Dr  Alex Spencer

Dr Alex Spencer

Immunology Lecturer

School of Biomedical Sciences and Pharmacy

Career Summary

Biography

Dr Spencer is an immunology researcher and lecturer in the School of Biomedical Sciences and Pharmacy, based at the Hunter Medical Research Institute. Her research is focused on understanding how the immune response is induced following vaccination or infection, to enabling tailoring vaccines approaches to each specific disease. 

Following a PhD at the University of Sydney in 2006, Dr Spencer moved to the Jenner Institute, University of Oxford, to apply her knowledge of T cells to the development of a liver-stage malaria vaccine. She was involved in the development of new vaccine vectors (ChAdOx1 and ChAdOx2), identification of new adjuvants, optimising antigen design and vaccination regimens, working across several vaccine programs (malaria, Influenza, Ebola, SARS CoV2). 

The translational focus of the Jenner Institute enabled Dr Spencer to follow some of these approaches from preclinical studies to clinical trials, working with both academic collaborators and industry partners. As part of the Oxford COVID vaccine team, Dr Spencer performed the preclinical assessment of ChAdOx1 nCoV-19, measuring immunogenicity across a range of animal species, in combination with mRNA vaccines and rapidly testing each new SARS CoV-2 variant vaccine.

Dr Spencer is passionate about supporting the development of the next generation of scientist. In addition to supervising research student, she enjoys opportunities to travel to partner sites to train staff or host international students and researchers. Through her roles on local and national committees and editorial boards, she promotes inclusivity, diversity and equity in science.

Research Interests:

Dr Spencer studies the type of T and B cell responses induced following vaccination in terms of function (cytokines and antibody isotypes/subclasses), phenotype (effector/memory) and organ localisation (tissue resident T and B cells). This work involves comparing vaccine platforms and vaccination regimens to understand the role antigen presentation and inflammatory signals can play in the induction and maintenance of immune responses. 

Current projects include:

  • Understanding the role of antigen presentation in vaccine induced immunity.
  • Develop and optimise vaccines and vaccination regimens to target immune responses to specific sites.
  • Assessing the functional capacity of antibodies induced by vaccination.

Skills and Expertise:

  • Preclinical animal models
  • Cellular assays
  • Humoral assays
  • Ex vivo immunology assays
  • Multi-parameter flow cytometry
  • Immunohistochemistry
  • Imaging


Qualifications

  • Doctor of Philosophy, University of Sydney
  • Bachelor of Medical Science, University of Sydney

Keywords

  • T cells
  • adaptive immunity
  • clinical trials
  • infectious diseases
  • preclinical studies
  • vaccines

Fields of Research

Code Description Percentage
320404 Cellular immunology 40
320405 Humoural immunology and immunochemistry 35
320499 Immunology not elsewhere classified 25

Professional Experience

UON Appointment

Title Organisation / Department
Immunology Lecturer University of Newcastle
School of Biomedical Sciences and Pharmacy
Australia

Academic appointment

Dates Title Organisation / Department
1/8/2022 - 1/8/2023 Honorary Visiting Research Fellow in Immunology University of Oxford
Nuffield Department of Clinical Medicine
United Kingdom
1/10/2011 - 12/8/2022 Senior Immunologist University of Oxford
Nuffield Department of Clinical Medicine
United Kingdom
10/5/2006 - 30/9/2011 Preclinical Immunologist University of Oxford
Nuffield Department of Clinical Medicine
United Kingdom

Professional appointment

Dates Title Organisation / Department
1/6/2022 -  Associate Editor Frontiers in Immunology
Switzerland

Invitations

Keynote Speaker

Year Title / Rationale
2023 ASI NSW-ACT Branch Meeting 2023
2023 ASMR Hunter Region Annual Meeting
2021 LASA Virtual Annual Conference 2021
2021 Cambridge 21st Immunology Symposium

Speaker

Year Title / Rationale
2022 Developing viral vectored vaccines for infectious diseases
2021 Parasitology Module
2021 Masters in Parasitology
2021 European Masters in Vaccinology “LIVE”
2020 Second Edition of MIE-2020: Malaria Immunology & Elimination - A Virtual Short Symposium
Edit

Publications

For publications that are currently unpublished or in-press, details are shown in italics.

Highlighted Publications

Year Citation Altmetrics Link
2020 van Doremalen N, Lambe T, Spencer A, Belij-Rammerstorfer S, Purushotham JN, Port JR, et al., 'ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques', Nature, 586 578-582 (2020) [C1]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 20191,2 and is responsible for the coronavirus disease 2019 (COVID-19) pandemic3. Vaccines are an ... [more]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 20191,2 and is responsible for the coronavirus disease 2019 (COVID-19) pandemic3. Vaccines are an essential countermeasure and are urgently needed to control the pandemic4. Here we show that the adenovirus-vector-based vaccine ChAdOx1¿nCoV-19, which encodes the spike protein of SARS-CoV-2, is immunogenic in mice and elicites a robust humoral and cell-mediated response. This response was predominantly mediated by type-1 T helper cells, as demonstrated by the profiling of the IgG subclass and the expression of cytokines. Vaccination with ChAdOx1¿nCoV-19 (using either a prime-only or a prime¿boost regimen) induced a balanced humoral and cellular immune response of type-1 and type-2 T helper cells in rhesus macaques. We observed a significantly reduced viral load in the bronchoalveolar lavage fluid and lower respiratory tract tissue of vaccinated rhesus macaques that were challenged with SARS-CoV-2 compared with control animals, and no pneumonia was observed in vaccinated SARS-CoV-2-infected animals. However, there was no difference in nasal shedding between vaccinated and control SARS-CoV-2-infected macaques. Notably, we found no evidence of immune-enhanced disease after viral challenge in vaccinated SARS-CoV-2-infected animals. The safety, immunogenicity and efficacy profiles of ChAdOx1¿nCoV-19 against symptomatic PCR-positive COVID-19 disease will now be assessed in randomized controlled clinical trials in humans.

DOI 10.1038/s41586-020-2608-y
Citations Scopus - 633Web of Science - 555
2020 Graham SP, McLean RK, Spencer AJ, Belij-Rammerstorfer S, Wright D, Ulaszewska M, et al., 'Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19', npj Vaccines, 5 (2020) [C1]

Clinical development of the COVID-19 vaccine candidate ChAdOx1 nCoV-19, a replication-deficient simian adenoviral vector expressing the full-length SARS-CoV-2 spike (S) protein wa... [more]

Clinical development of the COVID-19 vaccine candidate ChAdOx1 nCoV-19, a replication-deficient simian adenoviral vector expressing the full-length SARS-CoV-2 spike (S) protein was initiated in April 2020 following non-human primate studies using a single immunisation. Here, we compared the immunogenicity of one or two doses of ChAdOx1 nCoV-19 in both mice and pigs. Whilst a single dose induced antigen-specific antibody and T cells responses, a booster immunisation enhanced antibody responses, particularly in pigs, with a significant increase in SARS-CoV-2 neutralising titres.

DOI 10.1038/s41541-020-00221-3
Citations Scopus - 96Web of Science - 81
2021 Provine NM, Amini A, Garner LC, Spencer AJ, Dold C, Hutchings C, et al., 'MAIT cell activation augments adenovirus vector vaccine immunogenicity', Science, 371 (2021) [C1]

Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized that MAIT cells may ... [more]

Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized that MAIT cells may have inherent adjuvant activity in vaccine platforms that use replication-incompetent adenovirus vectors. In mice and humans, ChAdOx1 (chimpanzee adenovirus Ox1) immunization robustly activated MAIT cells. Activation required plasmacytoid dendritic cell (pDC)-derived interferon (IFN)-a and monocyte-derived interleukin-18. IFN-a-induced, monocyte-derived tumor necrosis factor was also identified as a key secondary signal. All three cytokines were required in vitro and in vivo. Activation of MAIT cells positively correlated with vaccine-induced T cell responses in human volunteers and MAIT cell-deficient mice displayed impaired CD8+ T cell responses to multiple vaccine-encoded antigens. Thus, MAIT cells contribute to the immunogenicity of adenovirus vectors, with implications for vaccine design.

DOI 10.1126/science.aax8819
Citations Scopus - 79Web of Science - 53
2021 Lambe T, Spencer AJ, Thomas KM, Gooch KE, Thomas S, White AD, et al., 'ChAdOx1 nCoV-19 protection against SARS-CoV-2 in rhesus macaque and ferret challenge models', Communications Biology, 4 (2021) [C1]

Vaccines against SARS-CoV-2 are urgently required, but early development of vaccines against SARS-CoV-1 resulted in enhanced disease after vaccination. Careful assessment of this ... [more]

Vaccines against SARS-CoV-2 are urgently required, but early development of vaccines against SARS-CoV-1 resulted in enhanced disease after vaccination. Careful assessment of this phenomena is warranted for vaccine development against SARS CoV-2. Here we report detailed immune profiling after ChAdOx1 nCoV-19 (AZD1222) and subsequent high dose challenge in two animal models of SARS-CoV-2 mediated disease. We demonstrate in rhesus macaques the lung pathology caused by SARS-CoV-2 mediated pneumonia is reduced by prior vaccination with ChAdOx1 nCoV-19 which induced neutralising antibody responses after a single intramuscular administration. In a second animal model, ferrets, ChAdOx1 nCoV-19 reduced both virus shedding and lung pathology. Antibody titre were boosted by a second dose. Data from these challenge models on the absence of enhanced disease and the detailed immune profiling, support the continued clinical evaluation of ChAdOx1 nCoV-19.

DOI 10.1038/s42003-021-02443-0
Citations Scopus - 13Web of Science - 12
2021 Spencer AJ, McKay PF, Belij-Rammerstorfer S, Ulaszewska M, Bissett CD, Hu K, et al., 'Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice', Nature Communications, 12 (2021) [C1]

Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens usin... [more]

Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens using alternate vaccine modalities. Here, we present a detailed description of the immune response, in mice, following vaccination with a self-amplifying RNA (saRNA) vaccine and an adenoviral vectored vaccine (ChAdOx1 nCoV-19/AZD1222) against SARS-CoV-2. We demonstrate that antibody responses are higher in two-dose heterologous vaccination regimens than single-dose regimens. Neutralising titres after heterologous prime-boost were at least comparable or higher than the titres measured after homologous prime boost vaccination with viral vectors. Importantly, the cellular immune response after a heterologous regimen is dominated by cytotoxic T cells and Th1+ CD4 T cells, which is superior to the response induced in homologous vaccination regimens in mice. These results underpin the need for clinical trials to investigate the immunogenicity of heterologous regimens with alternate vaccine technologies.

DOI 10.1038/s41467-021-23173-1
Citations Scopus - 86Web of Science - 73
2021 Collins KA, Brod F, Snaith R, Ulaszewska M, Longley RJ, Salman AM, et al., 'Ultra-low dose immunization and multi-component vaccination strategies enhance protection against malaria in mice', Scientific Reports, 11 (2021) [C1]

An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protectio... [more]

An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protection in a Phase 3 trial. R21 is a next-generation RTS,S-like vaccine. We have previously shown in mice that R21 administered in Matrix-M is highly immunogenic, able to elicit complete protection against sporozoite challenge, and can be successfully administered with TRAP based viral-vectors resulting in enhanced protection. In this study, we developed a novel, GMP-compatible purification process for R21, and evaluated the immunogenicity and protective efficacy of ultra-low doses of both R21 and RTS,S when formulated in AS01. We demonstrated that both vaccines are highly immunogenic and also elicit comparable high levels of protection against transgenic parasites in BALB/c mice. By lowering the vaccine dose there was a trend for increased immunogenicity and sterile protection, with the highest dose vaccine groups achieving the lowest efficacy (50% sterile protection). We also evaluated the ability to combine RTS,S/AS01 with TRAP based viral-vectors and observed concurrent induction of immune responses to both antigens with minimal interference when mixing the vaccines prior to administration. These studies suggest that R21 or RTS,S could be combined with viral-vectors for a multi-component vaccination approach and indicate that low dose vaccination should be fully explored in humans to maximize potential efficacy.

