-vector relationship involving a new group of pathogenic Leishmania in Ghana and midges, which can host these parasites to a mature, transmissible stage. Data from this study will enable us to develop better TBVs against leishmaniasis. Project Outcomes Transmission blocking vaccines could offer a new way of controlling Leishmania infection by preventing the parasite from establishing themselves in their sand fly vector. The aim of the project was to improve our understanding of this parasite-vector interaction to assess transmission blockade as a means of leishmanaisis control. Our initial modelling found that a hypothetical transmission blocking vaccine (TBV), which prevents the parasites from attaching to the gut of the sand fly, could result in effective protection or, worryingly, more transmission, depending on whether Leishmania influences the survival of the sand fly in the early or later stages of infection. Our results, over a series of sand fly infections using variable infecting doses of parasites, under stressed and unstressed conditions, demonstrated that parasite-induced mortality was determined by the density of infecting parasites. Furthermore, this was only manifest if the infected vector was subjected to other environmental stresses, including sugarmeal deprivation, induced flight and low temperatures. Experiments using mutant Leishmania which fail to attach or survive beyond the infecting bloodmeal, highlighted that sand fly mortality was associated with the early, bloodmeal, stages of the parasite. This was confirmed with wild type Leishmania infections stressed at specific times, which showed that parasite-induced mortality was restricted to the bloodmeal phase of development (days 1-4), not after, when the sand fly became infectious. These data indicate that a TBV which targets midgut attachment should work and have a proportional effect on transmission. Data acquired during these experiments are currently being used to parameterize our TBV model to confirm this. [This project was affected by COVID and received a no-cost extension]
Impact Publications: Hall A. R., Blakeman J. T., Eissa A. M., Chapman P., Morales-García A. L., Stennett L., Martin O., Giraud E., Dockrell D. H., Cameron N. R., Wiese M., Yakob L., Rogers M. E.* and Geoghegan M. (2020). Glycan-glycan interactions determine Leishmania attachment to the midgut of permissive sand fly vectors. Chemical Science. DOI:10.1039/D0SC03298K. (* corresponding author) Grants Global Health Innovative Technology fund (GHIT) (2020-2022): Testing the practicality of transmission blocking vaccines against leishmaniasis through the development of a novel screening technology platform combined with mathematical modelling. £571,000. Unsuccessful New collaborations and partnerships: Prof Mark Geoghegan, Newcastle University - Biophysics of Leishmania attachment. Dr Marion England, The Pirbright Institute - Culicoides-Leishmania infections in Ghana. Staff promotions or continuing professional development: - Promotion of Dr Mojca Kristan from Research Fellow to Assistant Professor - LMIC ECR career development for Dr Kwakye-Nuako, including knowledge transfer and training in terms of Leishmania vaccine development, validation and Leishmania transmission modelling in a TBV context, mentorship, and strengthening of UK-West Africa research collaboration Engagement activities (talks, presentations, workshops, posters, websites, radio/TV): - Presentation to Online VALIDATE Winter seminar series, 2021 - Instituto Nacional de Ciencia e Technologia em Entomologia - "New approaches and New Insights into Leishmania Transmission" Online seminar https://www.youtube.com/watch?v=oC5mKOt52lU - Bug Bitten Blog for Parasite and Vectors: New insight into the interaction between Leishmania and the sand fly midgut, May 2021. https://blogs.biomedcentral.com/bugbitten/2021/05/14/new-insight-into-the-interaction-between-leishmania-and-the-sand-fly-midgut/ - An article for the London Centre for Neglected Tropical Disease Research. Research booklet, 'An Innovative Research Collaboration: Selected Research Highlights 2021': "Atomic force microscopy identified a novel glycan mode of attachment between Leishmania and the sand fly midgut" Authors: Matthew Rogers, Laith Yakob, Mark Geoghegan. https://www.londonntd.org/news/lcntdr-publishes-2021-research-highlights Research databases or models, research tools/methods or software created: New experimental model for trabsmission blockade evaluation New experimental model for testing Leishmania vectorial capacity. New mathematical model for trabsmission blockade evaluation New mathematical model for testing Leishmania vectorial capacity.
Start Year 2019
 
Description VALIDATE Pump-priming project P030 
Organisation Addis Ababa University
Country Ethiopia 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution T cell receptor sequencing to identify correlates of protection in human tuberculosis vaccine studies Led by Dr Gabriele Pollara (UCL, UK), with Prof Mahdad Noursadeghi (UCL, UK), Assoc Prof Susanna Brighenti (Karolinska Institutet, Sweden), Assoc Prof Senait Ashenafi (Addis Ababa University, Ethiopia) and Prof Benny Chain (UCL, UK) Project Aims Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. We aim to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. We propose that measuring the number of distinct families of T cells at the site of infection may be more informative. In this project, we aim to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. Importantly, our approach to measure different families of T cells at the site of an immune response within people, can be applied in large scale clinical studies, first to validate these measurements as predicting immune protection against TB and second to test the effects of vaccination. Project Outcomes Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. The project aimed to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. The project proposed that measuring the number of distinct families of T cells at the site of infection may be more informative. We aimed to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. We have collected all the tissue and blood samples required for this project and extracted materials required for the assessment of the T cell families present. Analysis of our data is still ongoing, and we will share our findings as soon as possible. Our overall ambition remains to apply this assessment of T cells at the site of an immune response in large scale clinical studies to predict immune protection against TB and to test the effects of vaccination. [This project was significantly affected by the COVID-19 pandemic lockdowns and was granted a no-cost extension]
Impact This is a new collaboration between UCL, Karolinska and Addis Ababa University. The project lead, Dr Gabriele Pollara, is an ECR and this grant award supported his career building and learning about leading and managing a research project. Since the award, Dr Pollara has gained an Assistant Professor role at UCL. The transcriptomic data from the site of TST that has been collected to date will be placed in a public repository for public re-use and interrogation by the wider research community. The team will seek to publish the findings from both the primary project and the alternative investigations undertaken in peer-review publications.
Start Year 2019
 
Description VALIDATE Pump-priming project P030 
Organisation Karolinska Institute
Country Sweden 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution T cell receptor sequencing to identify correlates of protection in human tuberculosis vaccine studies Led by Dr Gabriele Pollara (UCL, UK), with Prof Mahdad Noursadeghi (UCL, UK), Assoc Prof Susanna Brighenti (Karolinska Institutet, Sweden), Assoc Prof Senait Ashenafi (Addis Ababa University, Ethiopia) and Prof Benny Chain (UCL, UK) Project Aims Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. We aim to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. We propose that measuring the number of distinct families of T cells at the site of infection may be more informative. In this project, we aim to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. Importantly, our approach to measure different families of T cells at the site of an immune response within people, can be applied in large scale clinical studies, first to validate these measurements as predicting immune protection against TB and second to test the effects of vaccination. Project Outcomes Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. The project aimed to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. The project proposed that measuring the number of distinct families of T cells at the site of infection may be more informative. We aimed to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. We have collected all the tissue and blood samples required for this project and extracted materials required for the assessment of the T cell families present. Analysis of our data is still ongoing, and we will share our findings as soon as possible. Our overall ambition remains to apply this assessment of T cells at the site of an immune response in large scale clinical studies to predict immune protection against TB and to test the effects of vaccination. [This project was significantly affected by the COVID-19 pandemic lockdowns and was granted a no-cost extension]
Impact This is a new collaboration between UCL, Karolinska and Addis Ababa University. The project lead, Dr Gabriele Pollara, is an ECR and this grant award supported his career building and learning about leading and managing a research project. Since the award, Dr Pollara has gained an Assistant Professor role at UCL. The transcriptomic data from the site of TST that has been collected to date will be placed in a public repository for public re-use and interrogation by the wider research community. The team will seek to publish the findings from both the primary project and the alternative investigations undertaken in peer-review publications.
Start Year 2019
 
Description VALIDATE Pump-priming project P030 
Organisation University College London
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution T cell receptor sequencing to identify correlates of protection in human tuberculosis vaccine studies Led by Dr Gabriele Pollara (UCL, UK), with Prof Mahdad Noursadeghi (UCL, UK), Assoc Prof Susanna Brighenti (Karolinska Institutet, Sweden), Assoc Prof Senait Ashenafi (Addis Ababa University, Ethiopia) and Prof Benny Chain (UCL, UK) Project Aims Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. We aim to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. We propose that measuring the number of distinct families of T cells at the site of infection may be more informative. In this project, we aim to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. Importantly, our approach to measure different families of T cells at the site of an immune response within people, can be applied in large scale clinical studies, first to validate these measurements as predicting immune protection against TB and second to test the effects of vaccination. Project Outcomes Tuberculosis (TB) is the disease caused by infection with Mycobacterium tuberculosis (Mtb). TB is the most common infectious cause of death world-wide. BCG vaccination against TB, is partially effective in children but does not provide long lasting protection for adults, who are most frequently affected. A critical barrier to the development of better vaccines is the lack of laboratory measurements that predict how well new vaccine candidates will work. In fact, we do not know precisely how the immune system protects us against TB. The project aimed to identify new measurements of immune protection against TB, which may be used to develop and test new vaccines in the future. We do know that immune cells called T cells make an important contribution to protection against TB. These are families of cells which recognise and respond to certain molecules within the bacteria. Simply measuring the number of T cells that recognise Mtb in blood samples has not provided a good measure of vaccine protection. The project proposed that measuring the number of distinct families of T cells at the site of infection may be more informative. We aimed to show proof of principle that these measurements can be made at the site of a skin test which stimulates an immune response to Mtb, and that the distinct T cell families which accumulate in the skin test are more similar to the site of disease than in blood samples. We have collected all the tissue and blood samples required for this project and extracted materials required for the assessment of the T cell families present. Analysis of our data is still ongoing, and we will share our findings as soon as possible. Our overall ambition remains to apply this assessment of T cells at the site of an immune response in large scale clinical studies to predict immune protection against TB and to test the effects of vaccination. [This project was significantly affected by the COVID-19 pandemic lockdowns and was granted a no-cost extension]
Impact This is a new collaboration between UCL, Karolinska and Addis Ababa University. The project lead, Dr Gabriele Pollara, is an ECR and this grant award supported his career building and learning about leading and managing a research project. Since the award, Dr Pollara has gained an Assistant Professor role at UCL. The transcriptomic data from the site of TST that has been collected to date will be placed in a public repository for public re-use and interrogation by the wider research community. The team will seek to publish the findings from both the primary project and the alternative investigations undertaken in peer-review publications.
Start Year 2019
 
Description VALIDATE Pump-priming project P033 
Organisation Institute of Post Graduate Medical Education And Research
Country India 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Characterising the cellular immunity and metabolic response to Mycobacterium tuberculosis and Burkholderia pseudomallei in Indian patients for vaccine design Led by Prof Chiranjay Mukhopadhyay (Manipal Academy of Higher Education, India), with Prof Susanna Dunachie (University of Oxford, UK) and Prof Mitali Chatterjee (IPGMER, India) Project Aims No vaccine is available for the neglected tropical disease melioidosis, and there is need of an improved vaccine for tuberculosis (TB). Both diseases are similar in that the bacteria live inside cells and their defence mechanisms are comparable. People suffering from diabetes mellitus (DM) have a three-fold higher risk of falling sick because of TB, and a twelve-fold higher risk of falling sick because of melioidosis than non-diabetic people. We therefore need to understand the reason why diabetic people are more susceptible to these bacteria. Likewise, understanding the response from the immune system of diabetic patients suffering from TB or melioidosis, will help in developing a new or improved vaccine. We will combine expertise at the Department of Microbioloy at the Manipal Academy of Higher Education (MAHE) in India and the University of Oxford, to establish how the metabolic status of immune cells differ in diabetic people that makes them susceptible to TB and melioidosis. In particular, we will investigate the metabolism of immune cells of these patient groups. Just like every other cell in the human body, immune cells need energy to function, especially when they have to be very active, as during an infection when they have to 'fight' the pathogen. Immune cells mostly use sugar (glucose) to acquire their energy, but the cells can use different pathways to convert glucose into energy. We will investigate how the metabolism of immune cells of diabetic patients responds to an infection with TB and melioidosis and if this differs from non-diabetics. We believe this will help to design better vaccines for TB and melioidosis in people with diabetes, allow development of better immune correlates of protection, and can also help us develop ways to use medicines at the time of vaccination to boost immune responses in people with diabetes. Project Outcomes The main goal of our project is to establish how the immunometabolic status of the diabetic people make them more susceptible to tuberculosis (TB) and melioidosis diseases in Indian subjects. In this regard, the first study objective includes understanding the cellular immune response to specific target antigens with respect to TB and melioidosis diseases that will help in getting a better insight into determining which antigens are immunogenic and which immune responses are upregulated and protective. This is followed by targeting the study towards metabolism of different T-cell population, the main players of the adaptive immune system, which will aid in obtaining insight into understanding the difference in the metabolism of T-cells in each patient groups, and examination of the interaction between T cell response and metabolism will illuminate potential mechanisms for immune impairment in diabetes leading to susceptibility to these infections. Finally, we will correlate the immunometabolic dysfunction to increased susceptibility towards TB and melioidosis. We have standardized peripheral blood mononuclear cells (PBMC) isolation and whole blood stimulation assay protocols. The blood samples of all the 9 recruited patients in our study have been processed for PBMC isolation and whole blood stimulation followed by cryopreservation until further downstream processing is performed. Initial standardization of ELISpot assay was performed using cryo-revived PBMCs' derived from healthy individuals. The standardized ELISpot plates will be couriered to nearby centre for further analysis. We will be carrying out ELISpot assay on the isolated PBMCs from the patients recruited for the study followed by further analysis of the plates based on the ELISpot readings obtained. Standardization of intracellular cytokine staining experiment will be performed prior to the patient sample processing for the same. Study of T-cell metabolism during infection and recovery will be completed using flow cytometry and Seahorse Extracellular Flux analysis. Our work will lay the foundation for the identification of drug candidates for protection in diabetes mellitus (DM) and drug discovery for possibilities of clinical interventions towards cure of TB and melioidosis. Standardization of immunometabolism experiment protocols are on the way preceding its application on the blood samples of the recruited patients. Antigen specific cell responses will be determined using flow cytometry. Study of T-cell metabolism during infection and recovery will be completed using flow cytometry and Seahorse Extracellular Flux analysis. Our work will lay the foundation for the identification of drug candidates for protection in diabetes mellitus (DM) and drug discovery for possibilities of clinical interventions towards cure of TB and melioidosis. Our studies will help understand the dependency/preference of nutrient availability and subsequent immunometabolic changes for the Bp infection progression. It will also help in understanding the increased susceptibility to infection that is a notable concern for diabetic patients. Studies from patient samples and cell line work will lay the foundation for more mechanistic studies that remain unexplored. [This project was significantly impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact Funding: Members of this project applied for extramural funding for some proposals related to this VALIDATE pump priming project: a. Evaluation of the immuno-metabolism is profile of patients with melioidosis and tuberculosis for developing immunotherapeutic strategies including vaccine design; ICMR, Govt of India; INR 86,94,000 (funded) b. Comparative analysis of sepsis pathophysiology and outcome measures in Burkholderia pseudomallei and other Gram-negative bacteria by multi-omics methods; ICMR, Govt of India; INR 83,71,940 (unsuccessful) c. An "omics" based approach to elucidate the molecular mechanisms behind the pathophysiology of Neurological Melioidosis caused by Burkholderia pseudomallei; ICMR, Govt of India; INR: 67,51,840 (unsuccessful) d. To explore the immunogenicity and vaccine potential of Burkholderia pseudomallei (Bps) candidate antigens: an application based approach to combat bps mediated bioterrorism; DRDO, Govt of India (Concept proposal) e. Understanding the effect of diabetes mellitus on immunometabolic response to Burkholderia pseudomallei in Indian patients f. Sepsis induced stress and Network Analysis on Pathogen Expression of stress-combat genes: the SNAPE study Funding Agency and Budget: ICMR, Govt of India; INR 35,50,648 (funded) Capacity building: The MAHE laboratory was performing serological, culture and molecular techniques for the diagnosis of melioidosis as a part of day-to-day diagnostic activities. It has now been upgraded further to perform immunological tests for this study which extended the research arena. Lab training for MAHE staff by Prof Susanna Dunachie helped improve strength in: 1) Elispot assay, 2) intracellular cytokine staining, 3) immunometabolism assay. Career development: The early student researcher working under this grant gained exposure to the advanced techniques involved in the immunology field from the expertise provided by the collaborating institute from University of Oxford, Prof. Susanna Dunachie's team and also the training on the immunometabolic work provided by Prof. Mitali Chatterjee's team from IPGMER, Kolkata, and also exposure to acquire clinical expertise provided by Dr. Chiranjay Mukhopadhyay's team in MAHE, Manipal. Influence on policy/patients/the public: While performing the research activities and recruitment of patients, extensive awareness has been created among the treating doctors and patients regarding melioidosis.
Start Year 2019
 