DOI 10.1038/s41598-021-90290-8
Citations Scopus - 8Web of Science - 1
2022 Noé A, Datoo MS, Flaxman A, Husainy MA, Jenkin D, Bellamy D, et al., 'Deep Immune Phenotyping and Single-Cell Transcriptomics Allow Identification of Circulating TRM-Like Cells Which Correlate With Liver-Stage Immunity and Vaccine-Induced Protection From Malaria', Frontiers in Immunology, 13 (2022) [C1]

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, inv... [more]

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral ¿TRM-like¿ cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.

DOI 10.3389/fimmu.2022.795463
Citations Scopus - 5Web of Science - 3
2022 Spencer AJ, Morris S, Ulaszewska M, Powers C, Kailath R, Bissett C, et al., 'The ChAdOx1 vectored vaccine, AZD2816, induces strong immunogenicity against SARS-CoV-2 beta (B.1.351) and other variants of concern in preclinical studies', eBioMedicine, 77 (2022) [C1]

Background: There is an ongoing global effort to design, manufacture, and clinically assess vaccines against SARS-CoV-2. Over the course of the ongoing pandemic a number of new SA... [more]

Background: There is an ongoing global effort to design, manufacture, and clinically assess vaccines against SARS-CoV-2. Over the course of the ongoing pandemic a number of new SARS-CoV-2 virus isolates or variants of concern (VoC) have been identified containing mutations in key proteins. Methods: In this study we describe the generation and preclinical assessment of a ChAdOx1-vectored vaccine (AZD2816) which expresses the spike protein of the Beta VoC (B.1.351). Findings: We demonstrate that AZD2816 is immunogenic after a single dose. When AZD2816 is used as a booster dose in animals primed with a vaccine encoding the original spike protein (ChAdOx1 nCoV-19/ [AZD1222]), an increase in binding and neutralising antibodies against Beta (B.1.351), Gamma (P.1) and Delta (B.1.617.2) is observed following each additional dose. In addition, a strong and polyfunctional T cell response was measured all booster regimens. Interpretation: Real world data is demonstrating that one or more doses of licensed SARS-CoV-2 vaccines confer reduced protection against hospitalisation and deaths caused by divergent VoC, including Omicron. Our data support the ongoing clinical development and testing of booster vaccines to increase immunity against highly mutated VoC. Funding: This research was funded by AstraZeneca with supporting funds from MRC and BBSRC.

DOI 10.1016/j.ebiom.2022.103902
Citations Scopus - 11Web of Science - 5

Chapter (1 outputs)

Year Citation Altmetrics Link
2011 Cottingham MG, Andersen RF, Spencer AJ, Saurya S, Furze J, Hill AVS, Gilbert SC, 'Recombination-mediated genetic engineering of a bacterial artificial chromosome clone of modified vaccinia virus ankara (MVA)', Genetic Engineering: Recent Developments in Applications 49-70 (2011)

The production, manipulation and rescue of a bacterial artificial chromosome clone of Vaccinia virus (VAC-BAC) in order to expedite construction of expression vectors and mutagene... [more]

The production, manipulation and rescue of a bacterial artificial chromosome clone of Vaccinia virus (VAC-BAC) in order to expedite construction of expression vectors and mutagenesis of the genome has been described (Domi & Moss, 2002, PNAS 99 12415-20). The genomic BAC clone was 'rescued' back to infectious virus using a Fowlpox virus helper to supply transcriptional machinery. We apply here a similar approach to the attenuated strain Modified Vaccinia virus Ankara (MVA), now widely used as a safe non-replicating recombinant vaccine vector in mammals, including humans. Four apparently full-length, rescuable clones were obtained, which had indistinguishable immunogenicity in mice. One clone was shotgun sequenced and found to be identical to the parent. We employed GalK recombination-mediated genetic engineering (recombineering) of MVA-BAC to delete five selected viral genes. Deletion of C12L, A44L, A46R or B7R did not significantly affect CD8+ T cell immunogenicity in BALB/c mice, but deletion of B15R enhanced specific CD8+ T cell responses to one of two endogenous viral epitopes (from the E2 and F2 proteins), in accordance with published work (Staib et al., 2005, J. Gen. Virol. 86, 1997-2006). In addition, we found a higher frequency of triple-positive IFN-¿, TNF-a and IL-2 secreting E3-specific CD8+ T-cells 8 weeks after vaccination with MVA lacking B15R. Furthermore, a recombinant vaccine capable of inducing CD8+ T cells against an epitope from Plasmodium berghei was created using GalK counterselection to insert an antigen expression cassette lacking a tandem marker gene into the traditional thymidine kinase locus of MVA-BAC. MVA continues to feature prominently in clinical trials of recombinant vaccines against diseases such as HIV-AIDS, malaria and tuberculosis. Here we demonstrate in proof-of-concept experiments that MVA-BAC recombineering is a viable route to more rapid and efficient generation of new candidate mutant and recombinant vaccines based on a clinically deployable viral vector.


Journal article (76 outputs)

Year Citation Altmetrics Link
2024 Lv X, Martin J, Hoover H, Joshi B, Wilkens M, Ullisch DA, et al., 'Chemical and biological characterization of vaccine adjuvant QS-21 produced via plant cell culture', iScience, 27 (2024) [C1]

Many vaccines, including those using recombinant antigen subunits, rely on adjuvant(s) to enhance the efficacy of the host immune responses. Among the few adjuvants clinically app... [more]

Many vaccines, including those using recombinant antigen subunits, rely on adjuvant(s) to enhance the efficacy of the host immune responses. Among the few adjuvants clinically approved, QS-21, a saponin-based immunomodulatory molecule isolated from the tree bark of Quillaja saponaria (QS) is used in complex formulations in approved effective vaccines. High demand of the QS raw material as well as manufacturing scalability limitation has been barriers here. We report for the first-time successful plant cell culture production of QS-21 having structural, chemical, and biologic, properties similar to the bark extracted product. These data ensure QS-21 and related saponins are broadly available and accessible to drug developers.

DOI 10.1016/j.isci.2024.109006
2023 Saunders JE, Gilbride C, Dowall S, Morris S, Ulaszewska M, Spencer AJ, et al., 'Adenoviral vectored vaccination protects against Crimean- Congo Haemorrhagic Fever disease in a lethal challenge model', EBIOMEDICINE, 90 (2023) [C1]
DOI 10.1016/j.ebiom.2023.104523
Citations Web of Science - 5
2023 Saunders JE, Gilbride C, Dowall S, Morris S, Ulaszewska M, Spencer AJ, et al., 'Adenoviral vectored vaccination protects against Crimean-Congo Haemorrhagic Fever disease in a lethal challenge model', eBioMedicine, 90 104523-104523 (2023) [C1]
DOI 10.1016/j.ebiom.2023.104523
Citations Scopus - 7
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse"', MBIO, 14 (2023)
DOI 10.1128/mbio.00815-23
Co-authors Nathan Bartlett, Camille Esneau
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse"', MSPHERE, 8 (2023)
DOI 10.1128/msphere.00165-23
Co-authors Nathan Bartlett, Camille Esneau
2023 Speck P, Mackenzie J, Bull RA, Slobedman B, Drummer H, Fraser J, et al., 'Statement in Support of: "Virology under the Microscope-a Call for Rational Discourse".', J Virol, 97 e0045123 (2023)
DOI 10.1128/jvi.00451-23
Co-authors Camille Esneau, Nathan Bartlett
2023 Foster WS, Newman J, Thakur N, Spencer AJ, Davies S, Woods D, et al., 'ChAdOx1 nCoV-19 vaccination generates spike-specific CD8
DOI 10.1111/imcb.12645
2023 Ni T, Mendonça L, Zhu Y, Howe A, Radecke J, Shah PM, et al., 'ChAdOx1 COVID vaccines express RBD open prefusion SARS-CoV-2 spikes on the cell surface', iScience, 26 (2023) [C1]

Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been proven to be an effective means of decreasing COVID-19 mortality, hospitalization rates, an... [more]

Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been proven to be an effective means of decreasing COVID-19 mortality, hospitalization rates, and transmission. One of the vaccines deployed worldwide is ChAdOx1 nCoV-19, which uses an adenovirus vector to drive the expression of the original SARS-CoV-2 spike on the surface of transduced cells. Using cryo-electron tomography and subtomogram averaging, we determined the native structures of the vaccine product expressed on cell surfaces in situ. We show that ChAdOx1-vectored vaccines expressing the Beta SARS-CoV-2 variant produce abundant native prefusion spikes predominantly in one-RBD-up conformation. Furthermore, the ChAdOx1-vectored HexaPro-stabilized spike yields higher cell surface expression, enhanced RBD exposure, and reduced shedding of S1 compared to the wild type. We demonstrate in situ structure determination as a powerful means for studying antigen design options in future vaccine development against emerging novel SARS-CoV-2 variants and broadly against other infectious viruses.

DOI 10.1016/j.isci.2023.107882
Citations Scopus - 2
2022 Foster WS, Lee JL, Thakur N, Newman J, Spencer AJ, Davies S, et al., 'Tfh cells and the germinal center are required for memory B cell formation & humoral immunity after ChAdOx1 nCoV-19 vaccination', Cell Reports Medicine, 3 (2022) [C1]

Emergence from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been facilitated by the rollout of effective vaccines. Successful vaccines generate hi... [more]

Emergence from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been facilitated by the rollout of effective vaccines. Successful vaccines generate high-affinity plasma blasts and long-lived protective memory B cells. Here, we show a requirement for T follicular helper (Tfh) cells and the germinal center reaction for optimal serum antibody and memory B cell formation after ChAdOx1 nCoV-19 vaccination. We found that Tfh cells play an important role in expanding antigen-specific B cells while identifying Tfh-cell-dependent and -independent memory B cell subsets. Upon secondary vaccination, germinal center B cells generated during primary immunizations can be recalled as germinal center B cells again. Likewise, primary immunization GC-Tfh cells can be recalled as either Tfh or Th1 cells, highlighting the pluripotent nature of Tfh cell memory. This study demonstrates that ChAdOx1 nCoV-19-induced germinal centers are a critical source of humoral immunity.

DOI 10.1016/j.xcrm.2022.100845
Citations Scopus - 9Web of Science - 2
2022 Noé A, Datoo MS, Flaxman A, Husainy MA, Jenkin D, Bellamy D, et al., 'Deep Immune Phenotyping and Single-Cell Transcriptomics Allow Identification of Circulating TRM-Like Cells Which Correlate With Liver-Stage Immunity and Vaccine-Induced Protection From Malaria', Frontiers in Immunology, 13 (2022) [C1]

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, inv... [more]

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral ¿TRM-like¿ cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.

DOI 10.3389/fimmu.2022.795463
Citations Scopus - 5Web of Science - 3
2022 van Doremalen N, Schulz JE, Adney DR, Saturday TA, Fischer RJ, Yinda CK, et al., 'ChAdOx1 nCoV-19 (AZD1222) or nCoV-19-Beta (AZD2816) protect Syrian hamsters against Beta Delta and Omicron variants', Nature Communications, 13 (2022) [C1]

ChAdOx1 nCoV-19 (AZD1222) is a replication-deficient simian adenovirus¿vectored vaccine encoding the spike (S) protein of SARS-CoV-2, based on the first published full-length sequ... [more]

ChAdOx1 nCoV-19 (AZD1222) is a replication-deficient simian adenovirus¿vectored vaccine encoding the spike (S) protein of SARS-CoV-2, based on the first published full-length sequence (Wuhan-1). AZD1222 has been shown to have 74% vaccine efficacy against symptomatic disease in clinical trials. However, variants of concern (VoCs) have been detected, with substitutions that are associated with a reduction in virus neutralizing antibody titer. Updating vaccines to include S proteins of VoCs may be beneficial, even though current real-world data is suggesting good efficacy following boosting with vaccines encoding the ancestral S protein. Using the Syrian hamster model, we evaluate the effect of a single dose of AZD2816, encoding the S protein of the Beta VoC, and efficacy of AZD1222/AZD2816 as a heterologous primary series against challenge with the Beta or Delta variant. Minimal to no viral sgRNA could be detected in lungs of vaccinated animals obtained at 3- or 5- days post inoculation, in contrast to lungs of control animals. In Omicron-challenged hamsters, a single dose of AZD2816 or AZD1222 reduced virus shedding. Thus, these vaccination regimens are protective against the Beta, Delta, and Omicron VoCs in the hamster model.

DOI 10.1038/s41467-022-32248-6
Citations Scopus - 16Web of Science - 1
2022 Spencer AJ, Morris S, Ulaszewska M, Powers C, Kailath R, Bissett C, et al., 'The ChAdOx1 vectored vaccine, AZD2816, induces strong immunogenicity against SARS-CoV-2 beta (B.1.351) and other variants of concern in preclinical studies', eBioMedicine, 77 (2022) [C1]

Background: There is an ongoing global effort to design, manufacture, and clinically assess vaccines against SARS-CoV-2. Over the course of the ongoing pandemic a number of new SA... [more]

Background: There is an ongoing global effort to design, manufacture, and clinically assess vaccines against SARS-CoV-2. Over the course of the ongoing pandemic a number of new SARS-CoV-2 virus isolates or variants of concern (VoC) have been identified containing mutations in key proteins. Methods: In this study we describe the generation and preclinical assessment of a ChAdOx1-vectored vaccine (AZD2816) which expresses the spike protein of the Beta VoC (B.1.351). Findings: We demonstrate that AZD2816 is immunogenic after a single dose. When AZD2816 is used as a booster dose in animals primed with a vaccine encoding the original spike protein (ChAdOx1 nCoV-19/ [AZD1222]), an increase in binding and neutralising antibodies against Beta (B.1.351), Gamma (P.1) and Delta (B.1.617.2) is observed following each additional dose. In addition, a strong and polyfunctional T cell response was measured all booster regimens. Interpretation: Real world data is demonstrating that one or more doses of licensed SARS-CoV-2 vaccines confer reduced protection against hospitalisation and deaths caused by divergent VoC, including Omicron. Our data support the ongoing clinical development and testing of booster vaccines to increase immunity against highly mutated VoC. Funding: This research was funded by AstraZeneca with supporting funds from MRC and BBSRC.

DOI 10.1016/j.ebiom.2022.103902
Citations Scopus - 11Web of Science - 5
2022 Rosadas C, Khan M, Parker E, Marchesin F, Katsanovskaja K, Sureda-Vives M, et al., 'Detection and quantification of antibody to SARS CoV 2 receptor binding domain provides enhanced sensitivity, specificity and utility', JOURNAL OF VIROLOGICAL METHODS, 302 (2022)
DOI 10.1016/j.jviromet.2022.114475
Citations Scopus - 8Web of Science - 6
2022 Padron-Regalado E, Ulaszewska M, Douglas AD, Hill AVS, Spencer AJ, 'STING-pathway modulation to enhance the immunogenicity of adenoviral-vectored vaccines', Scientific Reports, 12 (2022) [C1]

Traditional chemical adjuvants remain a practical means of enhancing the immunogenicity of vaccines. Nevertheless, it is recognized that increasing the immunogenicity of viral vec... [more]

Traditional chemical adjuvants remain a practical means of enhancing the immunogenicity of vaccines. Nevertheless, it is recognized that increasing the immunogenicity of viral vectors is challenging. Recently, STING ligands have been shown to enhance the efficacy of different vaccine platforms, but their affectivity on viral-vectored vaccination has not been fully assessed. In this study we used a multi-pronged approach to shed light on the immunological properties and potential mechanisms of action of this type of adjuvant and focused our study on replication-deficient human adenovirus serotype 5 (AdHu5). When the STING ligand 2'3'-cGAMP was mixed with AdHu5, the adjuvant enhanced anti-vector immune responses while decreasing the transgene-specific CD8+ T cell response. Studies employing STING-knockout mice and a 2'3'-cGAMP inactive analogue confirmed the aforementioned effects were STING dependent. In vitro assays demonstrated 2'3'-cGAMP induced the production of IFN-ß which in turn negatively affected AdHu5 transgene expression and CD8+ T cell immunogenicity. In an effort to overcome the negative impact of early 2'3'-cGAMP signaling on AdHu5 transgene immunogenicity, we generated a bicistronic vector encoding the 2'3'-cGAMP together with a model antigen. Intracellular production of 2'3'-cGAMP after AdHu5 infection was able to enhance transgene-specific CD8+ T cell immunogenicity, although not to a level that would warrant progression of this adjuvant to clinical assessment. This work highlights the importance of timing of 2'3'-cGAMP administration when assessing its adjuvant capacity with different vaccine modalities.

DOI 10.1038/s41598-022-18750-3
Citations Scopus - 2
2021 Voysey M, Clemens SAC, Madhi SA, Weckx LY, Folegatti PM, Aley PK, et al., 'Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK', The Lancet, 397 99-111 (2021) [C1]

Background: A safe and efficacious vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), if deployed with high coverage, could contribute to the control of... [more]

Background: A safe and efficacious vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), if deployed with high coverage, could contribute to the control of the COVID-19 pandemic. We evaluated the safety and efficacy of the ChAdOx1 nCoV-19 vaccine in a pooled interim analysis of four trials. Methods: This analysis includes data from four ongoing blinded, randomised, controlled trials done across the UK, Brazil, and South Africa. Participants aged 18 years and older were randomly assigned (1:1) to ChAdOx1 nCoV-19 vaccine or control (meningococcal group A, C, W, and Y conjugate vaccine or saline). Participants in the ChAdOx1 nCoV-19 group received two doses containing 5 × 1010 viral particles (standard dose; SD/SD cohort); a subset in the UK trial received a half dose as their first dose (low dose) and a standard dose as their second dose (LD/SD cohort). The primary efficacy analysis included symptomatic COVID-19 in seronegative participants with a nucleic acid amplification test-positive swab more than 14 days after a second dose of vaccine. Participants were analysed according to treatment received, with data cutoff on Nov 4, 2020. Vaccine efficacy was calculated as 1 - relative risk derived from a robust Poisson regression model adjusted for age. Studies are registered at ISRCTN89951424 and ClinicalTrials.gov, NCT04324606, NCT04400838, and NCT04444674. Findings: Between April 23 and Nov 4, 2020, 23 848 participants were enrolled and 11 636 participants (7548 in the UK, 4088 in Brazil) were included in the interim primary efficacy analysis. In participants who received two standard doses, vaccine efficacy was 62·1% (95% CI 41·0¿75·7; 27 [0·6%] of 4440 in the ChAdOx1 nCoV-19 group vs71 [1·6%] of 4455 in the control group) and in participants who received a low dose followed by a standard dose, efficacy was 90·0% (67·4¿97·0; three [0·2%] of 1367 vs 30 [2·2%] of 1374; pinteraction=0·010). Overall vaccine efficacy across both groups was 70·4% (95·8% CI 54·8¿80·6; 30 [0·5%] of 5807 vs 101 [1·7%] of 5829). From 21 days after the first dose, there were ten cases hospitalised for COVID-19, all in the control arm; two were classified as severe COVID-19, including one death. There were 74 341 person-months of safety follow-up (median 3·4 months, IQR 1·3¿4·8): 175 severe adverse events occurred in 168 participants, 84 events in the ChAdOx1 nCoV-19 group and 91 in the control group. Three events were classified as possibly related to a vaccine: one in the ChAdOx1 nCoV-19 group, one in the control group, and one in a participant who remains masked to group allocation. Interpretation: ChAdOx1 nCoV-19 has an acceptable safety profile and has been found to be efficacious against symptomatic COVID-19 in this interim analysis of ongoing clinical trials. Funding: UK Research and Innovation, National Institutes for Health Research (NIHR), Coalition for Epidemic Preparedness Innovations, Bill & Melinda Gates Foundation, Lemann Foundation, Rede D'Or, Brava and Telles Foundation, NIHR Oxford Biomedical Research Centre, Thames Valley and South Midland's NIHR Clinical Research Network, and AstraZeneca.

DOI 10.1016/S0140-6736(20)32661-1
Citations Scopus - 3281Web of Science - 2540
2021 van Doremalen N, Lambe T, Spencer A, Belij-Rammerstorfer S, Purushotham JN, Port JR, et al., 'ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques (vol 586, pg 578, 2020)', NATURE, 590 E24-E24 (2021)
DOI 10.1038/s41586-020-03099-2
Citations Scopus - 4Web of Science - 5
2021 Barrett JR, Belij-Rammerstorfer S, Dold C, Ewer KJ, Folegatti PM, Gilbride C, et al., 'Phase 1/2 trial of SARS-CoV-2 vaccine ChAdOx1 nCoV-19 with a booster dose induces multifunctional antibody responses (vol 27, pg 279, 2021)', NATURE MEDICINE, 27 1113-1113 (2021)
DOI 10.1038/s41591-021-01372-z
Citations Scopus - 8Web of Science - 4
2021 Ewer KJ, Barrett JR, Belij-Rammerstorfer S, Sharpe H, Makinson R, Morter R, et al., 'T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial (vol 27, pg 270, 2021)', NATURE MEDICINE, 27 1116-1116 (2021)
DOI 10.1038/s41591-021-01363-0
Citations Scopus - 7Web of Science - 3
2021 Provine NM, Amini A, Garner LC, Spencer AJ, Dold C, Hutchings C, et al., 'MAIT cell activation augments adenovirus vector vaccine immunogenicity', Science, 371 (2021) [C1]

Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized that MAIT cells may ... [more]

Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized that MAIT cells may have inherent adjuvant activity in vaccine platforms that use replication-incompetent adenovirus vectors. In mice and humans, ChAdOx1 (chimpanzee adenovirus Ox1) immunization robustly activated MAIT cells. Activation required plasmacytoid dendritic cell (pDC)-derived interferon (IFN)-a and monocyte-derived interleukin-18. IFN-a-induced, monocyte-derived tumor necrosis factor was also identified as a key secondary signal. All three cytokines were required in vitro and in vivo. Activation of MAIT cells positively correlated with vaccine-induced T cell responses in human volunteers and MAIT cell-deficient mice displayed impaired CD8+ T cell responses to multiple vaccine-encoded antigens. Thus, MAIT cells contribute to the immunogenicity of adenovirus vectors, with implications for vaccine design.