Description VALIDATE Pump-priming project P033 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Characterising the cellular immunity and metabolic response to Mycobacterium tuberculosis and Burkholderia pseudomallei in Indian patients for vaccine design Led by Prof Chiranjay Mukhopadhyay (Manipal Academy of Higher Education, India), with Prof Susanna Dunachie (University of Oxford, UK) and Prof Mitali Chatterjee (IPGMER, India) Project Aims No vaccine is available for the neglected tropical disease melioidosis, and there is need of an improved vaccine for tuberculosis (TB). Both diseases are similar in that the bacteria live inside cells and their defence mechanisms are comparable. People suffering from diabetes mellitus (DM) have a three-fold higher risk of falling sick because of TB, and a twelve-fold higher risk of falling sick because of melioidosis than non-diabetic people. We therefore need to understand the reason why diabetic people are more susceptible to these bacteria. Likewise, understanding the response from the immune system of diabetic patients suffering from TB or melioidosis, will help in developing a new or improved vaccine. We will combine expertise at the Department of Microbioloy at the Manipal Academy of Higher Education (MAHE) in India and the University of Oxford, to establish how the metabolic status of immune cells differ in diabetic people that makes them susceptible to TB and melioidosis. In particular, we will investigate the metabolism of immune cells of these patient groups. Just like every other cell in the human body, immune cells need energy to function, especially when they have to be very active, as during an infection when they have to 'fight' the pathogen. Immune cells mostly use sugar (glucose) to acquire their energy, but the cells can use different pathways to convert glucose into energy. We will investigate how the metabolism of immune cells of diabetic patients responds to an infection with TB and melioidosis and if this differs from non-diabetics. We believe this will help to design better vaccines for TB and melioidosis in people with diabetes, allow development of better immune correlates of protection, and can also help us develop ways to use medicines at the time of vaccination to boost immune responses in people with diabetes. Project Outcomes The main goal of our project is to establish how the immunometabolic status of the diabetic people make them more susceptible to tuberculosis (TB) and melioidosis diseases in Indian subjects. In this regard, the first study objective includes understanding the cellular immune response to specific target antigens with respect to TB and melioidosis diseases that will help in getting a better insight into determining which antigens are immunogenic and which immune responses are upregulated and protective. This is followed by targeting the study towards metabolism of different T-cell population, the main players of the adaptive immune system, which will aid in obtaining insight into understanding the difference in the metabolism of T-cells in each patient groups, and examination of the interaction between T cell response and metabolism will illuminate potential mechanisms for immune impairment in diabetes leading to susceptibility to these infections. Finally, we will correlate the immunometabolic dysfunction to increased susceptibility towards TB and melioidosis. We have standardized peripheral blood mononuclear cells (PBMC) isolation and whole blood stimulation assay protocols. The blood samples of all the 9 recruited patients in our study have been processed for PBMC isolation and whole blood stimulation followed by cryopreservation until further downstream processing is performed. Initial standardization of ELISpot assay was performed using cryo-revived PBMCs' derived from healthy individuals. The standardized ELISpot plates will be couriered to nearby centre for further analysis. We will be carrying out ELISpot assay on the isolated PBMCs from the patients recruited for the study followed by further analysis of the plates based on the ELISpot readings obtained. Standardization of intracellular cytokine staining experiment will be performed prior to the patient sample processing for the same. Study of T-cell metabolism during infection and recovery will be completed using flow cytometry and Seahorse Extracellular Flux analysis. Our work will lay the foundation for the identification of drug candidates for protection in diabetes mellitus (DM) and drug discovery for possibilities of clinical interventions towards cure of TB and melioidosis. Standardization of immunometabolism experiment protocols are on the way preceding its application on the blood samples of the recruited patients. Antigen specific cell responses will be determined using flow cytometry. Study of T-cell metabolism during infection and recovery will be completed using flow cytometry and Seahorse Extracellular Flux analysis. Our work will lay the foundation for the identification of drug candidates for protection in diabetes mellitus (DM) and drug discovery for possibilities of clinical interventions towards cure of TB and melioidosis. Our studies will help understand the dependency/preference of nutrient availability and subsequent immunometabolic changes for the Bp infection progression. It will also help in understanding the increased susceptibility to infection that is a notable concern for diabetic patients. Studies from patient samples and cell line work will lay the foundation for more mechanistic studies that remain unexplored. [This project was significantly impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact Funding: Members of this project applied for extramural funding for some proposals related to this VALIDATE pump priming project: a. Evaluation of the immuno-metabolism is profile of patients with melioidosis and tuberculosis for developing immunotherapeutic strategies including vaccine design; ICMR, Govt of India; INR 86,94,000 (funded) b. Comparative analysis of sepsis pathophysiology and outcome measures in Burkholderia pseudomallei and other Gram-negative bacteria by multi-omics methods; ICMR, Govt of India; INR 83,71,940 (unsuccessful) c. An "omics" based approach to elucidate the molecular mechanisms behind the pathophysiology of Neurological Melioidosis caused by Burkholderia pseudomallei; ICMR, Govt of India; INR: 67,51,840 (unsuccessful) d. To explore the immunogenicity and vaccine potential of Burkholderia pseudomallei (Bps) candidate antigens: an application based approach to combat bps mediated bioterrorism; DRDO, Govt of India (Concept proposal) e. Understanding the effect of diabetes mellitus on immunometabolic response to Burkholderia pseudomallei in Indian patients f. Sepsis induced stress and Network Analysis on Pathogen Expression of stress-combat genes: the SNAPE study Funding Agency and Budget: ICMR, Govt of India; INR 35,50,648 (funded) Capacity building: The MAHE laboratory was performing serological, culture and molecular techniques for the diagnosis of melioidosis as a part of day-to-day diagnostic activities. It has now been upgraded further to perform immunological tests for this study which extended the research arena. Lab training for MAHE staff by Prof Susanna Dunachie helped improve strength in: 1) Elispot assay, 2) intracellular cytokine staining, 3) immunometabolism assay. Career development: The early student researcher working under this grant gained exposure to the advanced techniques involved in the immunology field from the expertise provided by the collaborating institute from University of Oxford, Prof. Susanna Dunachie's team and also the training on the immunometabolic work provided by Prof. Mitali Chatterjee's team from IPGMER, Kolkata, and also exposure to acquire clinical expertise provided by Dr. Chiranjay Mukhopadhyay's team in MAHE, Manipal. Influence on policy/patients/the public: While performing the research activities and recruitment of patients, extensive awareness has been created among the treating doctors and patients regarding melioidosis.
Start Year 2019
 
Description VALIDATE Pump-priming project P034 
Organisation Federal University of Minas Gerais
Country Brazil 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Cytomegalovirus as a risk factor for TB and leishmaniasis Led by Dr Lisa Stockdale (University of Oxford, UK), with Dr Robindra Basu Roy (LSHTM, UK), Dr Vivian Tamietti Martins (Federal University of Minas Gerais, Brazil), Assoc Prof Eduardo Coelho (Federal University of Minas Gerais, Brazil), and Prof Helen Fletcher (LSHTM, UK) Project Aims Tuberculosis and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Caused by the microbes Mycobacterium tuberculosis and the microscopic parasite Leishmania spp. respectively, control of these diseases is hampered by a lack of effective vaccines. The drugs that are available require long regimes, have significant side effects, and treatment is complicated by antimicrobial resistance. In 2018, TB overtook HIV/AIDS as the largest cause of mortality due to a single infection, and visceral leishmaniasis (VL; the most serious form of the disease) is fatal in 95% of cases if left untreated. The role of an accompanying viral infection influencing the development of TB and VL is well-known through the increased risk in People Living With HIV. Cytomegalovirus (CMV), another virus, has also recently been implicated in increased risk of TB disease. Our current understanding of TB and leishmaniasis is that the body's protective immune responses are skewed, enabling the microbes to remain in the body and cause disease. One of the immune molecules involved in this balance between pathogen persistence and pathogen clearance for both diseases is IL-10. It is linked with shifting this balance towards disease persistence. CMV has its own viral version of this molecule which acts similarly to human IL-10. Using three unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, we hope to understand if and how CMV is implicated in increased risk of infection and disease. Not only will this information help to direct future VL and TB vaccine development but it may also drive development of a CMV vaccine as a possible cross-pathogen contribution to control of both diseases. Project Outcomes This project aimed to investigate the link between tuberculosis (TB), leishmaniasis and the ubiquitous human herpes virus, cytomegalovirus (CMV). TB and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Previous research found a link between CMV infection and increased risk of, and reduced time to development of, TB disease. Despite also being an intracellular pathogen with immunological parallels to TB pathogenesis, the role of co-infection of CMV in leishmaniasis has not been investigated. Our current understanding of progression of both TB and leishmaniasis is characterised by dysregulation of protective immune responses in favour of an immune environment conducive to pathogen persistence. CMV is widely distributed in human populations and infection is highly associated with immune variation and activation. It is also linked to a range of chronic diseases, and increased overall mortality. CMV vaccine development is already underway, however based upon a small target population (women of childbearing age to protect neonates from CMV-associated neurological disorders), progress is slower than it might be if CMV were found to be a risk factor for other infections. Using 3 unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, this project measured humoral responses to CMV infection along with markers of inflammation, a human and viral version of the cytokine IL10, and Th1 and Th2 cytokines. Among Gambian children, highly-TB exposed uninfected children had higher levels of CMV IgG than matched (on exposure to TB) infected children, suggesting that infection with CMV may not play as big a part in risk of initial infection with M.tb as compared with progression to TB disease once infected. Only 4/52 individuals had measurable levels of CMV vIL10. All 52 individuals were positive for CMV IgG suggesting that 100% of children in this cohort had been infected with CMV at some point. Only 7/52 individuals were positive for CMV IgM which is a marker of current or recent infection, however markers of general inflammation were not elevated for those 7 individuals compared with the rest of the cohort. Among individuals recruited to a rural Ugandan case-control study, we found a dose-response between odds of progression to active TB disease and increased levels of CMV IgM, CMV IgG avidity and CRP levels. Significant positive correlations were seen between CMV IgG avidity and CMV IgG antibody levels, between measures of inflammation and CMV IgG, and between Th2 cytokines IL4 and IL13. Despite this, the dose response associations were not altered by the addition of CMV IgG levels into a regression model. No samples had detectable CMV vIL10 levels. Among Brazilian leishmania patients, all the visceral leishmanisis patients were CMV IgG positive, 49/50 tegumentary leishmaniasis patients were CMV IgG positive, and 36/50 control individuals were CMV IgG positive. Individuals with tegumentary leishmaniasis had significantly higher levels of CMV IgG compared with visceral leishmania patients, and both groups had significantly higher CMV-specific IgG compared with control individuals without leishmania infection. None of the control individuals, and only 3/50 tegumentary and 3/50 visceral leishmaniasis patients were positive for CMV IgM. CMV IgG avidity data showed no difference in levels between visceral and tegumentary leishmania patients, however both these groups had significantly elevated CMV-specific IgG avidity compared with healthy controls. C-reactive protein, a measure of inflammation, was significantly elevated in visceral leishmaniasis patients compared with either tegumentary leishmaniasis patients or healthy controls. Similarly, tegumentary leishmaniasis patients had elevated CRP levels compared with healthy controls. Data from multiplex cytokine analysis showed visceral leishmaniasis patients had elevated CXCL10 (IP-10), IL-12 and IL-10 compared with either tegumentary leishmaniasis patients or healthy controls. Overall, a positive correlation was seen between CMV-specific IgM and CMV-specific IgG, and between measures of CMV IgG or IgM and CRP. As with the TB samples, no samples from the leishmania cohorts had detectable CMV vIL10 levels. In summary, this project has found further evidence that CMV may be associated with TB disease in a Ugandan population and found preliminary evidence of an association between CMV infection and leishmaniasis infection. [This project was significantly impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact - This is a new collaboration between University of Oxford and the Federal University of Minas Gerais (Brazil) - Capacity Building: Lead applicant, Dr Lisa Stockdale, is an ECR, and this project will be a major step towards building her reputation. Two further co-applicants, Dr Robindra Basu Roy (LSHTM, UK) and Dr Vivian Tamietti Martins (FUMG, Brazil), are also ECRs. - When final data is analysed, this work will be written up for publication. - Collaborative position paper with other VALIDATE members: Understanding the interaction between cytomegalovirus and tuberculosis in children: The way forward. Olbrich et al 2021. Plos Pathogens.
Start Year 2019
 
Description VALIDATE Pump-priming project P034 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Cytomegalovirus as a risk factor for TB and leishmaniasis Led by Dr Lisa Stockdale (University of Oxford, UK), with Dr Robindra Basu Roy (LSHTM, UK), Dr Vivian Tamietti Martins (Federal University of Minas Gerais, Brazil), Assoc Prof Eduardo Coelho (Federal University of Minas Gerais, Brazil), and Prof Helen Fletcher (LSHTM, UK) Project Aims Tuberculosis and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Caused by the microbes Mycobacterium tuberculosis and the microscopic parasite Leishmania spp. respectively, control of these diseases is hampered by a lack of effective vaccines. The drugs that are available require long regimes, have significant side effects, and treatment is complicated by antimicrobial resistance. In 2018, TB overtook HIV/AIDS as the largest cause of mortality due to a single infection, and visceral leishmaniasis (VL; the most serious form of the disease) is fatal in 95% of cases if left untreated. The role of an accompanying viral infection influencing the development of TB and VL is well-known through the increased risk in People Living With HIV. Cytomegalovirus (CMV), another virus, has also recently been implicated in increased risk of TB disease. Our current understanding of TB and leishmaniasis is that the body's protective immune responses are skewed, enabling the microbes to remain in the body and cause disease. One of the immune molecules involved in this balance between pathogen persistence and pathogen clearance for both diseases is IL-10. It is linked with shifting this balance towards disease persistence. CMV has its own viral version of this molecule which acts similarly to human IL-10. Using three unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, we hope to understand if and how CMV is implicated in increased risk of infection and disease. Not only will this information help to direct future VL and TB vaccine development but it may also drive development of a CMV vaccine as a possible cross-pathogen contribution to control of both diseases. Project Outcomes This project aimed to investigate the link between tuberculosis (TB), leishmaniasis and the ubiquitous human herpes virus, cytomegalovirus (CMV). TB and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Previous research found a link between CMV infection and increased risk of, and reduced time to development of, TB disease. Despite also being an intracellular pathogen with immunological parallels to TB pathogenesis, the role of co-infection of CMV in leishmaniasis has not been investigated. Our current understanding of progression of both TB and leishmaniasis is characterised by dysregulation of protective immune responses in favour of an immune environment conducive to pathogen persistence. CMV is widely distributed in human populations and infection is highly associated with immune variation and activation. It is also linked to a range of chronic diseases, and increased overall mortality. CMV vaccine development is already underway, however based upon a small target population (women of childbearing age to protect neonates from CMV-associated neurological disorders), progress is slower than it might be if CMV were found to be a risk factor for other infections. Using 3 unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, this project measured humoral responses to CMV infection along with markers of inflammation, a human and viral version of the cytokine IL10, and Th1 and Th2 cytokines. Among Gambian children, highly-TB exposed uninfected children had higher levels of CMV IgG than matched (on exposure to TB) infected children, suggesting that infection with CMV may not play as big a part in risk of initial infection with M.tb as compared with progression to TB disease once infected. Only 4/52 individuals had measurable levels of CMV vIL10. All 52 individuals were positive for CMV IgG suggesting that 100% of children in this cohort had been infected with CMV at some point. Only 7/52 individuals were positive for CMV IgM which is a marker of current or recent infection, however markers of general inflammation were not elevated for those 7 individuals compared with the rest of the cohort. Among individuals recruited to a rural Ugandan case-control study, we found a dose-response between odds of progression to active TB disease and increased levels of CMV IgM, CMV IgG avidity and CRP levels. Significant positive correlations were seen between CMV IgG avidity and CMV IgG antibody levels, between measures of inflammation and CMV IgG, and between Th2 cytokines IL4 and IL13. Despite this, the dose response associations were not altered by the addition of CMV IgG levels into a regression model. No samples had detectable CMV vIL10 levels. Among Brazilian leishmania patients, all the visceral leishmanisis patients were CMV IgG positive, 49/50 tegumentary leishmaniasis patients were CMV IgG positive, and 36/50 control individuals were CMV IgG positive. Individuals with tegumentary leishmaniasis had significantly higher levels of CMV IgG compared with visceral leishmania patients, and both groups had significantly higher CMV-specific IgG compared with control individuals without leishmania infection. None of the control individuals, and only 3/50 tegumentary and 3/50 visceral leishmaniasis patients were positive for CMV IgM. CMV IgG avidity data showed no difference in levels between visceral and tegumentary leishmania patients, however both these groups had significantly elevated CMV-specific IgG avidity compared with healthy controls. C-reactive protein, a measure of inflammation, was significantly elevated in visceral leishmaniasis patients compared with either tegumentary leishmaniasis patients or healthy controls. Similarly, tegumentary leishmaniasis patients had elevated CRP levels compared with healthy controls. Data from multiplex cytokine analysis showed visceral leishmaniasis patients had elevated CXCL10 (IP-10), IL-12 and IL-10 compared with either tegumentary leishmaniasis patients or healthy controls. Overall, a positive correlation was seen between CMV-specific IgM and CMV-specific IgG, and between measures of CMV IgG or IgM and CRP. As with the TB samples, no samples from the leishmania cohorts had detectable CMV vIL10 levels. In summary, this project has found further evidence that CMV may be associated with TB disease in a Ugandan population and found preliminary evidence of an association between CMV infection and leishmaniasis infection. [This project was significantly impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact - This is a new collaboration between University of Oxford and the Federal University of Minas Gerais (Brazil) - Capacity Building: Lead applicant, Dr Lisa Stockdale, is an ECR, and this project will be a major step towards building her reputation. Two further co-applicants, Dr Robindra Basu Roy (LSHTM, UK) and Dr Vivian Tamietti Martins (FUMG, Brazil), are also ECRs. - When final data is analysed, this work will be written up for publication. - Collaborative position paper with other VALIDATE members: Understanding the interaction between cytomegalovirus and tuberculosis in children: The way forward. Olbrich et al 2021. Plos Pathogens.
Start Year 2019
 