DOI 10.1126/science.aax8819
Citations Scopus - 79Web of Science - 53
2021 Voysey M, Costa Clemens SA, Madhi SA, Weckx LY, Folegatti PM, Aley PK, et al., 'Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: a pooled analysis of four randomised trials', The Lancet, 397 881-891 (2021) [C1]

Background: The ChAdOx1 nCoV-19 (AZD1222) vaccine has been approved for emergency use by the UK regulatory authority, Medicines and Healthcare products Regulatory Agency, with a r... [more]

Background: The ChAdOx1 nCoV-19 (AZD1222) vaccine has been approved for emergency use by the UK regulatory authority, Medicines and Healthcare products Regulatory Agency, with a regimen of two standard doses given with an interval of 4¿12 weeks. The planned roll-out in the UK will involve vaccinating people in high-risk categories with their first dose immediately, and delivering the second dose 12 weeks later. Here, we provide both a further prespecified pooled analysis of trials of ChAdOx1 nCoV-19 and exploratory analyses of the impact on immunogenicity and efficacy of extending the interval between priming and booster doses. In addition, we show the immunogenicity and protection afforded by the first dose, before a booster dose has been offered. Methods: We present data from three single-blind randomised controlled trials¿one phase 1/2 study in the UK (COV001), one phase 2/3 study in the UK (COV002), and a phase 3 study in Brazil (COV003)¿and one double-blind phase 1/2 study in South Africa (COV005). As previously described, individuals 18 years and older were randomly assigned 1:1 to receive two standard doses of ChAdOx1 nCoV-19 (5 × 1010 viral particles) or a control vaccine or saline placebo. In the UK trial, a subset of participants received a lower dose (2·2 × 1010 viral particles) of the ChAdOx1 nCoV-19 for the first dose. The primary outcome was virologically confirmed symptomatic COVID-19 disease, defined as a nucleic acid amplification test (NAAT)-positive swab combined with at least one qualifying symptom (fever =37·8°C, cough, shortness of breath, or anosmia or ageusia) more than 14 days after the second dose. Secondary efficacy analyses included cases occuring at least 22 days after the first dose. Antibody responses measured by immunoassay and by pseudovirus neutralisation were exploratory outcomes. All cases of COVID-19 with a NAAT-positive swab were adjudicated for inclusion in the analysis by a masked independent endpoint review committee. The primary analysis included all participants who were SARS-CoV-2 N protein seronegative at baseline, had had at least 14 days of follow-up after the second dose, and had no evidence of previous SARS-CoV-2 infection from NAAT swabs. Safety was assessed in all participants who received at least one dose. The four trials are registered at ISRCTN89951424 (COV003) and ClinicalTrials.gov, NCT04324606 (COV001), NCT04400838 (COV002), and NCT04444674 (COV005). Findings: Between April 23 and Dec 6, 2020, 24 422 participants were recruited and vaccinated across the four studies, of whom 17 178 were included in the primary analysis (8597 receiving ChAdOx1 nCoV-19 and 8581 receiving control vaccine). The data cutoff for these analyses was Dec 7, 2020. 332 NAAT-positive infections met the primary endpoint of symptomatic infection more than 14 days after the second dose. Overall vaccine efficacy more than 14 days after the second dose was 66·7% (95% CI 57·4¿74·0), with 84 (1·0%) cases in the 8597 participants in the ChAdOx1 nCoV-19 group and 248 (2·9%) in the 8581 participants in the control group. There were no hospital admissions for COVID-19 in the ChAdOx1 nCoV-19 group after the initial 21-day exclusion period, and 15 in the control group. 108 (0·9%) of 12 282 participants in the ChAdOx1 nCoV-19 group and 127 (1·1%) of 11 962 participants in the control group had serious adverse events. There were seven deaths considered unrelated to vaccination (two in the ChAdOx1 nCov-19 group and five in the control group), including one COVID-19-related death in one participant in the control group. Exploratory analyses showed that vaccine efficacy after a single standard dose of vaccine from day 22 to day 90 after vaccination was 76·0% (59·3¿85·9). Our modelling analysis indicated that protection did not wane during this initial 3-month period. Similarly, antibody levels were maintained during this period with minimal waning by day 90 (geometric mean ratio [GMR] 0·66 [95% CI 0·59¿0·74]). In...

DOI 10.1016/S0140-6736(21)00432-3
Citations Scopus - 796Web of Science - 705
2021 Ewer KJ, Barrett JR, Belij-Rammerstorfer S, Sharpe H, Makinson R, Morter R, et al., 'T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial', Nature Medicine, 27 270-278 (2021) [C1]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of Coronavirus Disease 2019 (COVID-19), has caused a global pandemic, and safe, effective vaccine... [more]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of Coronavirus Disease 2019 (COVID-19), has caused a global pandemic, and safe, effective vaccines are urgently needed1. Strong, Th1-skewed T cell responses can drive protective humoral and cell-mediated immune responses2 and might reduce the potential for disease enhancement3. Cytotoxic T cells clear virus-infected host cells and contribute to control of infection4. Studies of patients infected with SARS-CoV-2 have suggested a protective role for both humoral and cell-mediated immune responses in recovery from COVID-19 (refs. 5,6). ChAdOx1 nCoV-19 (AZD1222) is a candidate SARS-CoV-2 vaccine comprising a replication-deficient simian adenovirus expressing full-length SARS-CoV-2 spike protein. We recently reported preliminary safety and immunogenicity data from a phase 1/2 trial of the ChAdOx1 nCoV-19 vaccine (NCT04400838)7 given as either a one- or two-dose regimen. The vaccine was tolerated, with induction of neutralizing antibodies and antigen-specific T cells against the SARS-CoV-2 spike protein. Here we describe, in detail, exploratory analyses of the immune responses in adults, aged 18¿55 years, up to 8 weeks after vaccination with a single dose of ChAdOx1 nCoV-19 in this trial, demonstrating an induction of a Th1-biased response characterized by interferon-¿ and tumor necrosis factor-a cytokine secretion by CD4+ T cells and antibody production predominantly of IgG1 and IgG3 subclasses. CD8+ T cells, of monofunctional, polyfunctional and cytotoxic phenotypes, were also induced. Taken together, these results suggest a favorable immune profile induced by ChAdOx1 nCoV-19 vaccine, supporting the progression of this vaccine candidate to ongoing phase 2/3 trials to assess vaccine efficacy.

DOI 10.1038/s41591-020-01194-5
Citations Scopus - 407Web of Science - 333
2021 Ssemakalu CC, Ulaszewska M, Elias S, Spencer AJ, 'Solar inactivated Salmonella Typhimurium induces an immune response in BALB/c mice', Heliyon, 7 (2021) [C1]

Salmonella is contracted through the consumption of untreated water and contaminated food. The contraction and spread of water-related Salmonella in resource-poor communities can ... [more]

Salmonella is contracted through the consumption of untreated water and contaminated food. The contraction and spread of water-related Salmonella in resource-poor communities can be reduced by using solar disinfection (SODIS) to treat the water before its consumption. SODIS is a water sanitizing technique that relies on natural sunshine. It is a cost-effective, inexpensive, environmentally, and user-friendly means of treating microbiologically contaminated water. This water disinfection method has saved many lives in communities vulnerable to water-related infections worldwide. At present, the success of SODIS has mainly been attributed to permanent inactivation of water pathogens ability to grow. However, little to no information exists as to whether immune responses to the solar inactivated pathogens are induced in SODIS water consumers. This study assessed the potential for solar inactivated S. Typhimurium to induce an immune response in mice. Results show that solar inactivated S. Typhimurium can induce bactericidal antibodies against S. Typhimurium. Furthermore, an increase in the secretion of interferon-gamma (IFN-¿) was observed in mice given the solar inactivated S. Typhimurium. These findings suggest that solar inactivated S. Typhimurium induces a humoral and cellular immune response. However, the level of protection afforded by these responses requires further investigation.

DOI 10.1016/j.heliyon.2021.e05903
Citations Scopus - 2Web of Science - 2
2021 Silva-Cayetano A, Foster WS, Innocentin S, Belij-Rammerstorfer S, Spencer AJ, Burton OT, et al., 'A booster dose enhances immunogenicity of the COVID-19 vaccine candidate ChAdOx1 nCoV-19 in aged mice', Med, 2 243-262.e8 (2021) [C1]

Background: The spread of SARS-CoV-2 has caused a worldwide pandemic that has affected almost every aspect of human life. The development of an effective COVID-19 vaccine could li... [more]

Background: The spread of SARS-CoV-2 has caused a worldwide pandemic that has affected almost every aspect of human life. The development of an effective COVID-19 vaccine could limit the morbidity and mortality caused by infection and may enable the relaxation of social-distancing measures. Age is one of the most significant risk factors for poor health outcomes after SARS-CoV-2 infection; therefore, it is desirable that any new vaccine candidates elicit a robust immune response in older adults. Methods: Here, we use in-depth immunophenotyping to characterize the innate and adaptive immune response induced upon intramuscular administration of the adenoviral vectored ChAdOx1 nCoV-19 (AZD-1222) COVID-19 vaccine candidate in mice. Findings: A single vaccination generates spike-specific Th1 cells, Th1-like Foxp3+ regulatory T cells, polyfunctional spike-specific CD8+ T cells. and granzyme-B-producing CD8 effectors. Spike-specific IgG and IgM are generated from both the early extrafollicular antibody response and the T follicular helper cell-supported germinal center reaction, which is associated with the production of virus-neutralizing antibodies. A single dose of this vaccine generated a similar type of immune response in aged mice but of a reduced magnitude than in younger mice. We report that a second dose enhances the immune response to this vaccine in aged mice. Conclusions: This study shows that ChAdOx1 nCoV-19 induces both cellular and humoral immunity in adult and aged mice and suggests a prime-boost strategy is a rational approach to enhance immunogenicity in older persons. Funding: This study was supported by BBSRC, Lister institute of Preventative Medicine, EPSRC VaxHub, and Innovate UK.

DOI 10.1016/j.medj.2020.12.006
Citations Scopus - 56Web of Science - 37
2021 Lambe T, Spencer AJ, Thomas KM, Gooch KE, Thomas S, White AD, et al., 'ChAdOx1 nCoV-19 protection against SARS-CoV-2 in rhesus macaque and ferret challenge models', Communications Biology, 4 (2021) [C1]

Vaccines against SARS-CoV-2 are urgently required, but early development of vaccines against SARS-CoV-1 resulted in enhanced disease after vaccination. Careful assessment of this ... [more]

Vaccines against SARS-CoV-2 are urgently required, but early development of vaccines against SARS-CoV-1 resulted in enhanced disease after vaccination. Careful assessment of this phenomena is warranted for vaccine development against SARS CoV-2. Here we report detailed immune profiling after ChAdOx1 nCoV-19 (AZD1222) and subsequent high dose challenge in two animal models of SARS-CoV-2 mediated disease. We demonstrate in rhesus macaques the lung pathology caused by SARS-CoV-2 mediated pneumonia is reduced by prior vaccination with ChAdOx1 nCoV-19 which induced neutralising antibody responses after a single intramuscular administration. In a second animal model, ferrets, ChAdOx1 nCoV-19 reduced both virus shedding and lung pathology. Antibody titre were boosted by a second dose. Data from these challenge models on the absence of enhanced disease and the detailed immune profiling, support the continued clinical evaluation of ChAdOx1 nCoV-19.

DOI 10.1038/s42003-021-02443-0
Citations Scopus - 13Web of Science - 12
2021 Spencer AJ, McKay PF, Belij-Rammerstorfer S, Ulaszewska M, Bissett CD, Hu K, et al., 'Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice', Nature Communications, 12 (2021) [C1]

Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens usin... [more]

Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens using alternate vaccine modalities. Here, we present a detailed description of the immune response, in mice, following vaccination with a self-amplifying RNA (saRNA) vaccine and an adenoviral vectored vaccine (ChAdOx1 nCoV-19/AZD1222) against SARS-CoV-2. We demonstrate that antibody responses are higher in two-dose heterologous vaccination regimens than single-dose regimens. Neutralising titres after heterologous prime-boost were at least comparable or higher than the titres measured after homologous prime boost vaccination with viral vectors. Importantly, the cellular immune response after a heterologous regimen is dominated by cytotoxic T cells and Th1+ CD4 T cells, which is superior to the response induced in homologous vaccination regimens in mice. These results underpin the need for clinical trials to investigate the immunogenicity of heterologous regimens with alternate vaccine technologies.

DOI 10.1038/s41467-021-23173-1
Citations Scopus - 86Web of Science - 73
2021 Collins KA, Brod F, Snaith R, Ulaszewska M, Longley RJ, Salman AM, et al., 'Ultra-low dose immunization and multi-component vaccination strategies enhance protection against malaria in mice', Scientific Reports, 11 (2021) [C1]

An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protectio... [more]

An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protection in a Phase 3 trial. R21 is a next-generation RTS,S-like vaccine. We have previously shown in mice that R21 administered in Matrix-M is highly immunogenic, able to elicit complete protection against sporozoite challenge, and can be successfully administered with TRAP based viral-vectors resulting in enhanced protection. In this study, we developed a novel, GMP-compatible purification process for R21, and evaluated the immunogenicity and protective efficacy of ultra-low doses of both R21 and RTS,S when formulated in AS01. We demonstrated that both vaccines are highly immunogenic and also elicit comparable high levels of protection against transgenic parasites in BALB/c mice. By lowering the vaccine dose there was a trend for increased immunogenicity and sterile protection, with the highest dose vaccine groups achieving the lowest efficacy (50% sterile protection). We also evaluated the ability to combine RTS,S/AS01 with TRAP based viral-vectors and observed concurrent induction of immune responses to both antigens with minimal interference when mixing the vaccines prior to administration. These studies suggest that R21 or RTS,S could be combined with viral-vectors for a multi-component vaccination approach and indicate that low dose vaccination should be fully explored in humans to maximize potential efficacy.

DOI 10.1038/s41598-021-90290-8
Citations Scopus - 8Web of Science - 1
2021 Flaxman A, Marchevsky NG, Jenkin D, Aboagye J, Aley PK, Angus B, et al., 'Reactogenicity and immunogenicity after a late second dose or a third dose of ChAdOx1 nCoV-19 in the UK: a substudy of two randomised controlled trials (COV001 and COV002)', The Lancet, 398 981-990 (2021) [C1]

Background: COVID-19 vaccine supply shortages are causing concerns about compromised immunity in some countries as the interval between the first and second dose becomes longer. C... [more]

Background: COVID-19 vaccine supply shortages are causing concerns about compromised immunity in some countries as the interval between the first and second dose becomes longer. Conversely, countries with no supply constraints are considering administering a third dose. We assessed the persistence of immunogenicity after a single dose of ChAdOx1 nCoV-19 (AZD1222), immunity after an extended interval (44¿45 weeks) between the first and second dose, and response to a third dose as a booster given 28¿38 weeks after the second dose. Methods: In this substudy, volunteers aged 18¿55 years who were enrolled in the phase 1/2 (COV001) controlled trial in the UK and had received either a single dose or two doses of 5 × 1010 viral particles were invited back for vaccination. Here we report the reactogenicity and immunogenicity of a delayed second dose (44¿45 weeks after first dose) or a third dose of the vaccine (28¿38 weeks after second dose). Data from volunteers aged 18¿55 years who were enrolled in either the phase 1/2 (COV001) or phase 2/3 (COV002), single-blinded, randomised controlled trials of ChAdOx1 nCoV-19 and who had previously received a single dose or two doses of 5 × 1010 viral particles are used for comparison purposes. COV001 is registered with ClinicalTrials.gov, NCT04324606, and ISRCTN, 15281137, and COV002 is registered with ClinicalTrials.gov, NCT04400838, and ISRCTN, 15281137, and both are continuing but not recruiting. Findings: Between March 11 and 21, 2021, 90 participants were enrolled in the third-dose boost substudy, of whom 80 (89%) were assessable for reactogenicity, 75 (83%) were assessable for evaluation of antibodies, and 15 (17%) were assessable for T-cells responses. The two-dose cohort comprised 321 participants who had reactogenicity data (with prime-boost interval of 8¿12 weeks: 267 [83%] of 321; 15¿25 weeks: 24 [7%]; or 44¿45 weeks: 30 [9%]) and 261 who had immunogenicity data (interval of 8¿12 weeks: 115 [44%] of 261; 15¿25 weeks: 116 [44%]; and 44¿45 weeks: 30 [11%]). 480 participants from the single-dose cohort were assessable for immunogenicity up to 44¿45 weeks after vaccination. Antibody titres after a single dose measured approximately 320 days after vaccination remained higher than the titres measured at baseline (geometric mean titre of 66·00 ELISA units [EUs; 95% CI 47·83¿91·08] vs 1·75 EUs [1·60¿1·93]). 32 participants received a late second dose of vaccine 44¿45 weeks after the first dose, of whom 30 were included in immunogenicity and reactogenicity analyses. Antibody titres were higher 28 days after vaccination in those with a longer interval between first and second dose than for those with a short interval (median total IgG titre: 923 EUs [IQR 525¿1764] with an 8¿12 week interval; 1860 EUs [917¿4934] with a 15¿25 week interval; and 3738 EUs [1824¿6625] with a 44¿45 week interval). Among participants who received a third dose of vaccine, antibody titres (measured in 73 [81%] participants for whom samples were available) were significantly higher 28 days after a third dose (median total IgG titre: 3746 EUs [IQR 2047¿6420]) than 28 days after a second dose (median 1792 EUs [IQR 899¿4634]; Wilcoxon signed rank test p=0·0043). T-cell responses were also boosted after a third dose (median response increased from 200 spot forming units [SFUs] per million peripheral blood mononuclear cells [PBMCs; IQR 127¿389] immediately before the third dose to 399 SFUs per milion PBMCs [314¿662] by day 28 after the third dose; Wilcoxon signed rank test p=0·012). Reactogenicity after a late second dose or a third dose was lower than reactogenicity after a first dose. Interpretation: An extended interval before the second dose of ChAdOx1 nCoV-19 leads to increased antibody titres. A third dose of ChAdOx1 nCoV-19 induces antibodies to a level that correlates with high efficacy after second dose and boosts T-cell responses. Funding: UK Research and Innovation, Engineering and Physical Sciences Research Cou...

DOI 10.1016/S0140-6736(21)01699-8
Citations Scopus - 168Web of Science - 132
2021 Barrett JR, Belij-Rammerstorfer S, Dold C, Ewer KJ, Folegatti PM, Gilbride C, et al., 'Phase 1/2 trial of SARS-CoV-2 vaccine ChAdOx1 nCoV-19 with a booster dose induces multifunctional antibody responses', Nature Medicine, 27 279-288 (2021) [C1]

More than 190 vaccines are currently in development to prevent infection by the novel severe acute respiratory syndrome coronavirus 2. Animal studies suggest that while neutralizi... [more]

More than 190 vaccines are currently in development to prevent infection by the novel severe acute respiratory syndrome coronavirus 2. Animal studies suggest that while neutralizing antibodies against the viral spike protein may correlate with protection, additional antibody functions may also be important in preventing infection. Previously, we reported early immunogenicity and safety outcomes of a viral vector coronavirus vaccine, ChAdOx1 nCoV-19 (AZD1222), in a single-blinded phase 1/2 randomized controlled trial of healthy adults aged 18¿55 years (NCT04324606). Now we describe safety and exploratory humoral and cellular immunogenicity of the vaccine, from subgroups of volunteers in that trial, who were subsequently allocated to receive a homologous full-dose (SD/SD D56; n = 20) or half-dose (SD/LD D56; n = 32) ChAdOx1 booster vaccine 56 d following prime vaccination. Previously reported immunogenicity data from the open-label 28-d interval prime-boost group (SD/SD D28; n = 10) are also presented to facilitate comparison. Additionally, we describe volunteers boosted with the comparator vaccine (MenACWY; n = 10). In this interim report, we demonstrate that a booster dose of ChAdOx1 nCoV-19 is safe and better tolerated than priming doses. Using a systems serology approach we also demonstrate that anti-spike neutralizing antibody titers, as well as Fc-mediated functional antibody responses, including antibody-dependent neutrophil/monocyte phagocytosis, complement activation and natural killer cell activation, are substantially enhanced by a booster dose of vaccine. A booster dose of vaccine induced stronger antibody responses than a dose-sparing half-dose boost, although the magnitude of T cell responses did not increase with either boost dose. These data support the two-dose vaccine regime that is now being evaluated in phase 3 clinical trials.

DOI 10.1038/s41591-020-01179-4
Citations Scopus - 216Web of Science - 182
2020 Halbroth BR, Sebastian S, Salman AM, Ulaszewska M, Gola A, Longley RJ, et al., 'Preclinical development and assessment of viral vectors expressing a fusion antigen of plasmodium falciparum LSA1 and LSAP2 for efficacy against liver-stage malaria', Infection and Immunity, 88 (2020) [C1]

Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-m... [more]

Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-mediated protection from liver-stage malaria relies on a sufficient number of antigen-specific T cells reaching the liver during the time that parasites are present. A single vaccine expressing two antigens could potentially increase both the size and breadth of the antigen-specific response while halving vaccine production costs. In this study, we investigated combining two liver-stage antigens, P. falciparum LSA1 (PfLSA1) and PfLSAP2, and investigated the induction of protective efficacy by coadministration of single-antigen vectors or vaccination with dual-antigen vectors, using simian adenovirus and modified vaccinia virus Ankara vectors. The efficacy of these vaccines was assessed in mouse malaria challenge models using chimeric P. berghei parasites expressing the relevant P. falciparum antigens and challenging mice at the peak of the T cell response. Vaccination with a combination of the single-antigen vectors expressing PfLSA1 or PfLSAP2 was shown to improve protective efficacy compared to vaccination with each single-antigen vector alone. Vaccination with dual-antigen vectors expressing both PfLSA1 and PfLSAP2 resulted in responses to both antigens, particularly in outbred mice, and most importantly, the efficacy was equivalent to that of vaccination with a mixture of singleantigen vectors. Based on these promising data, dual-antigen vectors expressing PfLSA1 and PfLSAP2 will now proceed to manufacturing and clinical assessment under good manufacturing practice (GMP) guidelines.

DOI 10.1128/IAI.00573-19
Citations Scopus - 5Web of Science - 4
2020 van Doremalen N, Lambe T, Spencer A, Belij-Rammerstorfer S, Purushotham JN, Port JR, et al., 'ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques', Nature, 586 578-582 (2020) [C1]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 20191,2 and is responsible for the coronavirus disease 2019 (COVID-19) pandemic3. Vaccines are an ... [more]

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 20191,2 and is responsible for the coronavirus disease 2019 (COVID-19) pandemic3. Vaccines are an essential countermeasure and are urgently needed to control the pandemic4. Here we show that the adenovirus-vector-based vaccine ChAdOx1¿nCoV-19, which encodes the spike protein of SARS-CoV-2, is immunogenic in mice and elicites a robust humoral and cell-mediated response. This response was predominantly mediated by type-1 T helper cells, as demonstrated by the profiling of the IgG subclass and the expression of cytokines. Vaccination with ChAdOx1¿nCoV-19 (using either a prime-only or a prime¿boost regimen) induced a balanced humoral and cellular immune response of type-1 and type-2 T helper cells in rhesus macaques. We observed a significantly reduced viral load in the bronchoalveolar lavage fluid and lower respiratory tract tissue of vaccinated rhesus macaques that were challenged with SARS-CoV-2 compared with control animals, and no pneumonia was observed in vaccinated SARS-CoV-2-infected animals. However, there was no difference in nasal shedding between vaccinated and control SARS-CoV-2-infected macaques. Notably, we found no evidence of immune-enhanced disease after viral challenge in vaccinated SARS-CoV-2-infected animals. The safety, immunogenicity and efficacy profiles of ChAdOx1¿nCoV-19 against symptomatic PCR-positive COVID-19 disease will now be assessed in randomized controlled clinical trials in humans.

DOI 10.1038/s41586-020-2608-y
Citations Scopus - 633Web of Science - 555
2020 Rollier CS, Spencer AJ, Sogaard KC, Honeycutt J, Furze J, Bregu M, et al., 'Modification of Adenovirus vaccine vector-induced immune responses by expression of a signalling molecule', Scientific Reports, 10 (2020) [C1]

Adenoviral vectors are being developed as vaccines against infectious agents and tumour-associated antigens, because of their ability to induce cellular immunity. However, the pro... [more]

Adenoviral vectors are being developed as vaccines against infectious agents and tumour-associated antigens, because of their ability to induce cellular immunity. However, the protection afforded in animal models has not easily translated into primates and clinical trials, underlying the need for improving adenoviral vaccines-induced immunogenicity. A Toll-like receptor signalling molecule, TRAM, was assessed for its ability to modify the immune responses induced by an adenovirus-based vaccine. Different adenovirus vectors either expressing TRAM alone or co-expressing TRAM along with a model antigen were constructed. The modification of T-cell and antibody responses induced by TRAM was assessed in vivo in mice and in primates. Co-expression of TRAM and an antigen from adenoviruses increased the transgene-specific CD8+ T cell responses in mice. Similar effects were seen when a TRAM expressing virus was co-administered with the antigen-expressing adenovirus. However, in primate studies, co-administration of a TRAM expressing adenovirus with a vaccine expressing the ME-TRAP malaria antigen had no significant effect on the immune responses. While these results support the idea that modification of innate immune signalling by genetic vectors modifies immunogenicity, they also emphasise the difficulty in generalising results from rodents into primates, where the regulatory pathway may be different to that in mice.