Description VALIDATE Pump-priming project P034 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution Cytomegalovirus as a risk factor for TB and leishmaniasis Led by Dr Lisa Stockdale (University of Oxford, UK), with Dr Robindra Basu Roy (LSHTM, UK), Dr Vivian Tamietti Martins (Federal University of Minas Gerais, Brazil), Assoc Prof Eduardo Coelho (Federal University of Minas Gerais, Brazil), and Prof Helen Fletcher (LSHTM, UK) Project Aims Tuberculosis and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Caused by the microbes Mycobacterium tuberculosis and the microscopic parasite Leishmania spp. respectively, control of these diseases is hampered by a lack of effective vaccines. The drugs that are available require long regimes, have significant side effects, and treatment is complicated by antimicrobial resistance. In 2018, TB overtook HIV/AIDS as the largest cause of mortality due to a single infection, and visceral leishmaniasis (VL; the most serious form of the disease) is fatal in 95% of cases if left untreated. The role of an accompanying viral infection influencing the development of TB and VL is well-known through the increased risk in People Living With HIV. Cytomegalovirus (CMV), another virus, has also recently been implicated in increased risk of TB disease. Our current understanding of TB and leishmaniasis is that the body's protective immune responses are skewed, enabling the microbes to remain in the body and cause disease. One of the immune molecules involved in this balance between pathogen persistence and pathogen clearance for both diseases is IL-10. It is linked with shifting this balance towards disease persistence. CMV has its own viral version of this molecule which acts similarly to human IL-10. Using three unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, we hope to understand if and how CMV is implicated in increased risk of infection and disease. Not only will this information help to direct future VL and TB vaccine development but it may also drive development of a CMV vaccine as a possible cross-pathogen contribution to control of both diseases. Project Outcomes This project aimed to investigate the link between tuberculosis (TB), leishmaniasis and the ubiquitous human herpes virus, cytomegalovirus (CMV). TB and leishmaniasis are diseases that particularly affect some of the world's poorest populations. Previous research found a link between CMV infection and increased risk of, and reduced time to development of, TB disease. Despite also being an intracellular pathogen with immunological parallels to TB pathogenesis, the role of co-infection of CMV in leishmaniasis has not been investigated. Our current understanding of progression of both TB and leishmaniasis is characterised by dysregulation of protective immune responses in favour of an immune environment conducive to pathogen persistence. CMV is widely distributed in human populations and infection is highly associated with immune variation and activation. It is also linked to a range of chronic diseases, and increased overall mortality. CMV vaccine development is already underway, however based upon a small target population (women of childbearing age to protect neonates from CMV-associated neurological disorders), progress is slower than it might be if CMV were found to be a risk factor for other infections. Using 3 unique sample sets to investigate risk of infection with TB in Gambian children, risk of developing TB disease in Ugandan people, and severity of leishmaniasis in Brazilian people, this project measured humoral responses to CMV infection along with markers of inflammation, a human and viral version of the cytokine IL10, and Th1 and Th2 cytokines. Among Gambian children, highly-TB exposed uninfected children had higher levels of CMV IgG than matched (on exposure to TB) infected children, suggesting that infection with CMV may not play as big a part in risk of initial infection with M.tb as compared with progression to TB disease once infected. Only 4/52 individuals had measurable levels of CMV vIL10. All 52 individuals were positive for CMV IgG suggesting that 100% of children in this cohort had been infected with CMV at some point. Only 7/52 individuals were positive for CMV IgM which is a marker of current or recent infection, however markers of general inflammation were not elevated for those 7 individuals compared with the rest of the cohort. Among individuals recruited to a rural Ugandan case-control study, we found a dose-response between odds of progression to active TB disease and increased levels of CMV IgM, CMV IgG avidity and CRP levels. Significant positive correlations were seen between CMV IgG avidity and CMV IgG antibody levels, between measures of inflammation and CMV IgG, and between Th2 cytokines IL4 and IL13. Despite this, the dose response associations were not altered by the addition of CMV IgG levels into a regression model. No samples had detectable CMV vIL10 levels. Among Brazilian leishmania patients, all the visceral leishmanisis patients were CMV IgG positive, 49/50 tegumentary leishmaniasis patients were CMV IgG positive, and 36/50 control individuals were CMV IgG positive. Individuals with tegumentary leishmaniasis had significantly higher levels of CMV IgG compared with visceral leishmania patients, and both groups had significantly higher CMV-specific IgG compared with control individuals without leishmania infection. None of the control individuals, and only 3/50 tegumentary and 3/50 visceral leishmaniasis patients were positive for CMV IgM. CMV IgG avidity data showed no difference in levels between visceral and tegumentary leishmania patients, however both these groups had significantly elevated CMV-specific IgG avidity compared with healthy controls. C-reactive protein, a measure of inflammation, was significantly elevated in visceral leishmaniasis patients compared with either tegumentary leishmaniasis patients or healthy controls. Similarly, tegumentary leishmaniasis patients had elevated CRP levels compared with healthy controls. Data from multiplex cytokine analysis showed visceral leishmaniasis patients had elevated CXCL10 (IP-10), IL-12 and IL-10 compared with either tegumentary leishmaniasis patients or healthy controls. Overall, a positive correlation was seen between CMV-specific IgM and CMV-specific IgG, and between measures of CMV IgG or IgM and CRP. As with the TB samples, no samples from the leishmania cohorts had detectable CMV vIL10 levels. In summary, this project has found further evidence that CMV may be associated with TB disease in a Ugandan population and found preliminary evidence of an association between CMV infection and leishmaniasis infection. [This project was significantly impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact - This is a new collaboration between University of Oxford and the Federal University of Minas Gerais (Brazil) - Capacity Building: Lead applicant, Dr Lisa Stockdale, is an ECR, and this project will be a major step towards building her reputation. Two further co-applicants, Dr Robindra Basu Roy (LSHTM, UK) and Dr Vivian Tamietti Martins (FUMG, Brazil), are also ECRs. - When final data is analysed, this work will be written up for publication. - Collaborative position paper with other VALIDATE members: Understanding the interaction between cytomegalovirus and tuberculosis in children: The way forward. Olbrich et al 2021. Plos Pathogens.
Start Year 2019
 
Description VALIDATE Pump-priming project P036 
Organisation London School of Hygiene and Tropical Medicine (LSHTM)
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution An ex vivo model for understanding the impact of vaccination on Mycobacterial persister populations Led by Prof Samantha Sampson (Stellenbosch University, South Africa), with Asst Prof Andrea Zelmer (LSHTM, UK) and Dr Jomien Mouton (Stellenbosch University, South Africa) Project Aims Tuberculosis (TB) is a devastating disease worldwide. The available drugs, vaccines and tools for diagnosing the disease are sub-optimal, and more understanding of the bacterium that causes TB is needed. In particular, we need a better understanding of latent TB, a state in which the bacteria are thought to become "dormant", and are called "persisters". In this state, they are able to escape killing by conventional TB drugs, and survive within the host, but may later reawaken to cause active disease. In this study, we will investigate how the current vaccine against TB may influence the formation of these persister bacteria. To achieve this, we will use a special form of TB bacteria that express fluorescent proteins that tell us when the bacteria are in a persister state. We will use these specialised "reporter" bacteria to infect cells taken from vaccinated and non-vaccinated mice, and then characterise the host and bacterial cells within these samples. The results obtained could provide new insights into how host factors influence persister formation, and vice versa. This would help to rationally design new vaccines and therapies that reduce persister formation and therefore increase long-term protection against TB. Project Outcomes Tuberculosis (TB) is a devastating disease worldwide. The available drugs, vaccines and tools for diagnosing the disease are sub-optimal, and more understanding of the bacterium that causes TB is needed. In particular, we need a better understanding of latent TB, a state in which the bacteria are thought to become "dormant", and are called "persisters". In this state, they are able to escape killing by conventional TB drugs, and survive within the host, but may later reawaken to cause active disease. The VALIDATE pump-priming funding has allowed us to lay the foundation for work aimed at determining how the current vaccine against TB may influence the formation of these persister bacteria. To achieve this, we have used a special form of TB bacteria that express fluorescent proteins that tell us when the bacteria are in a persister state. We have optimised methods that will allow us to more rapidly determine how these specialised "reporter" bacteria respond when used to infect cells taken from vaccinated and non-vaccinated mice. We have developed recorded training videos that will allow others to learn how to perform the key techniques involved in this work. Work on this project is still ongoing, and we expect that the results obtained could provide new insights into how host factors influence persister formation, and vice versa. This would help to rationally design new vaccines and therapies that reduce persister formation and therefore increase long-term protection against TB. [This project was substantially impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact New collaboration Between Dr Andrea Zelmer (LSHTM) and the Stellenbosch team Publications Since the work is still ongoing, we have not yet submitted any manuscripts. However, we envisage that at least a brief methods paper (describing the use of the Mtb reporter strains in the MGIA system) will be submitted. Capacity development Ms. Su-Mari du Plessis and Dr. Zimvo Obasa (PhD student and post-doc involved in the in vitro and mouse work, respectively) attended a cthe NANO Boston conference in Boston, USA in October 2022, and the SA TB Conferencein Durban, South Africa in September, 2022. Dr. Obasa also undertook a research training visit to South Dakota State University in 2022, to further develop her expertise in murine models Dr. Obasa undertook postgraduate supervision training, and currently co-supervises an MSc student. Career development: The project included two female ECRs, Dr Jomien Mouton (Stellenbosch) and Dr Andrea Zelmer (LSHTM), and promoted the careers of each. Dr. Mouton has been granted an affiliate position at Stellenbosch University, which will allow her to continue student supervision and involvement in ongoing projects, as well as provide eligibility for additional funding opportunities (e.g. she recently submitted on NIH R01 application). In 2021 Prof. Sampson was appointed as a co-director of the VALIDATE network. Dr. Zelmer has moved to a new position as category level 3 laboratory manager at King's College London. Engagement: Dr. Mouton participated in VALIDATE for Schools, by presenting a lecture to school pupils at a UK-based school about her career path and research interests in May 2021. Prof. Sampson was featured on the VALIDATE network blog for International Day of Women and Girls in Science. February 2020 and 2021; Profiled on Division of Molecular Biology and Human Genetics Blog for Women's History month. March 2020; participated in Division of Molecular Biology and Human Genetics InstaLive interview series, profiling female scientists for Women's month in August 2021. P036 was presented at the VALIDATE Annual Meeting 2021. Research training resource: Dr. Zelmer developed a training video to provide instruction in the mouse handling and other techniques relating to the MGIA.
Start Year 2019
 
Description VALIDATE Pump-priming project P036 
Organisation University of Stellenbosch
Country South Africa 
Sector Academic/University 
PI Contribution VALIDATE funded project
Collaborator Contribution An ex vivo model for understanding the impact of vaccination on Mycobacterial persister populations Led by Prof Samantha Sampson (Stellenbosch University, South Africa), with Asst Prof Andrea Zelmer (LSHTM, UK) and Dr Jomien Mouton (Stellenbosch University, South Africa) Project Aims Tuberculosis (TB) is a devastating disease worldwide. The available drugs, vaccines and tools for diagnosing the disease are sub-optimal, and more understanding of the bacterium that causes TB is needed. In particular, we need a better understanding of latent TB, a state in which the bacteria are thought to become "dormant", and are called "persisters". In this state, they are able to escape killing by conventional TB drugs, and survive within the host, but may later reawaken to cause active disease. In this study, we will investigate how the current vaccine against TB may influence the formation of these persister bacteria. To achieve this, we will use a special form of TB bacteria that express fluorescent proteins that tell us when the bacteria are in a persister state. We will use these specialised "reporter" bacteria to infect cells taken from vaccinated and non-vaccinated mice, and then characterise the host and bacterial cells within these samples. The results obtained could provide new insights into how host factors influence persister formation, and vice versa. This would help to rationally design new vaccines and therapies that reduce persister formation and therefore increase long-term protection against TB. Project Outcomes Tuberculosis (TB) is a devastating disease worldwide. The available drugs, vaccines and tools for diagnosing the disease are sub-optimal, and more understanding of the bacterium that causes TB is needed. In particular, we need a better understanding of latent TB, a state in which the bacteria are thought to become "dormant", and are called "persisters". In this state, they are able to escape killing by conventional TB drugs, and survive within the host, but may later reawaken to cause active disease. The VALIDATE pump-priming funding has allowed us to lay the foundation for work aimed at determining how the current vaccine against TB may influence the formation of these persister bacteria. To achieve this, we have used a special form of TB bacteria that express fluorescent proteins that tell us when the bacteria are in a persister state. We have optimised methods that will allow us to more rapidly determine how these specialised "reporter" bacteria respond when used to infect cells taken from vaccinated and non-vaccinated mice. We have developed recorded training videos that will allow others to learn how to perform the key techniques involved in this work. Work on this project is still ongoing, and we expect that the results obtained could provide new insights into how host factors influence persister formation, and vice versa. This would help to rationally design new vaccines and therapies that reduce persister formation and therefore increase long-term protection against TB. [This project was substantially impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact New collaboration Between Dr Andrea Zelmer (LSHTM) and the Stellenbosch team Publications Since the work is still ongoing, we have not yet submitted any manuscripts. However, we envisage that at least a brief methods paper (describing the use of the Mtb reporter strains in the MGIA system) will be submitted. Capacity development Ms. Su-Mari du Plessis and Dr. Zimvo Obasa (PhD student and post-doc involved in the in vitro and mouse work, respectively) attended a cthe NANO Boston conference in Boston, USA in October 2022, and the SA TB Conferencein Durban, South Africa in September, 2022. Dr. Obasa also undertook a research training visit to South Dakota State University in 2022, to further develop her expertise in murine models Dr. Obasa undertook postgraduate supervision training, and currently co-supervises an MSc student. Career development: The project included two female ECRs, Dr Jomien Mouton (Stellenbosch) and Dr Andrea Zelmer (LSHTM), and promoted the careers of each. Dr. Mouton has been granted an affiliate position at Stellenbosch University, which will allow her to continue student supervision and involvement in ongoing projects, as well as provide eligibility for additional funding opportunities (e.g. she recently submitted on NIH R01 application). In 2021 Prof. Sampson was appointed as a co-director of the VALIDATE network. Dr. Zelmer has moved to a new position as category level 3 laboratory manager at King's College London. Engagement: Dr. Mouton participated in VALIDATE for Schools, by presenting a lecture to school pupils at a UK-based school about her career path and research interests in May 2021. Prof. Sampson was featured on the VALIDATE network blog for International Day of Women and Girls in Science. February 2020 and 2021; Profiled on Division of Molecular Biology and Human Genetics Blog for Women's History month. March 2020; participated in Division of Molecular Biology and Human Genetics InstaLive interview series, profiling female scientists for Women's month in August 2021. P036 was presented at the VALIDATE Annual Meeting 2021. Research training resource: Dr. Zelmer developed a training video to provide instruction in the mouse handling and other techniques relating to the MGIA.
Start Year 2019
 
Description VALIDATE Pump-priming project P038 
Organisation Addis Ababa University
Country Ethiopia 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution VALIDATE Pump-priming project P038 Innovative tools to measure mycobacterial antigen expression in tissue Led by Dr Simon Waddell (University of Sussex, UK), with Dr Javier Salguero Bodes (PHE, UK), Prof Gobena Ameni (Addis Ababa University, Ethiopia), Dr Sally Sharpe (PHE, UK) and Dr Ann Rawkins (PHE, UK) [note: PHE became UKHSA in 2021) Project Aims Before we can hope to eradicate tuberculosis suffering worldwide, we need to identify the key protective antigens to target for vaccination that will eliminate Mycobacterium tuberculosis from the lung. This project will develop new RNA extraction and profiling techniques that will allow us to define the expression of M. tuberculosis (M.tb) antigens in lung tissue, at the site of disease. Our unique approach, building on international expertise from the UK and Ethiopia, will identify the key M.tb antigens for novel vaccine development, offering new opportunities for transformational change. Project Outcomes This award enabled us to develop key techniques to measure Mycobacterium tuberculosis gene expression in fresh and FFPE (formalin-fixed paraffin-embedded) tissue. This will allow us to characterise previously intractable host-pathogen interactions at the site of disease. We have also built a strong UK and international collaborative team to further extend this research. [This project was significantly impacted by the COVID-19 pandemic and was granted a no-cost extension.]
Impact New collaborations: This project was a new partnerships between Sussex and three groups at Public Health England (now UKHSA) and Addis Ababa University in Ethiopia. Publications: Medley J, Goff A, Bettencourt PJG, Dare M, Cole L, Cantillon D, Waddell SJ (2022). Dissecting the Mycobacterium bovis BCG response to macrophage infection to help prioritise targets for anti-tuberculosis drug and vaccine discovery. Vaccines (Basel); 10(1):113. PMID: 35062774. PMCID: PMC8780277 Grants/Follow-on funding: The team applied to the MRC for postdoctoral funding to support this research theme (submitted Jan 2020) 'MICA: Spatial and temporal profiling of M.tuberculosis in lung tissue at subpopulation and single cell resolutions to drive vaccine and drug discovery'. The application was discussed by the panel in June 2020 but was unsuccessful. The team were awarded funding from Public Health England for a PhD studentship (starting in October 2020) to explore M.tb phenotypes alongside immune responses in tissue to extend this project (supervisors - Salguero, Rawkins, Sharpe and Waddell). £105,100 - 'Defining Mycobacterium tuberculosis in lung tissue - a novel discovery platform for new vaccine and drug targets' (PhD student Jamie Medley). Additional University of Sussex funds were awarded to conduct a small supplementary murine TB drug discovery study through the project team. We reallocated the PHE salary costs to this activity. The experiment is ongoing at PHE-Porton, and it will provide additional materials for this project. Alongside other VALIDATE members the team applied to the Delta-Tissue (?T) - Wellcome Trust/DARPA in July 2021 (£11M) 'SprintFISH: predicting active TB by counting individual bacteria, RNA and protein molecules in thick tissues', which was unsuccessful. Capacity Development: This award has helped in the training of a PhD student, and two MSc students. ECR Career Development: PDRA Leticia Wildner worked on this project from Jan 2019 until the first lockdown in mid-March. She moved to postdoctoral position at University College London in April 2020 to work on TB clinical trials after three years as a Wellcome Trust-funded PDRA at Sussex. The work has been taken forward by PhD student Jamie Medley. Jamie attended the Keystone Conference (virtual) "Tuberculosis: Science aimed at ending the Epidemic" 2nd-4th December 2020; the University of Sussex RNA Virtual Symposium; the Mycobacterial Acid Fast Club (virtual 9th Jan 2021, in person June 2022, virtual Jan 2023); and UCL World TB Day March 2022. Datasets: Generating transcriptional datasets for M.tb from in vivo models of infection. Awards/recognition: Best poster for JM/AG at RNA Biology Society Meeting in Portsmouth, December 2022. Standardised protocols/SOPs: Extraction methods to be published alongside manuscripts in late 2023. Other SW attended all three days of the VALIDATE conference 2021 and presented on 14th October 2021. Aaron Goff presented some of this work at the Acid Fast Club (mycobacterial society UK) in Aberystwyth in June 2022. JM and AG presented this work as a poster at Keystone Conference in Colorado, USA in August 2022, and at the RNA Biology Society Meeting in Portsmouth, December 2022.
Start Year 2019
 
Description VALIDATE Pump-priming project P038 
Organisation Public Health England
Country United Kingdom 
Sector Public 
PI Contribution VALIDATE funded this project.
Collaborator Contribution VALIDATE Pump-priming project P038 Innovative tools to measure mycobacterial antigen expression in tissue Led by Dr Simon Waddell (University of Sussex, UK), with Dr Javier Salguero Bodes (PHE, UK), Prof Gobena Ameni (Addis Ababa University, Ethiopia), Dr Sally Sharpe (PHE, UK) and Dr Ann Rawkins (PHE, UK) [note: PHE became UKHSA in 2021) Project Aims Before we can hope to eradicate tuberculosis suffering worldwide, we need to identify the key protective antigens to target for vaccination that will eliminate Mycobacterium tuberculosis from the lung. This project will develop new RNA extraction and profiling techniques that will allow us to define the expression of M. tuberculosis (M.tb) antigens in lung tissue, at the site of disease. Our unique approach, building on international expertise from the UK and Ethiopia, will identify the key M.tb antigens for novel vaccine development, offering new opportunities for transformational change. Project Outcomes This award enabled us to develop key techniques to measure Mycobacterium tuberculosis gene expression in fresh and FFPE (formalin-fixed paraffin-embedded) tissue. This will allow us to characterise previously intractable host-pathogen interactions at the site of disease. We have also built a strong UK and international collaborative team to further extend this research. [This project was significantly impacted by the COVID-19 pandemic and was granted a no-cost extension.]
Impact New collaborations: This project was a new partnerships between Sussex and three groups at Public Health England (now UKHSA) and Addis Ababa University in Ethiopia. Publications: Medley J, Goff A, Bettencourt PJG, Dare M, Cole L, Cantillon D, Waddell SJ (2022). Dissecting the Mycobacterium bovis BCG response to macrophage infection to help prioritise targets for anti-tuberculosis drug and vaccine discovery. Vaccines (Basel); 10(1):113. PMID: 35062774. PMCID: PMC8780277 Grants/Follow-on funding: The team applied to the MRC for postdoctoral funding to support this research theme (submitted Jan 2020) 'MICA: Spatial and temporal profiling of M.tuberculosis in lung tissue at subpopulation and single cell resolutions to drive vaccine and drug discovery'. The application was discussed by the panel in June 2020 but was unsuccessful. The team were awarded funding from Public Health England for a PhD studentship (starting in October 2020) to explore M.tb phenotypes alongside immune responses in tissue to extend this project (supervisors - Salguero, Rawkins, Sharpe and Waddell). £105,100 - 'Defining Mycobacterium tuberculosis in lung tissue - a novel discovery platform for new vaccine and drug targets' (PhD student Jamie Medley). Additional University of Sussex funds were awarded to conduct a small supplementary murine TB drug discovery study through the project team. We reallocated the PHE salary costs to this activity. The experiment is ongoing at PHE-Porton, and it will provide additional materials for this project. Alongside other VALIDATE members the team applied to the Delta-Tissue (?T) - Wellcome Trust/DARPA in July 2021 (£11M) 'SprintFISH: predicting active TB by counting individual bacteria, RNA and protein molecules in thick tissues', which was unsuccessful. Capacity Development: This award has helped in the training of a PhD student, and two MSc students. ECR Career Development: PDRA Leticia Wildner worked on this project from Jan 2019 until the first lockdown in mid-March. She moved to postdoctoral position at University College London in April 2020 to work on TB clinical trials after three years as a Wellcome Trust-funded PDRA at Sussex. The work has been taken forward by PhD student Jamie Medley. Jamie attended the Keystone Conference (virtual) "Tuberculosis: Science aimed at ending the Epidemic" 2nd-4th December 2020; the University of Sussex RNA Virtual Symposium; the Mycobacterial Acid Fast Club (virtual 9th Jan 2021, in person June 2022, virtual Jan 2023); and UCL World TB Day March 2022. Datasets: Generating transcriptional datasets for M.tb from in vivo models of infection. Awards/recognition: Best poster for JM/AG at RNA Biology Society Meeting in Portsmouth, December 2022. Standardised protocols/SOPs: Extraction methods to be published alongside manuscripts in late 2023. Other SW attended all three days of the VALIDATE conference 2021 and presented on 14th October 2021. Aaron Goff presented some of this work at the Acid Fast Club (mycobacterial society UK) in Aberystwyth in June 2022. JM and AG presented this work as a poster at Keystone Conference in Colorado, USA in August 2022, and at the RNA Biology Society Meeting in Portsmouth, December 2022.
Start Year 2019
 
Description VALIDATE Pump-priming project P038 
Organisation University of Sussex
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution VALIDATE Pump-priming project P038 Innovative tools to measure mycobacterial antigen expression in tissue Led by Dr Simon Waddell (University of Sussex, UK), with Dr Javier Salguero Bodes (PHE, UK), Prof Gobena Ameni (Addis Ababa University, Ethiopia), Dr Sally Sharpe (PHE, UK) and Dr Ann Rawkins (PHE, UK) [note: PHE became UKHSA in 2021) Project Aims Before we can hope to eradicate tuberculosis suffering worldwide, we need to identify the key protective antigens to target for vaccination that will eliminate Mycobacterium tuberculosis from the lung. This project will develop new RNA extraction and profiling techniques that will allow us to define the expression of M. tuberculosis (M.tb) antigens in lung tissue, at the site of disease. Our unique approach, building on international expertise from the UK and Ethiopia, will identify the key M.tb antigens for novel vaccine development, offering new opportunities for transformational change. Project Outcomes This award enabled us to develop key techniques to measure Mycobacterium tuberculosis gene expression in fresh and FFPE (formalin-fixed paraffin-embedded) tissue. This will allow us to characterise previously intractable host-pathogen interactions at the site of disease. We have also built a strong UK and international collaborative team to further extend this research. [This project was significantly impacted by the COVID-19 pandemic and was granted a no-cost extension.]
Impact New collaborations: This project was a new partnerships between Sussex and three groups at Public Health England (now UKHSA) and Addis Ababa University in Ethiopia. Publications: Medley J, Goff A, Bettencourt PJG, Dare M, Cole L, Cantillon D, Waddell SJ (2022). Dissecting the Mycobacterium bovis BCG response to macrophage infection to help prioritise targets for anti-tuberculosis drug and vaccine discovery. Vaccines (Basel); 10(1):113. PMID: 35062774. PMCID: PMC8780277 Grants/Follow-on funding: The team applied to the MRC for postdoctoral funding to support this research theme (submitted Jan 2020) 'MICA: Spatial and temporal profiling of M.tuberculosis in lung tissue at subpopulation and single cell resolutions to drive vaccine and drug discovery'. The application was discussed by the panel in June 2020 but was unsuccessful. The team were awarded funding from Public Health England for a PhD studentship (starting in October 2020) to explore M.tb phenotypes alongside immune responses in tissue to extend this project (supervisors - Salguero, Rawkins, Sharpe and Waddell). £105,100 - 'Defining Mycobacterium tuberculosis in lung tissue - a novel discovery platform for new vaccine and drug targets' (PhD student Jamie Medley). Additional University of Sussex funds were awarded to conduct a small supplementary murine TB drug discovery study through the project team. We reallocated the PHE salary costs to this activity. The experiment is ongoing at PHE-Porton, and it will provide additional materials for this project. Alongside other VALIDATE members the team applied to the Delta-Tissue (?T) - Wellcome Trust/DARPA in July 2021 (£11M) 'SprintFISH: predicting active TB by counting individual bacteria, RNA and protein molecules in thick tissues', which was unsuccessful. Capacity Development: This award has helped in the training of a PhD student, and two MSc students. ECR Career Development: PDRA Leticia Wildner worked on this project from Jan 2019 until the first lockdown in mid-March. She moved to postdoctoral position at University College London in April 2020 to work on TB clinical trials after three years as a Wellcome Trust-funded PDRA at Sussex. The work has been taken forward by PhD student Jamie Medley. Jamie attended the Keystone Conference (virtual) "Tuberculosis: Science aimed at ending the Epidemic" 2nd-4th December 2020; the University of Sussex RNA Virtual Symposium; the Mycobacterial Acid Fast Club (virtual 9th Jan 2021, in person June 2022, virtual Jan 2023); and UCL World TB Day March 2022. Datasets: Generating transcriptional datasets for M.tb from in vivo models of infection. Awards/recognition: Best poster for JM/AG at RNA Biology Society Meeting in Portsmouth, December 2022. Standardised protocols/SOPs: Extraction methods to be published alongside manuscripts in late 2023. Other SW attended all three days of the VALIDATE conference 2021 and presented on 14th October 2021. Aaron Goff presented some of this work at the Acid Fast Club (mycobacterial society UK) in Aberystwyth in June 2022. JM and AG presented this work as a poster at Keystone Conference in Colorado, USA in August 2022, and at the RNA Biology Society Meeting in Portsmouth, December 2022.
Start Year 2019
 
Description VALIDATE Pump-priming project P040 
Organisation MRC/UVRI Uganda Research Unit on AIDS
Country Uganda 
Sector Public 
PI Contribution VALIDATE funded this project.
Collaborator Contribution Investigating the BCG-induced Natural Killer (NK) cell response in cattle and humans Led by Dr Iman Satti (University of Oxford, UK), with Asst Prof Stephen Cose (UVRI, Uganda), Prof Helen McShane (University of Oxford, UK), Prof Alison Elliott (UVRI, Uganda) and Prof Jayne Hope (University of Edinburgh, UK) Project Aims Tuberculosis (TB) causes a huge burden of disease in humans and animals. In 2017, there were 10 million new cases of human TB and 1.6 million people died. The global prevalence of bovine TB is estimated at 7.4% of livestock. The only licensed vaccine, Bacillus Calmette Guerin (BCG), provides variable protection against human and bovine TB, particularly in high TB-burden countries. The variability in BCG efficacy has been attributed to many factors including genetic factors, coinfections, geographical location and socioeconomic status. Developing a more effective vaccine requires a better understanding of the BCG-induced immune response in populations where BCG provides good protection as well as in those where BCG is less efficacious. Natural Killer (NK) cells are immune cells that respond early to infections but are also now recognised to contribute to a more durable immune response. There is emerging evidence for a role of NK cells in the control of TB and in vaccine-induced protection. Here we propose to evaluate the induction of NK cells after BCG vaccination. We will utilize pre-existing samples from BCG-vaccinated humans to define the kinetics of the NK cell response. We will use samples from the UK, and two TB endemic settings, South Africa and Uganda. Any differences, in responses to BCG, between these populations may contribute to differences in protective efficacy. We have previously shown that Cytomegalovirus (CMV)+ infants who developed TB had lower expression of NK cell-associated gene signatures. We will therefore define the CMV status in our studies and determine if CMV co-infection modulates the NK cell response. We will also evaluate NK cell responses in subjects vaccinated with inhaled BCG. Parallel experiments in samples from BCG-vaccinated cattle will allow comparison between species. Understanding immune responses to BCG vaccination in different populations and species facilitates developing new improved universal TB vaccine. Project Outcomes In this project, we aimed at characterising NK cell response in BCG vaccinated humans and cattle. We have used samples from BCG-vaccinated individuals to define the kinetics of the NK cell response. We have characterised NK cell response using samples collected from aerosol and intradermal BCG vaccinated UK adults and BCG vaccinated South Africa infants. To investigate the influence of pre-existing CMV-specific responses on the mycobacterial-specific responses induced by BCG vaccination, we analysed the collected data in relation to the individuals' CMV responses. Experimental work was done using frozen PBMC. Briefly, the cells were thawed and stimulated with mycobacterial and CMV antigens. Harvested cells were stained with markers to characterise the following populations: NK and T cell (CD3, CD4, CD8, CD56, CD16, CD57, KIR2DL1, KIR2DL2, NKG2A, CD94, NKG2D, NKp30 NKp44, NKp46, CD161 and KLRG1), cell activation (HLA-DR), memory cells (CD45RO) and cytokine and cytotoxicity markers (IFN-?, CD107a, Granzyme-B, Granulysin, Perorin). Samples were acquired using LSRFORTESSA (BD). The collected data was analysed using Flowjo (v10.7, BD. Pestle (v6.1) and Spice (v2) (both from NIH) were used to perform polyfunctional cells analysis. Statistical analysis was done using SPSS (v27) and Prism (v8). In this study, in vitro stimulation with mycobacterial antigens (PPD and BCG) and CMV peptides and surface and intracellular staining of cells allowed characterisation of NK cell antigen-specific responses and provided insight into: • Interaction between BCG vaccination, NK cell response and pre-existing CMV response • Expression of NK cell markers on cells collected from individuals categorised based on: • The magnitude of BCG-induced ELISpot response (high vs low ELISpot responders) • Pre-existing (baseline) mycobacterial and CMV responses • Association between the expression of NK cell markers and: • Adaptive cellular and humoral immune responses • The outcome of in vitro Mycobacterial growth Inhibition assay • Gene expression profile in samples collected from South African infants • In vitro growth inhibition of non-mycobacterial pathogens (BCG non-specific effects) • Lung mucosal immune responses (existing data generated from in vitro stimulation of bronchoalveolar lavage cells collected two weeks following aerosol and intradermal BCG vaccination of UK adults) The study of vaccine induced cellular responses in cattle is important: bovine tuberculosis (bTB) caused by infection with Mycobacterium bovis is an important disease affecting large numbers of herds in the UK, and worldwide. A proportion of human cases of TB are caused by M. bovis, and the immunology and pathology of TB is similar in cattle when compared to humans. Cattle are well protected by BCG when it is administered in early life, similar to the protection observed in human populations. These parallels with humans make cross-species comparisons important, and may strengthen the utilisation of the bovine TB challenge model for vaccine studies. In this study we have assessed changes in the number, proportion and phenotype of NK cells in the blood of BCG vaccinated calves. We have also stored material to enable comparison with mucosal responses to vaccination. We have established protocols for multiparametric flow cytometry and used these to assess activation, memory, cytotoxicity and cytokine expression by NK cell subsets and how these are altered by BCG vaccination. Differences in NK cell subsets were evident The NK cell responses will be compared to ELISpot responses and additional NK-expressed molecules will be assessed using qRT-PCR to complete datasets for publication. [The project was impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact Publications: data analysis is being completed for the manuscript to be drafted. Career development: this grant is led by an ECR, Dr Iman Satti, progressing her career. Standardised protocols/SOPs: protocols and flow cytometry panels developed from this project are being used in other TB and SARS-COV-2 human immunology studies
Start Year 2019
 
Description VALIDATE Pump-priming project P040 
Organisation University of Edinburgh
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution Investigating the BCG-induced Natural Killer (NK) cell response in cattle and humans Led by Dr Iman Satti (University of Oxford, UK), with Asst Prof Stephen Cose (UVRI, Uganda), Prof Helen McShane (University of Oxford, UK), Prof Alison Elliott (UVRI, Uganda) and Prof Jayne Hope (University of Edinburgh, UK) Project Aims Tuberculosis (TB) causes a huge burden of disease in humans and animals. In 2017, there were 10 million new cases of human TB and 1.6 million people died. The global prevalence of bovine TB is estimated at 7.4% of livestock. The only licensed vaccine, Bacillus Calmette Guerin (BCG), provides variable protection against human and bovine TB, particularly in high TB-burden countries. The variability in BCG efficacy has been attributed to many factors including genetic factors, coinfections, geographical location and socioeconomic status. Developing a more effective vaccine requires a better understanding of the BCG-induced immune response in populations where BCG provides good protection as well as in those where BCG is less efficacious. Natural Killer (NK) cells are immune cells that respond early to infections but are also now recognised to contribute to a more durable immune response. There is emerging evidence for a role of NK cells in the control of TB and in vaccine-induced protection. Here we propose to evaluate the induction of NK cells after BCG vaccination. We will utilize pre-existing samples from BCG-vaccinated humans to define the kinetics of the NK cell response. We will use samples from the UK, and two TB endemic settings, South Africa and Uganda. Any differences, in responses to BCG, between these populations may contribute to differences in protective efficacy. We have previously shown that Cytomegalovirus (CMV)+ infants who developed TB had lower expression of NK cell-associated gene signatures. We will therefore define the CMV status in our studies and determine if CMV co-infection modulates the NK cell response. We will also evaluate NK cell responses in subjects vaccinated with inhaled BCG. Parallel experiments in samples from BCG-vaccinated cattle will allow comparison between species. Understanding immune responses to BCG vaccination in different populations and species facilitates developing new improved universal TB vaccine. Project Outcomes In this project, we aimed at characterising NK cell response in BCG vaccinated humans and cattle. We have used samples from BCG-vaccinated individuals to define the kinetics of the NK cell response. We have characterised NK cell response using samples collected from aerosol and intradermal BCG vaccinated UK adults and BCG vaccinated South Africa infants. To investigate the influence of pre-existing CMV-specific responses on the mycobacterial-specific responses induced by BCG vaccination, we analysed the collected data in relation to the individuals' CMV responses. Experimental work was done using frozen PBMC. Briefly, the cells were thawed and stimulated with mycobacterial and CMV antigens. Harvested cells were stained with markers to characterise the following populations: NK and T cell (CD3, CD4, CD8, CD56, CD16, CD57, KIR2DL1, KIR2DL2, NKG2A, CD94, NKG2D, NKp30 NKp44, NKp46, CD161 and KLRG1), cell activation (HLA-DR), memory cells (CD45RO) and cytokine and cytotoxicity markers (IFN-?, CD107a, Granzyme-B, Granulysin, Perorin). Samples were acquired using LSRFORTESSA (BD). The collected data was analysed using Flowjo (v10.7, BD. Pestle (v6.1) and Spice (v2) (both from NIH) were used to perform polyfunctional cells analysis. Statistical analysis was done using SPSS (v27) and Prism (v8). In this study, in vitro stimulation with mycobacterial antigens (PPD and BCG) and CMV peptides and surface and intracellular staining of cells allowed characterisation of NK cell antigen-specific responses and provided insight into: • Interaction between BCG vaccination, NK cell response and pre-existing CMV response • Expression of NK cell markers on cells collected from individuals categorised based on: • The magnitude of BCG-induced ELISpot response (high vs low ELISpot responders) • Pre-existing (baseline) mycobacterial and CMV responses • Association between the expression of NK cell markers and: • Adaptive cellular and humoral immune responses • The outcome of in vitro Mycobacterial growth Inhibition assay • Gene expression profile in samples collected from South African infants • In vitro growth inhibition of non-mycobacterial pathogens (BCG non-specific effects) • Lung mucosal immune responses (existing data generated from in vitro stimulation of bronchoalveolar lavage cells collected two weeks following aerosol and intradermal BCG vaccination of UK adults) The study of vaccine induced cellular responses in cattle is important: bovine tuberculosis (bTB) caused by infection with Mycobacterium bovis is an important disease affecting large numbers of herds in the UK, and worldwide. A proportion of human cases of TB are caused by M. bovis, and the immunology and pathology of TB is similar in cattle when compared to humans. Cattle are well protected by BCG when it is administered in early life, similar to the protection observed in human populations. These parallels with humans make cross-species comparisons important, and may strengthen the utilisation of the bovine TB challenge model for vaccine studies. In this study we have assessed changes in the number, proportion and phenotype of NK cells in the blood of BCG vaccinated calves. We have also stored material to enable comparison with mucosal responses to vaccination. We have established protocols for multiparametric flow cytometry and used these to assess activation, memory, cytotoxicity and cytokine expression by NK cell subsets and how these are altered by BCG vaccination. Differences in NK cell subsets were evident The NK cell responses will be compared to ELISpot responses and additional NK-expressed molecules will be assessed using qRT-PCR to complete datasets for publication. [The project was impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact Publications: data analysis is being completed for the manuscript to be drafted. Career development: this grant is led by an ECR, Dr Iman Satti, progressing her career. Standardised protocols/SOPs: protocols and flow cytometry panels developed from this project are being used in other TB and SARS-COV-2 human immunology studies
Start Year 2019
 