DOI 10.1038/s41598-020-61730-8
Citations Scopus - 9Web of Science - 7
2020 Sebastian S, Flaxman A, Cha KM, Ulaszewska M, Gilbride C, Sharpe H, et al., 'A multi-filovirus vaccine candidate: Co-expression of Ebola, Sudan, and Marburg antigens in a single vector', Vaccines, 8 (2020) [C1]

In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leavin... [more]

In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leaving individuals susceptible to disease after exposure to related virus species. This is a significant hurdle in the field of vaccine development, in which broadly protective vaccines represent an unmet need. This is particularly evident for filoviruses, as there are multiple family members that can cause lethal haemorrhagic fever, including Zaire ebolavirus, Sudan ebolavirus, and Marburg virus. In an attempt to address this need, both pre-clinical and clinical studies previously used mixed or co-administered monovalent vaccines to prevent filovirus mediated disease. However, these multi-vaccine and multi-dose vaccination regimens do not represent a practical immunisation scheme when considering the target endemic areas. We describe here the development of a single multi-pathogen filovirus vaccine candidate based on a replication-deficient simian adenoviral vector. Our vaccine candidate encodes three different filovirus glycoproteins in one vector and induces strong cellular and humoral immunity to all three viral glycoproteins after a single vaccination. Crucially, it was found to be protective in a stringent Zaire ebolavirus challenge in guinea pigs in a one-shot vaccination regimen. This trivalent filovirus vaccine offers a tenable vaccine product that could be rapidly translated to the clinic to prevent filovirus-mediated viral haemorrhagic fever.

DOI 10.3390/vaccines8020241
Citations Scopus - 9Web of Science - 6
2020 Graham SP, McLean RK, Spencer AJ, Belij-Rammerstorfer S, Wright D, Ulaszewska M, et al., 'Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19', npj Vaccines, 5 (2020) [C1]

Clinical development of the COVID-19 vaccine candidate ChAdOx1 nCoV-19, a replication-deficient simian adenoviral vector expressing the full-length SARS-CoV-2 spike (S) protein wa... [more]

Clinical development of the COVID-19 vaccine candidate ChAdOx1 nCoV-19, a replication-deficient simian adenoviral vector expressing the full-length SARS-CoV-2 spike (S) protein was initiated in April 2020 following non-human primate studies using a single immunisation. Here, we compared the immunogenicity of one or two doses of ChAdOx1 nCoV-19 in both mice and pigs. Whilst a single dose induced antigen-specific antibody and T cells responses, a booster immunisation enhanced antibody responses, particularly in pigs, with a significant increase in SARS-CoV-2 neutralising titres.

DOI 10.1038/s41541-020-00221-3
Citations Scopus - 96Web of Science - 81
2020 Folegatti PM, Ewer KJ, Aley PK, Angus B, Becker S, Belij-Rammerstorfer S, et al., 'Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial', The Lancet, 396 467-478 (2020) [C1]

Background: The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) might be curtailed by vaccination. We assessed the safety, reactogenicity, and immunogenic... [more]

Background: The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) might be curtailed by vaccination. We assessed the safety, reactogenicity, and immunogenicity of a viral vectored coronavirus vaccine that expresses the spike protein of SARS-CoV-2. Methods: We did a phase 1/2, single-blind, randomised controlled trial in five trial sites in the UK of a chimpanzee adenovirus-vectored vaccine (ChAdOx1 nCoV-19) expressing the SARS-CoV-2 spike protein compared with a meningococcal conjugate vaccine (MenACWY) as control. Healthy adults aged 18¿55 years with no history of laboratory confirmed SARS-CoV-2 infection or of COVID-19-like symptoms were randomly assigned (1:1) to receive ChAdOx1 nCoV-19 at a dose of 5 × 1010 viral particles or MenACWY as a single intramuscular injection. A protocol amendment in two of the five sites allowed prophylactic paracetamol to be administered before vaccination. Ten participants assigned to a non-randomised, unblinded ChAdOx1 nCoV-19 prime-boost group received a two-dose schedule, with the booster vaccine administered 28 days after the first dose. Humoral responses at baseline and following vaccination were assessed using a standardised total IgG ELISA against trimeric SARS-CoV-2 spike protein, a muliplexed immunoassay, three live SARS-CoV-2 neutralisation assays (a 50% plaque reduction neutralisation assay [PRNT50]; a microneutralisation assay [MNA50, MNA80, and MNA90]; and Marburg VN), and a pseudovirus neutralisation assay. Cellular responses were assessed using an ex-vivo interferon-¿ enzyme-linked immunospot assay. The co-primary outcomes are to assess efficacy, as measured by cases of symptomatic virologically confirmed COVID-19, and safety, as measured by the occurrence of serious adverse events. Analyses were done by group allocation in participants who received the vaccine. Safety was assessed over 28 days after vaccination. Here, we report the preliminary findings on safety, reactogenicity, and cellular and humoral immune responses. The study is ongoing, and was registered at ISRCTN, 15281137, and ClinicalTrials.gov, NCT04324606. Findings: Between April 23 and May 21, 2020, 1077 participants were enrolled and assigned to receive either ChAdOx1 nCoV-19 (n=543) or MenACWY (n=534), ten of whom were enrolled in the non-randomised ChAdOx1 nCoV-19 prime-boost group. Local and systemic reactions were more common in the ChAdOx1 nCoV-19 group and many were reduced by use of prophylactic paracetamol, including pain, feeling feverish, chills, muscle ache, headache, and malaise (all p<0·05). There were no serious adverse events related to ChAdOx1 nCoV-19. In the ChAdOx1 nCoV-19 group, spike-specific T-cell responses peaked on day 14 (median 856 spot-forming cells per million peripheral blood mononuclear cells, IQR 493¿1802; n=43). Anti-spike IgG responses rose by day 28 (median 157 ELISA units [EU], 96¿317; n=127), and were boosted following a second dose (639 EU, 360¿792; n=10). Neutralising antibody responses against SARS-CoV-2 were detected in 32 (91%) of 35 participants after a single dose when measured in MNA80 and in 35 (100%) participants when measured in PRNT50. After a booster dose, all participants had neutralising activity (nine of nine in MNA80 at day 42 and ten of ten in Marburg VN on day 56). Neutralising antibody responses correlated strongly with antibody levels measured by ELISA (R2=0·67 by Marburg VN; p<0·001). Interpretation: ChAdOx1 nCoV-19 showed an acceptable safety profile, and homologous boosting increased antibody responses. These results, together with the induction of both humoral and cellular immune responses, support large-scale evaluation of this candidate vaccine in an ongoing phase 3 programme. Funding: UK Research and Innovation, Coalition for Epidemic Preparedness Innovations, National Institute for Health Research (NIHR), NIHR Oxford Biomedical Research Centre, Thames Valley and South Midland's NIHR Clinical Research Network, and...

DOI 10.1016/S0140-6736(20)31604-4
Citations Scopus - 1761Web of Science - 1401
2020 Ramasamy MN, Minassian AM, Ewer KJ, Flaxman AL, Folegatti PM, Owens DR, et al., 'Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial', The Lancet, 396 1979-1993 (2020) [C1]

Background: Older adults (aged =70 years) are at increased risk of severe disease and death if they develop COVID-19 and are therefore a priority for immunisation should an effica... [more]

Background: Older adults (aged =70 years) are at increased risk of severe disease and death if they develop COVID-19 and are therefore a priority for immunisation should an efficacious vaccine be developed. Immunogenicity of vaccines is often worse in older adults as a result of immunosenescence. We have reported the immunogenicity of a novel chimpanzee adenovirus-vectored vaccine, ChAdOx1 nCoV-19 (AZD1222), in young adults, and now describe the safety and immunogenicity of this vaccine in a wider range of participants, including adults aged 70 years and older. Methods: In this report of the phase 2 component of a single-blind, randomised, controlled, phase 2/3 trial (COV002), healthy adults aged 18 years and older were enrolled at two UK clinical research facilities, in an age-escalation manner, into 18¿55 years, 56¿69 years, and 70 years and older immunogenicity subgroups. Participants were eligible if they did not have severe or uncontrolled medical comorbidities or a high frailty score (if aged =65 years). First, participants were recruited to a low-dose cohort, and within each age group, participants were randomly assigned to receive either intramuscular ChAdOx1 nCoV-19 (2·2 × 1010 virus particles) or a control vaccine, MenACWY, using block randomisation and stratified by age and dose group and study site, using the following ratios: in the 18¿55 years group, 1:1 to either two doses of ChAdOx1 nCoV-19 or two doses of MenACWY; in the 56¿69 years group, 3:1:3:1 to one dose of ChAdOx1 nCoV-19, one dose of MenACWY, two doses of ChAdOx1 nCoV-19, or two doses of MenACWY; and in the 70 years and older, 5:1:5:1 to one dose of ChAdOx1 nCoV-19, one dose of MenACWY, two doses of ChAdOx1 nCoV-19, or two doses of MenACWY. Prime-booster regimens were given 28 days apart. Participants were then recruited to the standard-dose cohort (3·5¿6·5 × 1010 virus particles of ChAdOx1 nCoV-19) and the same randomisation procedures were followed, except the 18¿55 years group was assigned in a 5:1 ratio to two doses of ChAdOx1 nCoV-19 or two doses of MenACWY. Participants and investigators, but not staff administering the vaccine, were masked to vaccine allocation. The specific objectives of this report were to assess the safety and humoral and cellular immunogenicity of a single-dose and two-dose schedule in adults older than 55 years. Humoral responses at baseline and after each vaccination until 1 year after the booster were assessed using an in-house standardised ELISA, a multiplex immunoassay, and a live severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) microneutralisation assay (MNA80). Cellular responses were assessed using an ex-vivo IFN-¿ enzyme-linked immunospot assay. The coprimary outcomes of the trial were efficacy, as measured by the number of cases of symptomatic, virologically confirmed COVID-19, and safety, as measured by the occurrence of serious adverse events. Analyses were by group allocation in participants who received the vaccine. Here, we report the preliminary findings on safety, reactogenicity, and cellular and humoral immune responses. This study is ongoing and is registered with ClinicalTrials.gov, NCT04400838, and ISRCTN, 15281137. Findings: Between May 30 and Aug 8, 2020, 560 participants were enrolled: 160 aged 18¿55 years (100 assigned to ChAdOx1 nCoV-19, 60 assigned to MenACWY), 160 aged 56¿69 years (120 assigned to ChAdOx1 nCoV-19: 40 assigned to MenACWY), and 240 aged 70 years and older (200 assigned to ChAdOx1 nCoV-19: 40 assigned to MenACWY). Seven participants did not receive the boost dose of their assigned two-dose regimen, one participant received the incorrect vaccine, and three were excluded from immunogenicity analyses due to incorrectly labelled samples. 280 (50%) of 552 analysable participants were female. Local and systemic reactions were more common in participants given ChAdOx1 nCoV-19 than in those given the control vaccine, and similar in nature to those previously reported (injection...