Description VALIDATE Pump-priming project P040 
Organisation University of Oxford
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution Investigating the BCG-induced Natural Killer (NK) cell response in cattle and humans Led by Dr Iman Satti (University of Oxford, UK), with Asst Prof Stephen Cose (UVRI, Uganda), Prof Helen McShane (University of Oxford, UK), Prof Alison Elliott (UVRI, Uganda) and Prof Jayne Hope (University of Edinburgh, UK) Project Aims Tuberculosis (TB) causes a huge burden of disease in humans and animals. In 2017, there were 10 million new cases of human TB and 1.6 million people died. The global prevalence of bovine TB is estimated at 7.4% of livestock. The only licensed vaccine, Bacillus Calmette Guerin (BCG), provides variable protection against human and bovine TB, particularly in high TB-burden countries. The variability in BCG efficacy has been attributed to many factors including genetic factors, coinfections, geographical location and socioeconomic status. Developing a more effective vaccine requires a better understanding of the BCG-induced immune response in populations where BCG provides good protection as well as in those where BCG is less efficacious. Natural Killer (NK) cells are immune cells that respond early to infections but are also now recognised to contribute to a more durable immune response. There is emerging evidence for a role of NK cells in the control of TB and in vaccine-induced protection. Here we propose to evaluate the induction of NK cells after BCG vaccination. We will utilize pre-existing samples from BCG-vaccinated humans to define the kinetics of the NK cell response. We will use samples from the UK, and two TB endemic settings, South Africa and Uganda. Any differences, in responses to BCG, between these populations may contribute to differences in protective efficacy. We have previously shown that Cytomegalovirus (CMV)+ infants who developed TB had lower expression of NK cell-associated gene signatures. We will therefore define the CMV status in our studies and determine if CMV co-infection modulates the NK cell response. We will also evaluate NK cell responses in subjects vaccinated with inhaled BCG. Parallel experiments in samples from BCG-vaccinated cattle will allow comparison between species. Understanding immune responses to BCG vaccination in different populations and species facilitates developing new improved universal TB vaccine. Project Outcomes In this project, we aimed at characterising NK cell response in BCG vaccinated humans and cattle. We have used samples from BCG-vaccinated individuals to define the kinetics of the NK cell response. We have characterised NK cell response using samples collected from aerosol and intradermal BCG vaccinated UK adults and BCG vaccinated South Africa infants. To investigate the influence of pre-existing CMV-specific responses on the mycobacterial-specific responses induced by BCG vaccination, we analysed the collected data in relation to the individuals' CMV responses. Experimental work was done using frozen PBMC. Briefly, the cells were thawed and stimulated with mycobacterial and CMV antigens. Harvested cells were stained with markers to characterise the following populations: NK and T cell (CD3, CD4, CD8, CD56, CD16, CD57, KIR2DL1, KIR2DL2, NKG2A, CD94, NKG2D, NKp30 NKp44, NKp46, CD161 and KLRG1), cell activation (HLA-DR), memory cells (CD45RO) and cytokine and cytotoxicity markers (IFN-?, CD107a, Granzyme-B, Granulysin, Perorin). Samples were acquired using LSRFORTESSA (BD). The collected data was analysed using Flowjo (v10.7, BD. Pestle (v6.1) and Spice (v2) (both from NIH) were used to perform polyfunctional cells analysis. Statistical analysis was done using SPSS (v27) and Prism (v8). In this study, in vitro stimulation with mycobacterial antigens (PPD and BCG) and CMV peptides and surface and intracellular staining of cells allowed characterisation of NK cell antigen-specific responses and provided insight into: • Interaction between BCG vaccination, NK cell response and pre-existing CMV response • Expression of NK cell markers on cells collected from individuals categorised based on: • The magnitude of BCG-induced ELISpot response (high vs low ELISpot responders) • Pre-existing (baseline) mycobacterial and CMV responses • Association between the expression of NK cell markers and: • Adaptive cellular and humoral immune responses • The outcome of in vitro Mycobacterial growth Inhibition assay • Gene expression profile in samples collected from South African infants • In vitro growth inhibition of non-mycobacterial pathogens (BCG non-specific effects) • Lung mucosal immune responses (existing data generated from in vitro stimulation of bronchoalveolar lavage cells collected two weeks following aerosol and intradermal BCG vaccination of UK adults) The study of vaccine induced cellular responses in cattle is important: bovine tuberculosis (bTB) caused by infection with Mycobacterium bovis is an important disease affecting large numbers of herds in the UK, and worldwide. A proportion of human cases of TB are caused by M. bovis, and the immunology and pathology of TB is similar in cattle when compared to humans. Cattle are well protected by BCG when it is administered in early life, similar to the protection observed in human populations. These parallels with humans make cross-species comparisons important, and may strengthen the utilisation of the bovine TB challenge model for vaccine studies. In this study we have assessed changes in the number, proportion and phenotype of NK cells in the blood of BCG vaccinated calves. We have also stored material to enable comparison with mucosal responses to vaccination. We have established protocols for multiparametric flow cytometry and used these to assess activation, memory, cytotoxicity and cytokine expression by NK cell subsets and how these are altered by BCG vaccination. Differences in NK cell subsets were evident The NK cell responses will be compared to ELISpot responses and additional NK-expressed molecules will be assessed using qRT-PCR to complete datasets for publication. [The project was impacted by the COVID-19 pandemic and granted a no-cost extension]
Impact Publications: data analysis is being completed for the manuscript to be drafted. Career development: this grant is led by an ECR, Dr Iman Satti, progressing her career. Standardised protocols/SOPs: protocols and flow cytometry panels developed from this project are being used in other TB and SARS-COV-2 human immunology studies
Start Year 2019
 
Description VALIDATE Pump-priming project P045 
Organisation Eduardo Mondlane University
Country Mozambique 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution Cross protection against TB by attenuated Mycobacterium ulcerans strain 5114 Led by Dr Justice Boakye-Appiah (SGUL, UK), with Dr Rajko Reljic (SGUL, UK) and Dr Tufária Mussá (Eduardo Mondlane University, Mozambique) Bacillus Calmette-Guerin (BCG) vaccine has been used since 1921 as a vaccine against tuberculosis (TB). Over these years, the extent of protection it offers has been a matter of controversy as it has been found to offer no protection at all to people of certain age groups and geographical locations, while offering up to 60% protection to others. It is commonly thought that the lack of better protection by BCG may be due to over-attenuation, that is, loss of too many virulence factors. However, we have recently shown that BCG, despite the lack of virulence, is still capable of down-regulating cellular processes which are required for optimal immune responses. We therefore propose that other mycobacteria which share significant homology with Mycobacterium tuberculosis but are less capable of manipulating the cellular immune responses involved could induce a more protective immune response against TB. Mycobacterium ulcerans, the bacteria that cause Buruli Ulcer (BU) disease is closely related to both BCG and MTB. We have identified an avirulent strain of Mycobacterium ulcerans which by virtue of its inability to secrete the toxin mycolactone, did not cause any lesions in animal models of BU disease when we infected mice footpads. When used as a subcutaneous vaccine against BU disease, 70% of the mice were protected. When a boost vaccine (BURULIVAC) was added, protection went up to 100%. Here, we want to determine whether or not this strain of Mycobacterium ulcerans can offer superior protection to BCG against TB, by carrying out immunizations in mice and challenging with aerosolised MTB. We will also test the responses of immune cells collected from TB patients, latently MTB-infected individuals and healthy BCG vaccinees to purified protein derivatives from this strain of Mycobacterium ulcerans, and determine the degree of immune cross-reactivity between these two pathogens. Project Outcomes Bacillus Calmette-Guerin (BCG) vaccine has been used since 1921 as a vaccine against tuberculosis (TB). Over these years, the extent of protection it offers has been a matter of controversy as it has been found to offer no protection at all to people of certain age groups and geographical locations, while offering up to 60% protection to others. It is commonly thought that the lack of better protection by BCG may be due to over-attenuation, that is, loss of too many virulence factors. However, we have recently shown that BCG, despite the lack of virulence, is still capable of down-regulating cellular processes which are required for optimal immune responses. We therefore propose that other mycobacteria which share significant homology with Mycobacterium tuberculosis but are less capable of manipulating the cellular immune responses involved could induce a more protective immune response against TB. Mycobacterium ulcerans, the bacteria that cause Buruli Ulcer (BU) disease is closely related to both BCG and MTB. We have identified an avirulent strain of Mycobacterium ulcerans which by virtue of its inability to secrete the toxin mycolactone, did not cause any lesions in animal models of BU disease when we infected mice footpads. When used as a subcutaneous vaccine against BU disease, 70% of the mice were protected. When a boost vaccine (BURULIVAC) was added, protection went up to 100%. In this project, we intended to determine whether or not this strain of Mycobacterium ulcerans can offer superior protection to BCG against TB, by carrying out immunizations in mice and challenging with aerosolised MTB. We were also to test the responses of immune cells collected from TB patients, latently MTB-infected individuals and healthy BCG vaccinees to purified protein derivatives from this strain of Mycobacterium ulcerans, and determine the degree of immune cross-reactivity between these two pathogens. We immunised mice with mycolactone deficient Mycobacterium ulcerans boosted by its protein antigen Ag85A and some adjuvants. We also collected immune cells from TB patients, latent MTB- Infected individuals and healthy BCG vaccinees. Challenge experiments are however pending because of the prolonged COVID lockdowns and restrictions. Experiments are scheduled to continue as laboratory and animal work resume fully. [This project was granted a no-cost extension due to the impact of COVID-19.]
Impact Project Outputs This project was a new collaboration between SGUL and Dr Dr Tufária Mussá at Eduardo Mondlane University, Mozambique. This project involves two ECRs, Dr Justice Boakye-Appiah (SGUL, UK - project lead), and Dr Tufária Mussá (Eduardo Mondlane University, Mozambique). This was Justice's first grant award. Justice wrote up his final PhD thesis and on submission he continued as a Post-doctoral researcher on this project. Presentation of project at SGUL seminar November 2020. Dr Justice Boakye-Appiah (SGUL, UK - project lead) used the successful acquisition of this grant and the experience gathered undertaking the research as a springboard, and was recruited as a part-time research fellow by NIHR on its COVID-19 Vaccine trials.
Start Year 2019
 
Description VALIDATE Pump-priming project P045 
Organisation St George's University of London
Country United Kingdom 
Sector Academic/University 
PI Contribution VALIDATE funded this project.
Collaborator Contribution Cross protection against TB by attenuated Mycobacterium ulcerans strain 5114 Led by Dr Justice Boakye-Appiah (SGUL, UK), with Dr Rajko Reljic (SGUL, UK) and Dr Tufária Mussá (Eduardo Mondlane University, Mozambique) Bacillus Calmette-Guerin (BCG) vaccine has been used since 1921 as a vaccine against tuberculosis (TB). Over these years, the extent of protection it offers has been a matter of controversy as it has been found to offer no protection at all to people of certain age groups and geographical locations, while offering up to 60% protection to others. It is commonly thought that the lack of better protection by BCG may be due to over-attenuation, that is, loss of too many virulence factors. However, we have recently shown that BCG, despite the lack of virulence, is still capable of down-regulating cellular processes which are required for optimal immune responses. We therefore propose that other mycobacteria which share significant homology with Mycobacterium tuberculosis but are less capable of manipulating the cellular immune responses involved could induce a more protective immune response against TB. Mycobacterium ulcerans, the bacteria that cause Buruli Ulcer (BU) disease is closely related to both BCG and MTB. We have identified an avirulent strain of Mycobacterium ulcerans which by virtue of its inability to secrete the toxin mycolactone, did not cause any lesions in animal models of BU disease when we infected mice footpads. When used as a subcutaneous vaccine against BU disease, 70% of the mice were protected. When a boost vaccine (BURULIVAC) was added, protection went up to 100%. Here, we want to determine whether or not this strain of Mycobacterium ulcerans can offer superior protection to BCG against TB, by carrying out immunizations in mice and challenging with aerosolised MTB. We will also test the responses of immune cells collected from TB patients, latently MTB-infected individuals and healthy BCG vaccinees to purified protein derivatives from this strain of Mycobacterium ulcerans, and determine the degree of immune cross-reactivity between these two pathogens. Project Outcomes Bacillus Calmette-Guerin (BCG) vaccine has been used since 1921 as a vaccine against tuberculosis (TB). Over these years, the extent of protection it offers has been a matter of controversy as it has been found to offer no protection at all to people of certain age groups and geographical locations, while offering up to 60% protection to others. It is commonly thought that the lack of better protection by BCG may be due to over-attenuation, that is, loss of too many virulence factors. However, we have recently shown that BCG, despite the lack of virulence, is still capable of down-regulating cellular processes which are required for optimal immune responses. We therefore propose that other mycobacteria which share significant homology with Mycobacterium tuberculosis but are less capable of manipulating the cellular immune responses involved could induce a more protective immune response against TB. Mycobacterium ulcerans, the bacteria that cause Buruli Ulcer (BU) disease is closely related to both BCG and MTB. We have identified an avirulent strain of Mycobacterium ulcerans which by virtue of its inability to secrete the toxin mycolactone, did not cause any lesions in animal models of BU disease when we infected mice footpads. When used as a subcutaneous vaccine against BU disease, 70% of the mice were protected. When a boost vaccine (BURULIVAC) was added, protection went up to 100%. In this project, we intended to determine whether or not this strain of Mycobacterium ulcerans can offer superior protection to BCG against TB, by carrying out immunizations in mice and challenging with aerosolised MTB. We were also to test the responses of immune cells collected from TB patients, latently MTB-infected individuals and healthy BCG vaccinees to purified protein derivatives from this strain of Mycobacterium ulcerans, and determine the degree of immune cross-reactivity between these two pathogens. We immunised mice with mycolactone deficient Mycobacterium ulcerans boosted by its protein antigen Ag85A and some adjuvants. We also collected immune cells from TB patients, latent MTB- Infected individuals and healthy BCG vaccinees. Challenge experiments are however pending because of the prolonged COVID lockdowns and restrictions. Experiments are scheduled to continue as laboratory and animal work resume fully. [This project was granted a no-cost extension due to the impact of COVID-19.]
Impact Project Outputs This project was a new collaboration between SGUL and Dr Dr Tufária Mussá at Eduardo Mondlane University, Mozambique. This project involves two ECRs, Dr Justice Boakye-Appiah (SGUL, UK - project lead), and Dr Tufária Mussá (Eduardo Mondlane University, Mozambique). This was Justice's first grant award. Justice wrote up his final PhD thesis and on submission he continued as a Post-doctoral researcher on this project. Presentation of project at SGUL seminar November 2020. Dr Justice Boakye-Appiah (SGUL, UK - project lead) used the successful acquisition of this grant and the experience gathered undertaking the research as a springboard, and was recruited as a part-time research fellow by NIHR on its COVID-19 Vaccine trials.
Start Year 2019
 
Description Representation of VALIDATE at Events, Meetings, Workshops or similar 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact VALIDATE has been represented at various events to promote our network and encourage new membership, as well as to speak to funding bodies about our work:

30 Jul 2017, VALIDATE Management Team Prof Helen McShane (Director), Prof Helen Fletcher (Co-Director) and Samantha Vermaak (Network Manager) attended the MRC Vaccines Network meeting in London hosted by MRC.

23-24 Nov 2017, talk about VALIDATE given by Dr Iman Satti (VALIDATE Associate) at HIC-VAC 1st Annual Meeting.

5 Dec 2017, VALIDATE Network Manager Samantha Vermaak co-hosted a stand at the BSI Congress with the other GCRF MRC-funded Vaccine Networks, with a pull-up banner and leaflets about VALIDATE, speaking to the event delegates about our Network and the benefits of membership.

15 Jan 2018, VALIDATE Director Prof Helen McShane presented a slide about VALIDATE, and had leaflets at the event to promote VALIDATE.

18 Jan 2018, VALIDATE Director Prof Helen McShane mentioned VALIDATE in her talk at the UK Veterinary Vaccinology Network Conference, and had leaflets.

12 Feb 2018, VALIDATE Co-Director Prof Helen Fletcher had a slide about VALIDATE in her talk at the UCL-AHRI Symposium "Towards HIV and TB elimination in South Africa".

22 Feb 2018, VALIDATE leaflets and representation (Samantha Vermaak) at University of Oxford's 'Global Research Day: Tackling International Challenges" event.

26-27 Feb 2018, VALIDATE talk by Director Prof Helen McShane at Bactivac Annual Meeting (leaflets too).

20 Mar 2018, VALIDATE talk by Network Manager Samantha Vermaak at European Melioidosis Congress. Our Associate Dr Panjaporn Chaichana (MORU, Thailand) also presented her VALIDATE-funded research and thanked VALIDATE for our funding.

23 Mar 2018, VALIDATE Director Prof Helen McShane had VALIDATE slide at UCL/LSHTM/UCSF World TB Day Super-Symposium https://panopto.lshtm.ac.uk/Panopto/Pages/Viewer.aspx?id=5ddad554-12f4-42e3-bab8-8bdc83ecbfb2

26-27 Mar 2018, VALIDATE Network Management Board member Prof Paul Kaye mentioned VALIDATE in his talk at IVVN's 1st Annual Meeting (leaflets at meeting).

19 Apr 2018, VALIDATE Co-Director Prof Helen Fletcher gave a careers talk to 30+ PhD students from LSHTM/UCL, mentioning VALIDATE.

10 May 2018, VALIDATE Research Data Analyst Dr Deniz Cizmeci gave a talk about VALIDATE at an IDDO meeting.

5 Jul 2018, VALIDATE Network Manager Samantha Vermaak gave a talk about VALIDATE at the Acid Fast Club meeting.

30 Jul 2018, Prof Helen McShane and Samantha Vermaak attended the MRC GCRF Vaccine Network first year meeting.

19 Sep 2018, Network Manager Samantha Vermaak gave a talk about VALIDATE at the 9th EDCTP Forum.

25 Sep 2018, Network Manager Samantha Vermaak gave a talk about VALIDATE at the IMPRINT 2nd Annual Meeting (leaflets available too).

10 Oct 2018, Prof Helen McShane and Samantha Vermaak had a teleconference with Willem Hanekom and Anne Kasmar at the Gates Foundation re VALIDATE.

12 Nov 2018, Prof Helen McShane and Samantha Vermaak met with Prof Sarah Rowland-Jones, President of RSTMH, to discuss how our two networks could work together to mutual benefit.

9 Jan 2019, representation of VALIDATE by Network Director Prof Helen McShane and Network Management Board member Prof Paul Kaye at HIC-VAC Regulatory Workshop "Human challenge agents: scoping potential manufacturing guidelines".

29 Jan 2019, Director Prof Helen McShane gave a talk about VALIDATE at the TBVAC2020 Les Diablerets meeting, as part of the TBVI Symposium "TB Vaccines Global Collaboration and Recent Advances in the Field."

29 Jan 2019, Director Prof Helen McShane met with Ole Olesen at EDCTP to discuss VALIDATE.

18 March 2019, Fellow and NMB member Dr Rachel Tanner presented at the 3Rs Research Review Launch and 2018 Prize Presentation "An in vitro functional assay to replace virulent M.tb challenge for the testing of TB vaccine candidates".

20-21 March 2019, VALIDATE Manager Samantha Vermaak represented VALIDATE at Bactivac 2nd Annual Meeting in Birmingham.

29 April - 3 May 2019, VALIDATE Director Prof Helen Fletcher and member Prof Jayne Sutherland were speakers at the Vaccinology in Africa course.

16 May 2019, VALIDATE Director Prof Helen McShane spoke at the 20th ADVAC (Advanced course of vaccinology) in Annecy, France, and had VALIDATE leaflets.

1-7 July 2019, VALIDATE member Dr Amanda Gibson and the RVC TB research group had a stand at the Royal Society Summer exhibition, with VALIDATE logo to show they are supported by our Network.

24 Sept 2019, VALIDATE NMB member Prof Jo Prior had a VALIDATE poster and chatted to the MRC/UKRI team during their visit to dstl.

7 Nov 2019, VALIDATE Fellow and NMB member Dr Rachel Tanner won 'Woman of the Future award for Science'.

18 Nov 2019, VALIDATE Director Prof Helen McShane presented on why we need a human TB challenge model at HIC-VAC Annual Meeting.

18 Nov 2019, VALIDATE NMB member Prof Paul Kaye presented on progress with leishmaniasis challenge studies at HIV-VAC Annual Meeting.

31 Jan 2020, VALIDATE is a partner of the first World NTD Day (https://worldntdday.org/partners/).

In 2020 our representation at face-to-face meetings was curtailed by the COVID-19 pandemic, but we continued to talk via Skype/Zoom/Teams to funders about possibilities for VALIDATE and VALIDATE's members:

10 Mar 2020, Samantha Vermaak and Mirvat Surakhy (VALIDATE Bioinformatician) met IDDO staff to discuss data sharing opportunities.

Aug 2020 VALIDATE Director Prof Helen McShane was phone interviewed by Cristiana Vagnoni (Biological Sciences and Health Advisor at UK Parliamentary Office of Science and Technology [POST], an office of both Houses of Parliament) for research for a four-page briefing on advances in vaccine technologies and policy approaches to stimulate and fast-track vaccine research and development 'given VALIDATE Network's role in supporting the development of vaccines for complex neglected pathogens'. Publication 29.09.21 "Advances in vaccine technologies" UK Parliament POSTnote https://post.parliament.uk/research-briefings/post-pn-0657/

21 Oct 2020, Helen McShane and Samantha Vermaak talk to Hannu Laang (GTBVP).

11 Nov 2020, Helen McShane and Samantha Vermaak talk to Ann Ginsberg (BMGF).

13 Nov 2020, Helen McShane and Samantha Vermaak talk to Katrin Eichelberg (NIH).

Oct 2020, VALIDATE Fellow Dr Rachel Tanner presented her research project findings at the TBScience 2020 meeting (part of 51st Union World Conference on Lung Health). and was invited as a panellist to talk about her work and experience as a female scientist for the 2020 Women of the Future Awards and the 2021 Oxford Women in Engineering, Science and Technology (OxWEST) Annual Symposium.

30 Jan 2021, VALIDATE is a partner of World NTD Day (https://worldntdday.org/partners/).

9 Mar 2021, Helen McShane and Samantha Vermaak talk to Ann Ginsberg (BMGF) and Ksenia Koon (CTVD) [this talk eventually led to the BMGF follow-on funding VALIDATE received].

1 Oct 2021, Helen McShane gave CTVD (virtual) workshop talk re VALIDATE-CTVD partnership & VALIDATE progree to date and future plans (including VALIDATE-CTVD joint grant calls).

28 Oct 2021, Helen McShane and Samantha Vermaak present about VALIDATE to GTBVP.

Nov 2021, Prof Susie Dunachie gave a talk about her P033 India-UK pump-priming project, as well as an intro to VALIDATE at FIS conference.

8-9 Nov 2021, Prof Samantha Sampson (VALIDATE Co-Director) gave presentation about VALIDATE at Birmingham University BBSRC Chaperonins workshop.

10 Jan 2022, Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with Mike Whelan at CEPI re possibilities to work together and for VALIDATE funding.

31 Jan 2022, Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with BMGF NTD team about VALIDATE; invited to submit small grant proposal (awarded Oct 2022).

18 Feb 2022, Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with Indian British High Commission re possibilities.

18 Mar 2022, Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with Francesco Berlanda Scorza at GSK to discuss possibilities to work together.

4 Apr 2022, Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with Fleming Fund Manager at Mott MacDonald re possibilities to work together.

26 May 2022 Samantha Vermaak virtual meeting with Suzanna Francis at IAVI who are looking for potential MTBVAC trial sites (where they could build capacity). Agreed she will add VALIDATE to their grant proposal and will get back in touch if successful.

17 Aug 2022 Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with Afrigen CEO Prof Petro Terblanche about possibilities of working together (Afrigen subsequently joined VALIDATE and participated in a joint grant proposal).

17 Sept 2022 Helen McShane, Samantha Sampson & Samantha Vermaak virtual meeting with SA MRC. Samantha Sampson invited to give talk re VALIDATE at MRC's Funders Forum 20-22 November 2022 (bringing together local and global funders in the SADC region).
Year(s) Of Engagement Activity 2017,2018,2019,2020,2021,2023
 
Description VALIDATE 1st Annual Meeting 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 5-6 Sept 2017 we hosted our first VALIDATE Annual Meeting, with 35 members attending from 5 countries (UK, India, Brazil, Thailand, and Uganda), plus our 3 Scientific Advisory Board members. We awarded 3 travel scholarships to facilitate LMIC attendees. This was a fantastic opportunity for our members to meet face-to-face, many for the first time, and it gave our Management Team a chance to showcase VALIDATE and its aims, vision, and upcoming grant calls, as well as all the opportunities and benefits we provide for our members. We had talks by world-leading researchers about each of our focus pathogens (to spark cross-pathogen thinking), presentations by our industry partners (to spark academic-industry collaborations), a speed-dating session (to facilitate new collaborations and networking), as well as a session to find out what workshops our members would find useful, and a Dragons Den style session where we showcased the type of pump-priming projects VALIDATE was looking for (to encourage successful ideas and applications from our members), followed by a session that facilitated cross-pathogen discussion and idea sharing to set up future pump-priming ideas. We had positive feedback from our members about the relevance and usefulness of this first meeting, and it was a very successful couple of days for jump-starting our Network and engaging our members in this new community.
Year(s) Of Engagement Activity 2017
URL https://www.validate-network.org/annual-meeting#collapse1-0
 
Description VALIDATE 2023 Annual Meeting 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 7-8 March 2023, VALIDATE held an in-person Annual Meeting for its members at BMA House, London. 95 members joined the meeting in-person, with a further >30 online (as the meeting was hybrid to increase access). Members came from 31 countries (21 LMIC): Argentina, Australia, Bangladesh, Canada, China, Colombia, Ethiopia, Ghana, India, Ireland, Ivory Coast, Japan, Malawi, Mexico, Mozambique, Nepal, Netherlands, New Zealand, Nigeria, Pakistan, Paraguay, South Africa, Spain, Sweden, Thailand, The Gambia, Tunisia, Uganda, UK, USA and Vietnam.

58% of delegates identified as female. 49% of delegates came from LMICs (many of the latter had travel grants from VALIDATE to enable them to join in-person).

The meeting was held across two days (with an ECR workshop the day before, on the 6th March) as an optional extra), and included three keynote talks, presentations by our Fellows and Pump-priming grant recipients about their projects, as well as a collaboration clinic for networking, a poster session, and a conference dinner (again, for networking).

Feedback has been extremely positive, with many members making new connections and sharing research ideas. We hope this will lead to new collaborations down the line.
Year(s) Of Engagement Activity 2023
URL https://www.validate-network.org/annual-meeting
 
Description VALIDATE 2nd Annual Meeting 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 6-7th November 2018, we held our 2nd Annual Meeting in York, UK. The meeting contained updates about VALIDATE, Prof Heidi Larson (LSHTM) talking about vaccine hesitancy, our pump-priming funded projects each giving an update about their research, as well as some animated and engaged networking to get our members meeting each other and sparking collaboration ideas. We had an ECR showcase, where our network's ECRs hosted posters outlining their current research and future career and research ambitions to facilitate and spark new collaborative relationships amongst our members and to network them with both their peers and senior scientists and PIs in VALIDATE. We also had an industry showcase where our industry members had posters highlighting their research experience and expertise, again to network them in with other members.

On day 2 we held a facilitated 'Ideas Laboratory' aiming to help our delegates think big to tackle vaccine development for our focus pathogens - what breakthroughs need to be achieved, what challenges need to be overcome, what do we need to start/stop/continue doing to achieve success? The room buzzed with energy as people exchanged ideas, gradually building up to concrete research project concepts that everyone worked collaboratively on to develop further, and some of which have been submitted as pump-priming grants in our subsequent round of funding.

62 delegates from 13 countries (Brazil, Cameroon, Ireland, Italy, Mexico, Mozambique, Nepal, Nigeria, South Africa, Thailand, Uganda, UK, USA) attended, as well as two members of our SAB. We awarded 9 travel scholarships to LMIC members to encourage and facilitate their attendance (7 to female scientists), 7 or which were to ECRs.

The meeting had excellent feedback from our members. We also used the meeting to produce three short videos, showcasing VALIDATE to encourage further new members to join and to highlight the benefits of VALIDATE membership (viewable at https://www.youtube.com/channel/UC4jnwrHLkvK-xJjYJYpnYog).

Outputs include:
- (from the 2nd AM and the VALIDATE melioidosis 2019 workshop) a new collaboration between Prof Susanna Dunachie (PI - Oxford), Assoc Prof Paul Brett (Nevada), Assoc Prof Mary Burtnick (Nevada), Prof Helen McShane (Oxford) and Prof Narisara Chantratita (Mahidol) applied for and were awarded an MRC Biomedical Catalyst DPFS / US Defense Threat Reduction Agency grant, "Developing a vaccine for Burkholderia pseudomallei - a Phase I Clinical Trial" of £3,238,214 in 2020 (to 2023). This will be the first ever human clinical trial of a melioidosis vaccine.
This new collaboration have also been awarded a Defense Threat Reduction Agency (DTRA) Subaward "Developing a vaccine for Burkholderia pseudomallei - a Phase I Clinical Trial" (Brett PI, Dunachie Oxford subcontracting PI to value of $733,976.61(£524,269) to Dunachie lab) for 2020-2021, and a US Defense Threat Reduction Agency 2020-2021 award "Optimization of lead melioidosis and glanders vaccine formulations" Prof Paul Brett University of Nevada, Reno PI with sub-contract award to Dunachie, Oxford PI, (awarded in principle, awaiting contract) $733,976.61 USD (£524,269).
- During the 2nd Annual Meeting Facilitated 'Ideas Laboratory', Dr Barbara Kronsteiner-Dobramysl (University of Oxford ECR) was approached by Prof Javier Sanchez (IPN, Mexico) who also had an interest in immunometabolism and they formed a new collaboration, being successfully awarded a VALIDATE pump-priming grant in 2019.
- A new collaboration was formed between Dr Julen Tomas Cortazar and Dr Siobhan McClean (UCD), Dr Louise Gourlay (University of Milan) and Prof Susanna Dunachie (University of Oxford), which resulted in a successful pump-priming grant application in 2019.
Year(s) Of Engagement Activity 2018
URL https://www.validate-network.org/article/validate-2nd-annual-meeting-report
 
Description VALIDATE 3rd Annual Meeting and VALIDATE-BSI 2019 Conference 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact From 30th September to 3rd October 2019, VALIDATE held three back-to-back events, our 2nd Early Career Researcher (ECR) Workshop (reported separately elsewhere), the 3rd VALIDATE Annual Meeting, and a VALIDATE-BSI Conference 'Vaccine Development for Neglected Intracellular Pathogens', at The Drum at Wembley, London. We welcomed just under 90 researchers, almost all VALIDATE members, from 19 countries (13 LMIC) worldwide.

On Tuesday 1st October the VALIDATE NMB and SAB held a closed face-to-face meeting, and then we kicked off the 3rd VALIDATE Annual Meeting. VALIDATE Director Prof Helen McShane welcomed our 78 delegates and Network Manager Samantha Vermaak gave an update on VALIDATE's activities over the last two years, and an insight into what's ahead for the Network, highlighting our final pump-priming call that closes on 22nd October 2019. We then had talks by our most recently funded round (#3) of pump-priming projects, with Prof Myron Christodoulides (University of Southampton) on 'Protective efficacy of conserved Leishmania hypothetical proteins against visceral leishmaniasis', Dr Barbara Kronsteiner-Dobramysl (University of Oxford) on 'Metabolic reprograming of skin microenvironment for improved BCG vaccine efficacy', Dr Sasha Larsen (IDRI) on 'Development of an RNA based vaccine against Mycobacterium tuberculosis', Dr Elena Stylianou (University of Oxford) on 'Developing a mouse model of diabetes to evaluate vaccines for TB and melioidosis', and Dr Julen Tomás Cortázar (University College Dublin) on 'Elucidating the T-cell epitopes and T-cell responses of two B.pseudomallei vaccine antigens'. These interesting talks showcasing the great research VALIDATE has coming up in the next 12 months were followed by Emilie Karafillakis (LSHTM) discussing the 'Monitoring and addressing challenging risks, rumours and reputations across the GCRF Vaccinology Networks' project co-funded by VALIDATE, IMPRINT and HIC-VAC, which aims to help all our members with vaccine hesitancy and media/online reputation management.

After lunch, our round #1 pump-priming projects (funded in November 2017) discussed their completed research, with Sally Hayward (SGUL/LSHTM) discussing 'Enhancing BCG efficacy: the Social Technology Lab Initiative', Dr Javier Salguero Bodes (PHE) 'The effect of BCG vaccination in immune responses against visceral leishmaniasis in a natural (canine) model of infection', Asst Prof Steven Smith (LSHTM) 'How do functional and metabolic characteristics of trained monocytes affect their anti-bacterial activity?', Dr Panjaporn Chaichana (MORU) 'Establishment of a functional assay panel to evaluate the role of antibodies in defence against melioidosis and tuberculosis', Dr Rajko Reljic (SGUL) 'Overcoming innate immune tolerance in the respiratory tract for optimal vaccine design', and Prof Bernardo Villarreal-Ramos (Aberystwyth University & APHA) 'Natural variation of the bovine lymph node microenvironment and its possible effect on BCG immunogenicity'. These projects had had some interesting scientific outputs that we look forward to seeing in publications soon, and some great career outcomes!

After a break, our Keynote speaker, Affiliate Prof Rhea Coler (IDRI) spoke about 'Novel TB and leprosy vaccines: from the lab bench to clinical trials' from an industry perspective. We closed the formal part of the day with a panel discussion by our funded pump-priming project awardees about their experiences applying for and receiving VALIDATE funding, and the lessons they have learned on the way.

Many of the delegates then joined a behind-the-scenes tour of Wembley Stadium, with two engaging and entertaining guides who soon had us all trying out the press box and running out of the tunnel into the stadium as two teams. The day closed with those delegates who wanted to chat further visiting nearby Zizzi's for dinner together.

The last morning of the Annual Meeting opened on Wednesday 2nd October with talks from two round #2 pump-priming projects (funded August 2018), with Mohamed Osman (University of York) on 'Identification of Leishmania donovani and Mycobacterium tuberculosis- derived proteins on the surface of infected macrophages that are associated with ADCC induction', and Prof Susie Dunachie (University of Oxford) with Asst Prof Jackie Cliff (LSHTM) and Patpong Rongkard (Oxford/MORU) on 'Vaccines to target people with diabetes: characterising the pathways of immune response to M.tuberculosis and B.pseudomallei in people with diabetes compared to non-diabetics'. We look forward to seeing the final data of these projects soon.

This session then moved into talks by our VALIDATE Fellows, with Prof Samantha Sampson (University of Stellenbosch) on behalf of Dr Jomien Mouton (who is on maternity leave) talking about 'Identification of latency associated antigens and biosignatures associated with Mycobacterium tuberculosis' and Dr Rachel Tanner (University of Oxford) about 'Characterising the BCG-induced antibody response to inform the design of improved vaccines against M.tuberculosis, M. leprae and M.bovis'.

After a coffee break, we went into our final session - funder talks aiming to help our members find future funding opportunities. Prof Helen Fletcher spoke about the Global Challenges Research Fund (GCRF) and Newton Fund, Dr Peter Hart on Wellcome Vaccines Funding, Dr Martin Broadstock on MRC Funding Opportunities, and Dr Ceri-Wyn Thomas on BBSRC funding - all very relevant funders for our members and our research field. VALIDATE Director, Prof Helen McShane, then wrapped up a very successful 3rd Annual Meeting; congratulations to Dr Sasha Larsen (IDRI) for winning the 'Best Talk by an ECR' prize.