DOI 10.1016/S0140-6736(20)32466-1
Citations Scopus - 1061Web of Science - 889
2018 Halbroth BR, Sebastian S, Poyntz HC, Bregu M, Cottingham MG, Hill AVS, Spencer AJ, 'Development of a Molecular Adjuvant to Enhance Antigen-Specific CD8(+) T Cell Responses', SCIENTIFIC REPORTS, 8 (2018) [C1]
DOI 10.1038/s41598-018-33375-1
Citations Scopus - 16Web of Science - 13
2018 Gola A, Silman D, Walters AA, Sridhar S, Uderhardt S, Salman AM, et al., 'Prime and target immunization protects against liver-stage malaria in mice', Science Translational Medicine, 10 (2018) [C1]

Despite recent advances in treatment and vector control, malaria is still a leading cause of death, emphasizing the need for an effective vaccine. The malaria life cycle can be su... [more]

Despite recent advances in treatment and vector control, malaria is still a leading cause of death, emphasizing the need for an effective vaccine. The malaria life cycle can be subdivided into three stages: the invasion and growth within liver hepatocytes (pre-erythrocytic stage), the blood stage (erythrocytic stage), and, finally, the sexual stage (occurring within the mosquito vector). Antigen (Ag)-specific CD8+ T cells are effectively induced by heterologous prime-boost viral vector immunization and known to correlate with liver-stage protection. However, liver-stage malaria vaccines have struggled to generate and maintain the high numbers of Plasmodium-specific circulating T cells necessary to confer sterile protection. We describe an alternative "prime and target" vaccination strategy aimed specifically at inducing high numbers of tissue-resident memory T cells present in the liver at the time of hepatic infection. This approach bypasses the need for very high numbers of circulating T cells and markedly increases the efficacy of subunit immunization against liver-stage malaria with clinically relevant Ags and clinically tested viral vectors in murine challenge models. Translation to clinical use has begun, with encouraging results from a pilot safety and feasibility trial of intravenous chimpanzee adenovirus vaccination in humans. This work highlights the value of a prime-target approach for immunization against malaria and suggests that this strategy may represent a more general approach for prophylaxis or immunotherapy of other liver infections and diseases.

DOI 10.1126/scitranslmed.aap9128
Citations Scopus - 57Web of Science - 44
2018 Brod F, Miura K, Taylor I, Li Y, Marini A, Salman AM, et al., 'Combination of RTS,S and Pfs25-IMX313 Induces a Functional Antibody Response Against Malaria Infection and Transmission in Mice', Frontiers in Immunology, 9 (2018) [C1]

The last two decades saw a dramatic reduction in malaria incidence rates, but this decrease has been stalling recently, indicating control measures are starting to fail. An effect... [more]

The last two decades saw a dramatic reduction in malaria incidence rates, but this decrease has been stalling recently, indicating control measures are starting to fail. An effective vaccine, particularly one with a marked effect on disease transmission, would undoubtedly be an invaluable tool for efforts to control and eliminate malaria. RTS,S/AS01, the most advanced malaria vaccine to date, targets the parasite before it invades the liver and has the potential to prevent malaria disease as well as transmission by preventing blood stage infection and therefore gametocytogenesis. Unfortunately efficacy in a phase III clinical trial was limited and it is widely believed that a malaria vaccine needed to contain multiple antigens from different life-cycle stages to have a realistic chance of success. A recent study in mice has shown that partially efficacious interventions targeting the pre-erythrocytic and the sexual lifecycle stage synergise in eliminating malaria from a population over multiple generations. Hence, the combination of RTS,S/AS01 with a transmission blocking vaccine (TBV) is highly appealing as a pragmatic and powerful way to increase vaccine efficacy. Here we demonstrate that combining Pfs25-IMX313, one of the TBV candidates currently in clinical development, with RTS,S/AS01 readily induces a functional immune response against both antigens in outbred CD1 mice. Formulation of Pfs25-IMX313 in AS01 significantly increased antibody titres when compared to formulation in Alhydrogel, resulting in improved transmission reducing activity in standard membrane feeding assays (SMFA). Upon co-formulation of Pfs25-IMX313 with RTS,S/AS01, the immunogenicity of both vaccines was maintained, and functional assessment of the induced antibody response by SMFA and inhibition of sporozoite invasion assay (ISI) showed no reduction in biological activity against parasites of both lifecycle stages. Should this findings be translatable to human vaccination this could greatly aid efforts to eliminate and eventually eradicate malaria.

DOI 10.3389/fimmu.2018.02780
Citations Scopus - 23Web of Science - 20
2017 Longley RJ, Halbroth BR, Salman AM, Ewer KJ, Hodgson SH, Janse CJ, et al., 'Assessment of the Plasmodium falciparum preerythrocytic antigen UIS3 as a potential candidate for a malaria vaccine', Infection and Immunity, 85 (2017) [C1]

Efforts are under way to improve the efficacy of subunit malaria vaccines through assessments of new adjuvants, vaccination platforms, and antigens. In this study, we further asse... [more]

Efforts are under way to improve the efficacy of subunit malaria vaccines through assessments of new adjuvants, vaccination platforms, and antigens. In this study, we further assessed the Plasmodium falciparum antigen upregulated in infective sporozoites 3 (PfUIS3) as a vaccine candidate. PfUIS3 was expressed in the viral vectors chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) and used to immunize mice in a prime-boost regimen. We previously demonstrated that this regimen could provide partial protection against challenge with chimeric P. berghei parasites expressing PfUIS3. We now show that ChAd63-MVA PfUIS3 can also provide partial cross-species protection against challenge with wild-type P. berghei parasites. We also show that PfUIS3-specific cellular memory responses could be recalled in human volunteers exposed to P. falciparum parasites in a controlled human malaria infection study. When ChAd63-MVA PfUIS3 was coadministered with the vaccine candidate P. falciparum thrombospondin-related adhesion protein (PfTRAP) expressed in the ChAd63-MVA system, there was no significant change in immunogenicity to either vaccine. However, when mice were challenged with double chimeric P. berghei-P. falciparum parasites expressing both PfUIS3 and PfTRAP, vaccine efficacy was improved to 100% sterile protection. This synergistic effect was evident only when the two vaccines were mixed and administered at the same site. We have therefore demonstrated that vaccination with PfUIS3 can induce a consistent delay in patent parasitemia across mouse strains and against chimeric parasites expressing PfUIS3 as well as wild-type P. berghei; when this vaccine is combined with another partially protective regimen (ChAd63-MVA PfTRAP), complete protection is induced.

DOI 10.1128/IAI.00641-16
Citations Scopus - 15Web of Science - 15
2017 Spencer AJ, Longley RJ, Gola A, Ulaszewska M, Lambe T, Hill AVS, 'The Threshold of protection from liver-stage malaria relies on a fine balance between the number of infected hepatocytes and effector cd8

Since the demonstration of sterile protection afforded by injection of irradiated sporozoites, CD8+ T cells have been shown to play a significant role in protection from liver-sta... [more]

Since the demonstration of sterile protection afforded by injection of irradiated sporozoites, CD8+ T cells have been shown to play a significant role in protection from liver-stage malaria. This is, however, dependent on the presence of an extremely high number of circulating effector cells, thought to be necessary to scan, locate, and kill infected hepatocytes in the short time that parasites are present in the liver. We used an adoptive transfer model to elucidate the kinetics of the effector CD8+ T cell response in the liver following Plasmodium berghei sporozoite challenge. Although effector CD8+ T cells require <24 h to find, locate, and kill infected hepatocytes, active migration of Ag-specific CD8+ T cells into the liver was not observed during the 2-d liver stage of infection, as divided cells were only detected from day 3 postchallenge. However, the percentage of donor cells recruited into division was shown to indicate the level of Ag presentation from infected hepatocytes. By titrating the number of transferred Ag-specific effector CD8+ T cells and sporozoites, we demonstrate that achieving protection toward liver-stage malaria is reliant on CD8+ T cells being able to locate infected hepatocytes, resulting in a protection threshold dependent on a fine balance between the number of infected hepatocytes and CD8+ T cells present in the liver. With such a fine balance determining protection, achieving a high number of CD8+ T cells will be critical to the success of a cell-mediated vaccine against liver-stage malaria.

DOI 10.4049/jimmunol.1601209
Citations Scopus - 13Web of Science - 11
2017 Ewer K, Sebastian S, Spencer AJ, Gilbert S, Hill AVS, Lambe T, 'Chimpanzee adenoviral vectors as vaccines for outbreak pathogens', Human Vaccines and Immunotherapeutics, 13 3020-3032 (2017) [C1]

The 2014-15 Ebola outbreak in West Africa highlighted the potential for large disease outbreaks caused by emerging pathogens and has generated considerable focus on preparedness f... [more]

The 2014-15 Ebola outbreak in West Africa highlighted the potential for large disease outbreaks caused by emerging pathogens and has generated considerable focus on preparedness for future epidemics. Here we discuss drivers, strategies and practical considerations for developing vaccines against outbreak pathogens. Chimpanzee adenoviral (ChAd) vectors have been developed as vaccine candidates for multiple infectious diseases and prostate cancer. ChAd vectors are safe and induce antigen-specific cellular and humoral immunity in all age groups, as well as circumventing the problem of pre-existing immunity encountered with human Ad vectors. For these reasons, such viral vectors provide an attractive platform for stockpiling vaccines for emergency deployment in response to a threatened outbreak of an emerging pathogen. Work is already underway to develop vaccines against a number of other outbreak pathogens and we will also review progress on these approaches here, particularly for Lassa fever, Nipah and MERS.

DOI 10.1080/21645515.2017.1383575
Citations Scopus - 60Web of Science - 50
2017 Rodríguez-Galán A, Salman AM, Bowyer G, Collins KA, Longley RJ, Brod F, et al., 'An in vitro assay to measure antibody-mediated inhibition of P. berghei sporozoite invasion against P. falciparum antigens', Scientific Reports, 7 (2017) [C1]

A large research effort is currently underway to find an effective and affordable malaria vaccine. Tools that enable the rapid evaluation of protective immune responses are essent... [more]

A large research effort is currently underway to find an effective and affordable malaria vaccine. Tools that enable the rapid evaluation of protective immune responses are essential to vaccine development as they can provide selection criteria to rank order vaccine candidates. In this study we have revisited the Inhibition of Sporozoite Invasion (ISI) assay to assess the ability of antibodies to inhibit sporozoite infection of hepatocytes. By using GFP expressing sporozoites of the rodent parasite P. berghei we are able to robustly quantify parasite infection of hepatocyte cell lines by flow cytometry. In conjunction with recently produced transgenic P. berghei parasites that express P. falciparum sporozoite antigens, we have been able to use this assay to measure antibody mediated inhibition of sporozoite invasion against one of the lead malaria antigens P. falciparum CSP. By combining chimeric rodent parasites expressing P. falciparum antigens and a flow cytometric readout of infection, we are able to robustly assess vaccine-induced antibodies, from mice, rhesus macaques and human clinical trials, for their functional ability to block sporozoite invasion of hepatocytes.

DOI 10.1038/s41598-017-17274-5
Citations Scopus - 10Web of Science - 9
2016 Alharbi NK, Spencer AJ, Salman AM, Tully CM, Chinnakannan SK, Lambe T, et al., 'Enhancing cellular immunogenicity of MVA-vectored vaccines by utilizing the F11L endogenous promoter', Vaccine, 34 49-55 (2016) [C1]

Modified vaccinia virus Ankara (MVA)-vectored vaccines against malaria, influenza, tuberculosis and recently Ebola virus are in clinical development. Although this vector is safe ... [more]

Modified vaccinia virus Ankara (MVA)-vectored vaccines against malaria, influenza, tuberculosis and recently Ebola virus are in clinical development. Although this vector is safe and immunogenic in humans, efforts remain on-going to enhance immunogenicity through various approaches such as using stronger promoters to boost transgene expression. We previously reported that endogenous MVA promoters such as pB8 and pF11 increased transgene expression and immunogenicity, as compared to the conventional p7.5 promoter. Here, we show that both promoters also rivalled the mH5 promoter in enhancing MVA immunogenicity. We investigated the mechanisms behind this improved immunogenicity and show that it was a result of strong early transgene expression in vivo, rather than in vitro as would normally be assessed. Moreover, keeping the TK gene intact resulted in a modest improvement in immunogenicity. Utilizing pB8 or pF11 as ectopic promoters at the TK locus instead of their natural loci also increased transgene expression and immunogenicity. In addition to a reporter antigen, the pF11 promoter was tested with the expression of two vaccine antigens for which cellular immunogenicity was significantly increased as compared to the p7.5 promoter. Our data support the use of the pF11 and pB8 promoters for improved immunogenicity in future MVA-vectored candidate vaccines.