After lunch, we moved into the VALIDATE-BSI 2019 Conference 'Vaccine Development for Neglected Intracellular Pathogens', with Prof Helen McShane (University of Oxford/VALIDATE) and Dr Doug Brown (British Society for Immunology [BSI]) each giving a welcome address to the Conference delegates. Our first session talks centred on tissue-specific immunity, host-pathogen interactions, macrophages and the microenvironment, chaired by Prof Andrea Cooper (University of Leicester). Our first Keynote Speaker, Prof Clare Lloyd (ICL), spoke about how 'Local and environmental cues dictate the nature of the pulmonary immune response', and was followed by Dr Barbara Kronsteiner-Dobramysl (University of Oxford) on 'Diabetes alters cellular metabolism of T cells: implications for vaccine development', Dr Chidchamai Kewcharoenwong (Khon Kaen University) on ' Glibenclamide alters cytokine production of primary human monocytes from diabetic individuals against Mycobacterium tuberculosis infection', and Dr Isadora Lima (FIOCRUZ) on 'Canine visceral leishmaniasis: the role of tissue-specific immunity in the progression of disease'. After a quick break, our second Keynote Speaker for this session, Prof Tracy Hussell (University of Manchester), spoke on 'Innate sensing of lung health and disease', followed by Dr Lisa Stockdale (University of Oxford) on 'Cytomegalovirus as a correlate of risk for TB?'.

Next followed an interesting 45 minutes of 3-minute turbo talks by poster hosts, with all the speakers impressively sticking to time! Asst Prof Kiran Afshan (Quaid-i-Azam University) spoke on 'Cutaneous Leishmaniasis in Khyber Pakhtunkhwa, Pakistan', Dr Anthony Afum-Adjei Awuah (KCCR) on 'Why fix when you can prevent; towards TB Vaccines Research in Ghana', Dr Federico Carlos Blanco (INTA) on 'The isolate Mb04-303 induce a proinflammatory innate immune profile and is a potential candidate for an attenuated mutant vaccine against bovine tuberculosis', Assoc Prof Mary Burtnick (University of Nevada, Reno) on 'Optimization of melioidosis subunit vaccines using Burkholderia thailandensis E555', Dr Panjaporn Chaichana (MORU) on 'Identification of protective antibody profiles for vaccine development against melioidosis', Assoc Prof Narisara Chantratita (Mahidol University) on 'Risk of Burkholderia pseudomallei infection, host immune response and biomarkers for melioidosis', Asst Prof Ganjana Lertmemongkolchai (Khon Kaen University) on 'Metformin-induced suppression of IFN-a expression via mTORC1 signalling following seasonal vaccination is associated with impaired antibody responses in diabetes mellitus', Dr Olayinka Osuolale (Elizade University) on 'Leishmaniasis in patients attending a medical health center in the rural community of Ilara-Mokin, Ondo State, Nigeria', Asst Prof Vijay Kumar Prajapati (Central University of Rajasthan) on 'Evaluation of immunogenicity of the engineered multi-epitope subunit vaccine against visceral leishmaniasis infection', Dr Lok Bahadur Shrestha (BPKIHS) on 'Emerging Clinical Microbiologist looking for research opportunity in TB Vaccinology', Dr Suwarti (EOCRU) on 'Intranasal boosting regimen of Human Parainfluenza Virus Type 2-Ag85B vaccines induces superior CD4 immune response against Mycobacterium tuberculosis', Dr Julen Tomás Cortázar (University College Dublin) on 'Elucidating the mechanisms of protection of the Burkholderia pseudomallei antigen, BpOmpW', and Chathika Weersuriya (LSHTM) rounded the session off with 'Mathematical modelling of the epidemiological impact, cost-effectiveness and budget impact of novel tuberculosis vaccines on multi-drug resistant tuberculosis'. This energetic session was followed by a group photo, and a drinks reception with posters, before everyone headed into the Conference Dinner.

The second day of the conference on Thursday 3 October saw the start of our second session on Biomarkers, chaired by Prof Helen Fletcher (LSHTM). Our first Keynote Speaker, Dr January Weiner (Berlin Institute of Health) spoke about 'Biomarkers in tuberculosis and individual variability of the host'. January was followed by Dr Fatoumatta Darboe (MRC The Gambia) discussing 'Predicting and diagnosing TB disease in HIV-infected persons' and Dr Eduardo Milton Ramos Sanchez (USP) on 'Altered expression of microRNAs in infections with cutaneous and visceral strains of Leishmania'. After a break, our second Keynote Speaker for this session, Prof Stephen Gordon (University College Dublin) discussed the 'One Health route to exploring virulence in the Mycobacterium tuberculosis complex', with Hannah Painter following on 'Development of an ex vivo mycobacterial inhibition assay using murine lung cells for tuberculosis vaccine testing' and Asst Prof Tufária Mussa (Eduardo Mondlane University) on 'The fusion protein Ag85B-HBHA (FP1) elicits IFN-?, IL-2 and TNF-a in conventional T cells of Tuberculosis contacts with latent infection over time'.

After lunch, we moved into our final session 'Latest vaccine developments', chaired by Prof Helen McShane (University of Oxford). Keynote Speaker Assoc Prof Paul Brett kicked us off discussing 'Development of multivalent subunit vaccines for immunization against melioidosis', followed by Dr Sophie Rhodes (LSHTM) on 'Dose finding for new vaccines: the role of immunostimulation/immunodynamic modelling' and Dr Deepa Paliwal (University of Reading) on 'Progress in a reverse vaccinology approach for bovine TB'. After coffee, our final Keynote Speaker, Prof Mark Hatherill (University of Cape Town) spoke about 'Major advances and next steps for tuberculosis vaccine development', after which Dr Justice Boakye-Appiah discussed 'Mycolactone based vaccine against Buruli ulcer disease', and then Asst Prof Rebecca Harris (LSHTM) gave our last talk on 'Potential impact of new tuberculosis vaccines in China, South Africa and India and implications for vaccine development'. VALIDATE Director Prof Helen McShane gave a brief wrap-up, and the prizes - congratulations to Dr Panjaporn Chaichana for winning Best ECR Poster and Dr Sophie Rhodes for winning Best ECR Talk at the Conference - and then we were done! Four great days of thoroughly interesting talks, and fantastic networking opportunities were over.

Slides from the presentations (where the speaker was happy to share them) are accessible to our members on the VALIDATE Document Library. Seven video compilations of interviews of our members at the events were created and added to the VALIDATE YouTube channel (https://www.youtube.com/channel/UC4jnwrHLkvK-xJjYJYpnYog).

Feedback from our members included:
'Great talks, great opportunity to network. Really nice, collegiate atmosphere.'
'Fantastic idea to put together M.tb, leishmania and B.pseudomallei and see cross-translational approaches in vaccine research against these pathogens.'

Outputs from this meeting include:
- Dr Mark Chambers (University of Surrey) obtained research reagent and know-how from Assoc Prof Paul Brett (University of Nevada), and has started collaborating with Emilie Karrafillakis (LSHTM Vaccine Confidence Project).
- Dr Federico Blanco (INTA, Argentina) was awarded a travel scholarship to attend the VALIDATE 2019 events. Through this attendance he (and his home research group, including Dr Fabiana Bigi) have established a collaboration with Dr Bernardo Villarreal-Ramos (APHA). They submitted an unsuccessful application for pump-priming funding to VALIDATE, but have since been awarded funds from an Argentine agency and will start their project in March 2020. In this collaborative project, Bernardo will be training the INTA researchers in techniques to study the innate bovine response to TB.
- Dr Julen Tomas Cortazar and Dr Siobhan McClean (UCD) have been collaborating with Assoc Prof Paul Brett (University of Nevada) since this Annual Meeting.
Year(s) Of Engagement Activity 2019
URL https://www.validate-network.org/article/validate-2019-events-report
 
Description VALIDATE 4th Annual Meeting 2021 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact VALIDATE hosted our 4th Annual Meeting across three days in Sept, Oct and Nov 2021. This was held virtually, with each session in a different time-zone so that all VALIDATE members across the globe could access at least one day live. VALIDATE's pump-priming and Fellowship awardees presented their projects in this closed meeting for VALIDATE members. In addition each session had a welcome from a local UK High Commission/Newton representative (details below). Recordings of the talks are available via our Hub for all members to access.

Day 1 (time zone for SE Asia/Australasia): Chaired by Prof Mitali Chatterjee, IPMGER, India. ~80 attendees from 19 countries: Bangladesh, Belgium, Brazil, Cameroon, China, Ethiopia, Ghana, India, Indonesia, Ireland, Malaysia, Nigeria, Pakistan, Sri Lanka, South Africa, Switzerland, The Gambia, Uganda, UK. Welcome address by Dr Himangi Bhardawaj MBE, Newton Fund India.

Day 2 (time zone for Europe/Africa): Chaired by Prof Samantha Sampson, Stellenbosch University, South Africa. 51 attendees from 21 countries: Argentina, Bangladesh, Belgium, Brazil, Colombia, Ghana, India, Ireland, Japan, Nigeria, Pakistan, Paraguay, Portugal, Sri Lanka, South Africa, Sweden, Thailand, Uganda, UK, USA, Vietnam. Welcome address by Mr Anthony Phillipson, UK High Commission in South Africa.

Day 3 (time zone for the Americas): Chaired by Dr Maria Adelaida Gomez, CIDEIM, Colombia. 70 attendees from 22 countries: Argentina, Bangladesh, Belgium, Brazil, Colombia, Ghana, India, Ireland, Japan, Kenya, Mexico, Netherlands, Nigeria, Pakistan, Paraguay, Portugal, Serbia, South Africa, The Gambia, Uganda, UK, USA. Welcome address by Colin Martin-Reynolds CMG, British Ambassador to Colombia.
Year(s) Of Engagement Activity 2021
URL https://www.validate-network.org/annual-meeting-2021#collapse3064486
 
Description VALIDATE BCG100 Programme 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact To celebrate the centenary, in 2021, of the first use of the Bacille Calmette-Guérin, or 'BCG', vaccine, VALIDATE ran a 'BCG100 Programme' throughout 2021. BCG is the first and, still a century later, only vaccine approved for humans that protects against tuberculosis (TB).

We marked the centenary with a programme of events and activities to raise awareness about TB, and to highlight the work researchers around the world are doing to try to develop a better vaccine against TB. The campaign also aimed to provide resources to allow our members to undertake their own public engagement.

The BCG100 project received additional funding via a small grant from the University of Oxford Enriching Engagement scheme funded by the Wellcome Trust.

The BCG100 Programme contained two aspects:
1. Online Talks: A series of online talks, aimed at the public, to raise awareness and to answer questions about vaccines, vaccine hesitancy, and vaccines for TB. We delivered two public lectures, and one school talk. Attendees were very engaged during the Q&A sessions, and feedback has been overwhelmingly positive. The three events were (all virtual due to COVID pandemic):

'BCG Then and Now' (online public lecture) - 24 March 2021 (World TB Day)
Prof Paul Fine (LSHTM) and Prof Helen McShane (Oxford) discussed the initial development of the BCG vaccine in 1921 and its story through to the modern-day fight against TB and the future challenges researchers face in replacing BCG with a more effective vaccine. 150 attendees from around the world. Recording available on VALIDATE YouTube channel (342 views).

BCG100 For Schools - 11 May 2021
A follow up/spin-off from our very successful VALIDATE For Schools event, Prof Michele Miller and Dr Wynand Goosen from Stellenbosch University talked to students from around the world about their careers, their TB vaccine research, and their work with African wildlife. ~60 children attended mostly from the UK. Recording available on VALIDATE YouTube channel (72 views).

'TB and the use of BCG in Animals: Does it matter to people?' (online public lecture) -9 June 2021
Prof Michele Miller and Prof Glyn Hewinson discussed why effective vaccine programmes for animals are essential to the health and wellbeing of humans as well, and the challenges that we still face in making such programmes a reality. 78 people (42 non-members) attended from 53 institutions in 22 countries (Argentina, Brazil, China, Ethiopia, Germany, Ghana, India, Ireland, Malaysia , México , Nigeria, Paraguay, Poland, South Korea, South Africa, Spain, Sri Lanka , Turkey, Uganda, United Kingdom, United States, and Zambia). Recording available on our YouTube channel (173 views).

2. Video Games
The VALIDATE team and members created three educational computer games, 'BCG Adventures', aiming to raise interest and awareness of vaccine development and deployment, particularly for TB. They are fun and educational for all ages but designed as educational tools for Key Stage 3 students (11-14 years). Each game begins with an anonymous survey asking for age, country of origin and previous knowledge of vaccines and ends with a quiz about the information learnt whilst playing the game. The games are:
a. Albert and Camille's Little Lille Adventure (Little Lille) - a classic adventure game where the player explores the war-torn city of Little Lille in 1917. Taking the part of both Albert Calmette and Camille Guérin, players interact with the city's citizens to obtain the necessary supplies they need for their important vaccine research. As items are collected, players learn about the methods used to create BCG, the first vaccine against tuberculosis.
b. Vaccine Delivery Adventure (VDA) - a classic side-scrolling action game where as the player is a healthcare worker trying to overcome the challenges of delivering the BCG vaccine to communities that need it. As they deliver the vaccine in different locations, the player learns about the BCG vaccine and the challenges of protecting communities against tuberculosis.
c. Welcome to Your Immune System (WTYIS) - an arcade-style game in which the player defends the human body against an attack from Mycobacteria tuberculosis. The player can use macrophages, killer T cells and cytokine storms in the fight - and learn what these actually are and how they are used by our immune system.

The games have an accompanying document written for teachers, with further information about both the games and the content, to support them in using the games in their lessons to spark discussion.

The games are open-access and free to play (and to use, hopefully by teachers for classes and researchers for public engagement/outreach) via Itch.io and STEAM.

>27k people from 99 countries had engaged with the games within the first year. STEAM player reviews are respectively 86%, 50% and 92% positive. The games have been rated overall by players as 4.1, 3.6, 4.1 (average 3.9) out of 5 for 'fun', and 3.9, 3.4, 3.9 (average 3.7) out of 5 for 'informative'. End of game quiz scores are respectively 3.8, 2.4, and 3.1 out of 5. Clearly the Vaccine Delivery Adventure was the weaker game, and we have learned from this for future outreach efforts.

One of the aims of the BCG100 Programme was to encourage a better understanding and increased confidence in vaccines, and to encourage young people to consider a career in STEMM. Obtaining data on these was not possible in part due to lack of feedback from our main schools event and the lack of face-to-face engagement/events due to the COVID pandemic. We hope to get more data as the games are used by researchers in face-to-face public engagement as the COVID-19 pandemic eases.
Year(s) Of Engagement Activity 2021
URL https://www.validate-network.org/bcg100
 
Description VALIDATE Leprosy Video 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Public/other audiences
Results and Impact As part of our activities marking World NTD Day and World Leprosy Day in 2021, we produced a short video about leprosy. In 'A Vaccine for leprosy?', Dr Hua Wang (new PI at Glasgow University) talks about leprosy, how it can be tackled and what VALIDATE is doing to develop vaccines to help eradicate the disease. The video is publically available on our YouTube channel at https://www.youtube.com/watch?v=1fzDWmAc2YY and has had 6.3k views.
Year(s) Of Engagement Activity 2021,2022,2023
URL https://www.youtube.com/watch?v=1fzDWmAc2YY
 
Description VALIDATE LinkedIn 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact We have set up a closed LinkedIn VALIDATE group with three aims:
1. To make it easier for our members to connect to each other.
2. To create a further social media forum on a different platform for dissemination of relevant and interesting news as well as VALIDATE outputs, to reach members who may not be using Twitter.
3. To add a way to keep up to date with outputs and news from our members e.g. job vacancies are often posted to LinkedIn, that we can then share and also publicise on our website and Twitter accounts.

Currently 329 of our members have connected and are now following our account.
Year(s) Of Engagement Activity 2018,2019,2020,2021,2022,2023
URL https://www.linkedin.com/in/validate-network/
 
Description VALIDATE Seminars 
Form Of Engagement Activity A talk or presentation
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact VALIDATE aims to host regular seminars by our members about their research to inform members and the general public, and to spark new research ideas and collaborations. These are live-streamed online to enable all our members and the general public, no matter where they are based in the world, to join and benefit from the information, ideas, and findings being disseminated.

Our first talk was held on 6 Dec 2017, by Assoc Prof Helen Fletcher and Dr Andrea Zelmer at LSHTM, entitled "Correlates of risk of TB disease and their back-translation into animal models", and was viewed by c.43 people.

Our second talk was held on 15 Feb 2019, by Prof Helen McShane and Dr Rachel Tanner at University of Oxford, entitled "Developing a better vaccine against TB" and was viewed by 52 unique viewers.

Our 2020 Summer Seminar Series involved:
30 June 2020 'Developing a vaccine for Melioidosis: Probing the protective immune response'
By Prof Siobhán McClean and Dr Julen Tomás Cortázar (UCD, Ireland). 52 people from 16 countries registered for the event (India, Nigeria, Bangladesh, USA, UK, UCD, Ghana, Brazil, Italy, Hungary, Pakistan, Camaroon, Algeria, Thailand, Vietnam, Indonesia).

23 July 2020 'Melioidosis and Glanders' by Prof Mary Burtnick (University of Nevada, Reno School of Medicine, USA). 35 people registered to attend from 15 countries (Vietnam, India, Hungary, Ireland, UK, Sri Lanka, USA, Nigeria, Cameroon, Ghana, China, Pakistan, Bangladesh, Thailand, Malaysia).

29 July 2020 'Nonspecific factors in the immunopathogenesis of leishmaniasis' by Assoc Prof Hiro Goto and Dr Eduardo Milton Ramos Sanchez (Universidade de Sao Paulo, Brazil). 12 people attended from 6 countries (UK, India, Sri Lanka, Hungary, Pakistan, Brazil).

01 September 2020 'Efforts to understand the immunology of bovine TB' by Prof Bernardo Villarreal-Ramos (University of Aberystwyth and APHA) and Dr Amanda Gibson (Aberystwyth University). This seminar was pre-recorded, with 57 people registering to watch the recording from 12 countries (Brazil, Mexico, Argentina, UK, India, Uganda, Algeria, Nigeria, South Africa, Tanzania, Ghana, Ethiopia).

Our 2020 Winter seminar series included:
24 November 2020 'Mtb in the host Persisters and PE&PPE Proteins' by Prof Samantha Sampson and Dr Jomien Mouton (Stellenbosch University, South Africa). 60 people joined the seminar from 18 countries (Hungary, Brazil, UK, Argentina, India, Mexico, South Korea, India, Ghana, Uganda, Italy, South Africa, Malaysia, Germany, Australia, Sri Lanka, USA, Lithuania). 111 views on YouTube.

3 December 2020 'Computational approaches in structural vaccinology - Structural biology for antigen-based therapeutics & diagnostics' by Prof Giorgio Colombo (University of Pavia, Italy) and Asst Prof Louise Gourlay (University of Milan, Italy). There were 31 attendees from 20 countries (UK, Brazil, Bangladesh, Vietnam, Mexico, Burkina Faso, UK, USA, Ghana, Kenya, India, Uganda, Tanzania, Nigeria, Pakistan, Italy, Malaysia, Ireland, Cameroon, Sri Lanka). 118 views on YouTube.

15 December 2020 'Immune profiles of healing in cutaneous leishmaniasis, what to target and when?' by Dr Maria Adelaida Gomez (CIDEIM, Colombia). 39 people attended from 15 countries (Nigeria, USA, India, Colombia, Sri Lanka, Morocco, Vietnam, Belgium, UK, Uganda, Algeria, Pakistan, Spain, Brazil, Cameroon, Ghana, Lithuania). 108 views on YouTube.

2021 Industry Seminar Series:
11 March 2021 'Capturing and recreating recombinant polycloncal antibodies for passive immunization to treat difficult to treat infections' by Dr Sheila Keating of GigaGen. 18 people attended from 9 countries joined (Uganda, UK, Ghana, India, USA, Colombia, Nigeria, South Africa, and Thailand). 83 views on YouTube.

27 April 2021 'Addressing pathogen and human diversity' by Graham Clarke, Chair and former CEO of ImmBio. ~30 attended with 60 registered from 25 countries (Austria, Brazil, Burkina Faso, China, Croatia, Ethiopia, Gambia, Germany, Ghana, India, Indonesia, Ireland, Mexico, Nigeria, Nigeria , Pakistan, Paraguay, South Africa, Tanzania, Thailand, The Gambia, Uganda, United Kingdom, United States and Venezuela). 25 views on YouTube.

15 Jul 2021 'Tuberculosis Infection and T cell-Mediated Immunity' by Dr Yupei Xiao, Medical Science Liaison, Oxford Immunotec. This was the last in our series of Industry Seminars. 64 attended with people registering from Argentina, Bangladesh, Belgium, Brazil, Bulgaria, China, Colombia, Ethiopia, France, Germany, Ghana, Hungary, India, Indonesia, Ireland, Japan, México , Nigeria, Pakistan, South Africa, South Korea, Sri Lanka, Sweden, Switzerland, Tanzania, Thailand, The Gambia, Uganda, United Kingdom, United States, Venezuela and Zambia. 302 views on YouTube.

30 November 2021 - seminar by Dr Yumi Maeda (Leprosy Research Center, NIID, Japan) 'Vaccine development and sero-diagnosis of leprosy'. 23 people attended from 14 countries (Japan, Peru, United Kingdom, Ghana, Sri Lanka, Nigeria, Portugal, Albania, India, South Africa, Senegal, Japan, Burkina Faso).

6 Apr 2022 "A vaccine for leishmaniasis?" by Prof Mitali Chatterjee (IPGMER, India) and Dr Mohamed Osman (University of York, UK) provided members with an overview and update on the current state of research into vaccines for leishmaniasis. 55 online attendees from 21 countries (14 LMIC). 137 views on YouTube.

18 May 2022 "A vaccine for melioidosis?" by Prof Chiranjay Mukhopadhyay and Dr Somasish Ghosh Dastidar (MAHE, India) discussed the prospects for a melioidosis vaccine. 38 online attendees from 16 countries (13 LMIC). 96 views on YouTube.

22 June 2022 - ECR Summer Showcase 1 online seminars - Dr Nguyen Ho Lam (University of Medicine & Pharmacy, Vietnam), Dr Nidhi Gupta (CURAJ, India) and Dr Rachel Tanner (University of Oxford, UK) gave short talks on their research. 36 online attendees from 16 countries (11 LMIC).

28 June 2022 "A vaccine for leprosy?" - Dr Khushboo Slater (University of Surrey, UK) and Dr Chyntia Diaz Acosta (IICS, Paraguay) discussed the prospects for a leprosy vaccine. 40 online attendees from 24 countries (14 LMIC). 110 views on YouTube.

13 July 2022 - ECR Summer Showcase 2 online seminars - Asst Prof Nargis Khan (University of Calgary, Canada), Dr Laura Sibley (UKHSA, UK) and Dr Cristian Segura-Cerda (CIATEJ, Mexico) gave short talks about their research. 31 online attendees from 25 countries (20 LMIC).

16 Nov 2022 "A flexible and efficient platform to develop a nanovaccine against Burkholderia pseudomallei" by Prof Alfredo G Torres (UTMB, USA). 31 online attendees from 13 countries (8 LMIC). 83 views on YouTube.

14 Dec 2022 "Targeting Immunometabolism in Host Defense Against Mycobacterium Tuberculosis" by Dr Luciana Balboa (CONICET, Argentina). 53 online attendees from 26 countries (16 LMIC). 70 views on YouTube.

27 Jan 2023 Prof Makram Essafi "Targeting FOXO3 transcription factor as a host-directed approach to enhance the efficacy of BCG against Mtb" & Prof Dhafer Laouini "Vaccine against human leishmaniasis: Institut Pasteur de Tunis contribution". 31 online attendees from 16 countries (9 LMIC). 30 views on YouTube.

Nearly all the talks were recorded and these recordings are available to VALIDATE members via the VALIDATE Hub; open-access talk recordings are also shared on the VALIDATE YouTube Channel.

In addition, in 2020, Dr Julen Tomas Cortazar (UCD, Ireland) made three videos talking about his research on T-cell inducing vaccines, in English, Spanish and Basque respectively. These have 512 YouTube views in total.

One of our ECRs, Wilfred Aniagyei (KNUST/KCCR) said 'VALIDATE online seminar gave me insight into other research methodologies that helped with my current research' while Christine Petersen (University of Iowa) commented 'VALIDATE has provided some wonderful seminars and other platforms for scientific discourse during this year [2020] of limited ability to otherwise interact with our scientific peers. This has been very helpful to maintain productivity and connection.'

Prof Samantha Sampson's seminar led to an invitation to speak at University of Surrey, which has led to several follow-on conversations about possible collaborations (with Suzie Hingley-Wilson, VALIDATE member). One of her PhD students has been assisting one of Prof Sampson's MSc students with protocol development.
Year(s) Of Engagement Activity 2017,2018,2019,2020,2021,2022,2023
URL https://www.validate-network.org/seminars
 
Description VALIDATE Twitter account 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 28 July 2017 we launched our VALIDATE Twitter account @NetworkVALIDATE. We post regularly about news, opinions and activities relevant to our Network members, including grant calls, training opportunities, events, journal articles, opinion pieces, tips/advice, and disease statistics from VALIDATE and relevant external organisations, to promote both our activities and items that could help our members advance their research and careers - or inform general public followers about our research, vaccine development, vaccine hesitancy etc. We highlight papers authored by our members, raising their profile and potential citations, as well as papers relevant to our members' research, making keeping up to date with new research in the field easier for our members. We also have social media engagement activities for World Leprosy Day, World NTD Day, International Day of Women & Girls in Science, World TB Day, Immunization Week and others. To date we have 1,140 followers, which we hope will continue to increase, forming an engaged community.
Year(s) Of Engagement Activity 2017,2018,2019,2020,2021,2022,2023
URL https://twitter.com/NetworkValidate
 
Description VALIDATE YouTube Channel 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 4 Dec 2018 VALIDATE launched its You Tube Channel, with videos of our members talking about VALIDATE and the benefits of membership, to promote our network in another format and to hopefully encourage new members to join VALIDATE. https://www.youtube.com/channel/UC4jnwrHLkvK-xJjYJYpnYog.

We added 7 further videos of member interviews taken during our 3rd Annual Meeting and ECR workshop (2019) e.g. 'How VALIDATE is accelerating vaccine development', 'How VALIDATE is capacity building' as well as overview videos of our Annual Meetings and the benefits of being a VALIDATE member.

In 2020/2021 we expanded our Channel aims to include educating the general public about VALIDATE's focus pathogens and research, and encouraging teenagers towards careers in STEM. We added an interview with Prof Helen McShane for World TB Day, three videos (English, Spanish, Basque) by Dr Julen Tomas Cortazar explaining his research, a recording of our first VALIDATE for Schools talk by Prof Helen McShane and Dr Rachel Tanner (attended live by >200 school kids), three open-access scientific seminars by VALIDATE members based in South Africa, Italy and Colombia, and a video by Dr Hua Wang about leprosy for World Leprosy Day 2021. The latter is our most successful video, with 6.3k views.

In 2021/2022 we added recordings of our two BCG100 public talks, 3 industry open-access seminars, a further VALIDATE for Schools recorded talk, two trailers for our BCG Adventure computer games, and an original video 'A few things you might not know about Leishmaniasis' (as part of World NTD Day 2022), as well as further member scientific open-access seminars.
Year(s) Of Engagement Activity 2018,2019,2020,2021,2022,2023
URL https://www.youtube.com/channel/UC4jnwrHLkvK-xJjYJYpnYog
 
Description VALIDATE for Schools 
Form Of Engagement Activity Participation in an activity, workshop or similar
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Schools
Results and Impact On 9 December 2020 over 200 students in schools around the UK joined VALIDATE members Prof Helen McShane and Dr Rachel Tanner on Microsoft Teams for the first VALIDATE for Schools virtual event. Helen and Rachel talked to GCSE and A Level students about their research, tuberculosis vaccine development and immunology, and their careers in STEMM. They also responded to some fantastic questions from the students on subjects that ranged from scientist salaries to CRISPR. The event had fantastic feedback from the teachers involved (who would like more such events). The talk was recorded and is available on the VALIDATE YouTube channel at https://www.youtube.com/watch?v=oTrp0JnQ2LI.

ON 11 May 2021, Prof Michele Miler and Dr Wynand Goosen (Stellenbosch University) gave a further VALIDATE for Schools virtual talk to ~60 children.
Year(s) Of Engagement Activity 2020,2021
URL https://www.youtube.com/watch?v=oTrp0JnQ2LI
 
Description VALIDATE in the spotlight - in writing 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact In 2020, VALIDATE Fellow Dr Rachel Tanner took part in BSI's 'a day in the life of a vaccine researcher' blog. She was also interviewed for a 500WomenScientists article, and contacted by school student in California who interviewed her for a presentation on 'scientists that don't fit the conventional mold'

12 May 2020, '10 minutes with Prof Helen McShane' in BMJ Leader bmjleader.bmj.com/content/early/2020/05/10/leader-2020-000268

29 Apr 2020, VALIDATE highlighted in BSI's 'protecting the world' report on UK vaccinology www.immunology.org/sites/default/files/BSI_Celebrate_vaccines_report_2020_FINAL_0.pdf 29.04.20

Apr 2020, World Immunization Week, VALIDATE is part of BSI's Instagram campaign www.immunology.org/news/world-immunisation-week-2020

11 Feb 2020, VALIDATE blog post for International Day of Women and Girls in Science, chatting to five of our female members about their research, inspiration, and advice/tips they would give to girls wanting to aim for a career in scientific research: www.validate-network.org/article/international-day-of-women-and-girls-in-science-2020

31 Jan 2020, VALIDATE First World NTD Day blog post, discussing NTDs and new NMB member Dr Alvaro Acosta-Serrano's research on leishmaniasis: www.validate-network.org/article/world-ntd-day-2020

27 Jan 2020, VALIDATE World Leprosy Day blog post, flagging the news and views for this day from around the world: www.validate-network.org/article/world-leprosy-day-2020

22 Mar 2019, VALIDATE Director Prof Helen McShane spoke to Phys.org ahead of World TB Day https://phys.org/wire-news/314702574/a-new-tb-vaccine-is-within-reach-prof-helen-mcshane.html

8 Mar 2019, VALIDATE blog post for International Women's Day 2019 interviewing 5 of our female scientists https://www.validate-network.org/article/international-womens-day-2019

27 Jan 2019, VALIDATE World Leprosy Day blog post, talking to two ECR researchers working on leprosy vaccine development: www.validate-network.org/article/world-leprosy-day-2019

26 Nov 2018, VALIDATE featured in RSTMH newsletter

18 Jun 2018, VALIDATE Director Helen McShane spoke to www.openaccessgovernment.org about "The current status of tuberculosis vaccine development" https://www.openaccessgovernment.org/current-status-tuberculosis-vaccine-development/46726/

30 Apr 2018, VALIDATE blog post for World Immunization week https://www.validate-network.org/article/vaccineswork-world-immunization-week-vaccines-validate

5 Dec 2017, VALIDATE featured in a blog post "Building international partnerships to tackle disease" on the University of Oxford Science blog, which has an extensive worldwide reach. http://www.ox.ac.uk/news/science-blog/building-international-partnerships-tackle-disease

22 Nov 2017, VALIDATE featured in article on GCRF Networks by Martin Broadstock for BSI Immunology News magazine https://www.immunology.org/news/the-network-effect-promoting-collaborative-research?utm_content=buffer15fca&utm_medium=social&utm_source=twitter.com&utm_campaign=buffer

24 Oct 2017, VALIDATE launch featured on TBVI website 'news' https://www.tbvi.eu/new-international-vaccine-rd-network-validate/
Year(s) Of Engagement Activity 2017,2018,2019,2020,2021
 
Description VALIDATE in the spotlight - media 
Form Of Engagement Activity A broadcast e.g. TV/radio/film/podcast (other than news/press)
Part Of Official Scheme? No
Geographic Reach National
Primary Audience Media (as a channel to the public)
Results and Impact 24 Mar 2018, VALIDATE Director Prof Helen McShane gave an interview for World TB Day at BBCOxford Radio https://www.bbc.co.uk/programmes/p060zj9g 1:02:30 for the headline; 1:09:05 for the interview.

29 Apr 2020, VALIDATE Director Prof Helen McShane featured in The Guardian podcast 'What has the BCG vaccine got to do with COVID-19' www.theguardian.com/science/audio/2020/apr/30/covid-19-what-has-the-bcg-vaccine-got-to-do-with-it-podcast.

2020, VALIDATE Fellow, Dr Rachel Tanner was filmed in the lab for the YouTube 'Life in a Day' movie directed by Ridley Scott and Kevin McDonald which premiered at Sundance Film Festival (her submission was featured as part of the press release).

11 Dec 2020, VALIDATE Director Prof Helen McShane interviewed on Sky News re vaccine hesitancy studies (relating to COVID): https://twitter.com/SkyNews/status/1337320456211361794

In 2020 VALIDATE Fellow Dr Rachel Tanner took part in 'I'm a Scientist, Stay at Home!' to help school pupils stay connected with STEM during the lockdown. Rachel was invited as a panellist to talk about her work and experience as a female scientist for the 2020 Women of the Future Awards and the 2021 Oxford Women in Engineering, Science and Technology (OxWEST) Annual Symposium.
Year(s) Of Engagement Activity 2018,2020
 
Description VALIDATE leishmaniasis video 
Form Of Engagement Activity A broadcast e.g. TV/radio/film/podcast (other than news/press)
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Public/other audiences
Results and Impact 'A few things you might not know about leishmaniasis' video created and added to VALIDATE YouTube channel for World NTD Day 2022. 217 views to date.
Year(s) Of Engagement Activity 2022,2023
URL https://www.youtube.com/watch?v=DgchaulbtrY
 
Description VALIDATE website 
Form Of Engagement Activity Engagement focused website, blog or social media channel
Part Of Official Scheme? No
Geographic Reach International
Primary Audience Other audiences
Results and Impact On 28 July 2017 we launched the VALIDATE Network website www.validate-network.org. The website publicises our network, our members (including a searchable members' directory to facilitate new collaborations), our funding opportunities, events, our mentoring scheme, projects and grants we have funded, seminars and workshops provided, external funding and training/job opportunities relevant to our members, and a weekly round-up of new grant calls/jobs/training/events and news. According to Google Analytics, to date the website has been viewed by 149k users from 215 countries/territories, with around 4,000 users per month in 2023.

Our aim is to make our website a central 'go to' hub for all relevant information for researchers working in our field (from academia, industry, government and non-profits), and for it to act as a hub of reliable information about vaccine research and our focus diseases for the general public and other interested parties. In 2018 we had 3 LMIC ECR members feedback outputs from our website information - one saw the Newton Travel Award listed in our external grants list and applied for and was awarded a grant; a second ECR member (from Uganda) applied for the "Data Management for Early Career Researchers" workshop in Nairobi that we listed in our training opportunities and was given a place and has now attended this course; while a 3rd ECR member from Cameroon applied for the Oxford University summer internship 2019 programme, having seen it advertised on the VALIDATE website. A further UK PhD (medical) student, when asked about any impact on career/research commented 'Easy notification of funding and training opportunities via validate website'.

In 2020 we also added a VALIDATE Microsoft Teams 'Hub' where members can more easily contact each other via several channels (including an ECR channel for ECR peer networking and support, and a collaborators channel for people looking for collaborators), and can access shared documents (including seminar recordings, Annual Meeting presentation slides, shared standardised SOPs and protocols).
Year(s) Of Engagement Activity 2017,2018,2019,2020,2021,2022,2023
URL http://www.validate-network.org
 
Description VALIDATE work experience 
Form Of Engagement Activity Participation in an open day or visit at my research institution
Part Of Official Scheme? No
Geographic Reach Regional
Primary Audience Schools
Results and Impact In February 2019 a Six Form student from a school in Surrey attended two days' work experience/shadowing at the Jenner Institute, visiting the laboratories and the clinical trial clinics and talking to staff about their research and careers in science, after contacting VALIDATE with a request to visit. This hopefully helped Zack with choosing his degree and future career plans.
Year(s) Of Engagement Activity 2017,2019