DOI 10.1016/j.vaccine.2015.11.028
Citations Scopus - 13Web of Science - 13
2016 Ewer KJ, Lambe T, Rollier CS, Spencer AJ, Hill AVS, Dorrell L, 'Viral vectors as vaccine platforms: From immunogenicity to impact', Current Opinion in Immunology, 41 47-54 (2016) [C1]

Viral vectors are the vaccine platform of choice for many pathogens that have thwarted efforts towards control using conventional vaccine approaches. Although the STEP trial encum... [more]

Viral vectors are the vaccine platform of choice for many pathogens that have thwarted efforts towards control using conventional vaccine approaches. Although the STEP trial encumbered development of recombinant human adenovirus vectors only a few years ago, replication-deficient simian adenoviruses have since emerged as a crucial component of clinically effective prime-boost regimens. The vectors discussed here elicit functionally relevant cellular and humoral immune responses, at extremes of age and in diverse populations. The recent Ebola virus outbreak highlighted the utility of viral vectored vaccines in facilitating a rapid response to public health emergencies. Meanwhile, technological advances in manufacturing to support scale-up of viral vectored vaccines have helped to consolidate their position as a leading approach to tackling 'old' and emerging infections.

DOI 10.1016/j.coi.2016.05.014
Citations Scopus - 137Web of Science - 113
2016 Morris SJ, Sebastian S, Spencer AJ, Gilbert SC, 'Simian adenoviruses as vaccine vectors', Future Virology, 11 649-659 (2016) [C1]

Replication incompetent human adenovirus serotype 5 (HAdV-C5) has been extensively used as a delivery vehicle for gene therapy proteins and infectious disease antigens. These vect... [more]

Replication incompetent human adenovirus serotype 5 (HAdV-C5) has been extensively used as a delivery vehicle for gene therapy proteins and infectious disease antigens. These vectors infect replicating and nonreplicating cells, have a broad tissue tropism, elicit high immune responses and are easily purified to high titers. However, the utility of HAdV-C5 vectors as potential vaccines is limited due to pre-existing immunity within the human population that significantly reduces the immunogenicity of HAdV-C5 vaccines. In recent years, adenovirus vaccine development has focused on simian-derived adenoviral vectors, which have the desirable vector characteristics of HAdV-C5 but with negligible seroprevalence in the human population. Here, we discuss recent advances in simian adenovirus vaccine vector development and evaluate current research specifically focusing on clinical trial data.

DOI 10.2217/fvl-2016-0070
Citations Scopus - 65Web of Science - 51
2016 Hodgson SH, Llewellyn D, Silk SE, Milne KH, Elias SC, Miura K, et al., 'Changes in serological immunology measures in UK and Kenyan adults post-controlled human malaria infection', Frontiers in Microbiology, 7 (2016) [C1]

Background: The timing of infection is closely determined in controlled human malaria infection (CHMI) studies, and as such they provide a unique opportunity to dissect changes in... [more]

Background: The timing of infection is closely determined in controlled human malaria infection (CHMI) studies, and as such they provide a unique opportunity to dissect changes in immunological responses before and after a single infection. The first Kenyan Challenge Study (KCS) (Pan African Clinical Trial Registry: PACTR20121100033272) was performed in 2013 with the aim of establishing the CHMI model in Kenya. This study used aseptic, cryopreserved, attenuated Plasmodium falciparum sporozoites administered by needle and syringe (PfSPZ Challenge) and was the first to evaluate parasite dynamics post-CHMI in individuals with varying degrees of prior exposure to malaria. Methods: We describe detailed serological and functional immunological responses pre- and post-CHMI for participants in the KCS and compare these with those from malaria-naïve UK volunteers who also underwent CHMI (VAC049) (ClinicalTrials.gov NCT01465048) using PfSPZ Challenge. We assessed antibody responses to three key blood-stage merozoite antigens [merozoite surface protein 1 (MSP1), apical membrane protein 1 (AMA1), and reticulocyte-binding protein homolog 5 (RH5)] and functional activity using two candidate measures of anti-merozoite immunity; the growth inhibition activity (GIA) assay and the antibody-dependent respiratory burst activity (ADRB) assay. Results:Clear serological differences were observed pre- and post-CHMI by ELISA between malaria-naïve UK volunteers in VAC049, and Kenyan volunteers who had prior malaria exposure. Antibodies to AMA1 and schizont extract correlated with parasite multiplication rate (PMR) post-CHMI in KCS. Serum from volunteer 110 in KCS, who demonstrated a dramatically reduced PMR in vivo, had no in vitro GIA prior to CHMI but the highest level of ADRB activity. A significant difference in ADRB activity was seen between KCS volunteers with minimal and definite prior exposure to malaria and significant increases were seen in ADRB activity post-CHMI in Kenyan volunteers. Quinine and atovaquone/proguanil, previously assumed to be removed by IgG purification, were identified as likely giving rise to aberrantly high in vitro GIA results. Conclusions: The ADRB activity assay is a promising functional assay that warrants further investigation as a measure of prior exposure to malaria and predictor of control of parasite growth. The CHMI model can be used to evaluate potential measures of naturally-acquired immunity to malaria.

DOI 10.3389/fmicb.2016.01604
Citations Scopus - 15Web of Science - 14
2015 Dicks MDJ, Guzman E, Spencer AJ, Gilbert SC, Charleston B, Hill AVS, Cottingham MG, 'The relative magnitude of transgene-specific adaptive immune responses induced by human and chimpanzee adenovirus vectors differs between laboratory animals and a target species', VACCINE, 33 1121-1128 (2015)
DOI 10.1016/j.vaccine.2015.01.042
Citations Scopus - 20Web of Science - 20
2015 Longley RJ, Hill AVS, Spencer AJ, 'Malaria vaccines: identifying Plasmodium falciparum liver-stage targets', FRONTIERS IN MICROBIOLOGY, 6 (2015)
DOI 10.3389/fmicb.2015.00965
Citations Scopus - 14Web of Science - 13
2015 Douglas AD, Baldeviano GC, Lucas CM, Lugo-Roman LA, Crosnier C, Bartholdson SJ, et al., 'A PfRH5-Based Vaccine Is Efficacious against Heterologous Strain Blood-Stage Plasmodium falciparum Infection in Aotus Monkeys', CELL HOST & MICROBE, 17 130-139 (2015)
DOI 10.1016/j.chom.2014.11.017
Citations Scopus - 151Web of Science - 137
2015 Longley RJ, Bauza K, Ewer KJ, Hill AVS, Spencer AJ, 'Development of an In Vitro Assay and Demonstration of Plasmodium berghei Liver-Stage Inhibition by TRAP-Specific CD8(+) T Cells', PLOS ONE, 10 (2015)
DOI 10.1371/journal.pone.0119880
Citations Scopus - 17Web of Science - 15
2015 Alharbi NK, Spencer AJ, Hill AVS, Gilbert SC, 'Deletion of Fifteen Open Reading Frames from Modified Vaccinia Virus Ankara Fails to Improve Immunogenicity', PLOS ONE, 10 (2015)
DOI 10.1371/journal.pone.0128626
Citations Scopus - 10Web of Science - 9
2015 Longley RJ, Halbroth BR, Ewer KJ, Hill AVS, Spencer AJ, 'Identification of Immunodominant Responses to the Plasmodium falciparum Antigens PfUIS3, PfLSA1 and PfLSAP2 in Multiple Strains of Mice', PLOS ONE, 10 (2015)
DOI 10.1371/journal.pone.0144515
Citations Scopus - 5Web of Science - 5
2015 Longley RJ, Salman AM, Cottingham MG, Ewer K, Janse CJ, Khan SM, et al., 'Comparative assessment of vaccine vectors encoding ten malaria antigens identifies two protective liver-stage candidates', SCIENTIFIC REPORTS, 5 (2015)
DOI 10.1038/srep11820
Citations Scopus - 45Web of Science - 44
2015 Dicks MDJ, Spencer AJ, Coughlan L, Bauza K, Gilbert SC, Hill AVS, Cottingham MG, 'Differential immunogenicity between HAdV-5 and chimpanzee adenovirus vector ChAdOx1 is independent of fiber and penton RGD loop sequences in mice', SCIENTIFIC REPORTS, 5 (2015)
DOI 10.1038/srep16756
Citations Scopus - 28Web of Science - 26
2014 Bliss CM, Spencer AJ, Salman AM, Longley RJ, Galan AR, Khan SM, et al., 'Development of an in vitro Plasmodium parasite killing assay for the evaluation of cell-mediated immune responses following vaccination with pre-erythrocytic malaria vaccine candidates', IMMUNOLOGY, 143 74-75 (2014)
2014 Kimani D, Jagne YJ, Cox M, Kimani E, Bliss CM, Gitau E, et al., 'Translating the Immunogenicity of Prime-boost Immunization With ChAd63 and MVA ME-TRAP From Malaria Naive to Malaria-endemic Populations', MOLECULAR THERAPY, 22 1992-2003 (2014)
DOI 10.1038/mt.2014.109
Citations Scopus - 45Web of Science - 41
2014 Spencer AJ, Cottingham MG, Jenks JA, Longley RJ, Capone S, Colloca S, et al., 'Enhanced Vaccine-Induced CD8(+) T Cell Responses to Malaria Antigen ME-TRAP by Fusion to MHC Class II Invariant Chain', PLOS ONE, 9 (2014)
DOI 10.1371/journal.pone.0100538
Citations Scopus - 32Web of Science - 22
2014 Spencer AJ, Furze J, Honeycutt JD, Calvert A, Saurya S, Colloca S, et al., '4-1BBL Enhances CD8(+) T Cell Responses Induced by Vectored Vaccines in Mice but Fails to Improve Immunogenicity in Rhesus Macaques', PLOS ONE, 9 (2014)
DOI 10.1371/journal.pone.0105520
Citations Scopus - 7Web of Science - 6
2013 Warimwe GM, Lorenzo G, Lopez-Gil E, Reyes-Sandoval A, Cottingham MG, Spencer AJ, et al., 'Immunogenicity and efficacy of a chimpanzee adenovirus-vectored Rift Valley Fever vaccine in mice', VIROLOGY JOURNAL, 10 (2013)
DOI 10.1186/1743-422X-10-349
Citations Scopus - 51Web of Science - 44
2013 Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, Bliss CM, et al., 'Safety and Immunogenicity of Heterologous Prime-Boost Immunisation with Plasmodium falciparum Malaria Candidate Vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in Healthy Gambian and Kenyan Adults', PLOS ONE, 8 (2013)
DOI 10.1371/journal.pone.0057726
Citations Scopus - 62Web of Science - 51
2013 Sheehy SH, Spencer AJ, Douglas AD, Sim BKL, Longley RJ, Edwards NJ, et al., 'Optimising Controlled Human Malaria Infection Studies Using Cryopreserved P. falciparum Parasites Administered by Needle and Syringe', PLOS ONE, 8 (2013)
DOI 10.1371/journal.pone.0065960
Citations Scopus - 72Web of Science - 67
2013 Lambe T, Carey JB, Li Y, Spencer AJ, van Laarhoven A, Mullarkey CE, et al., 'Immunity Against Heterosubtypic Influenza Virus Induced By Adenovirus And MVA Expressing Nucleoprotein And Matrix Protein-1', SCIENTIFIC REPORTS, 3 (2013)
DOI 10.1038/srep01443
Citations Scopus - 64Web of Science - 63
2013 'Abstracts of the 3rd Annual Infection and Immunity Meeting, a meeting of the BSI Infection & Immunity Affinity Group. April 11, 2013. London, United Kingdom.', Clinical and experimental immunology, 173 Suppl 1 1-4 (2013)
DOI 10.1111/j.1365-2249.2013.12169.x
2012 Biswas S, Spencer AJ, Forbes EK, Gilbert SC, Holder AA, Hill AVS, Draper SJ, 'Recombinant Viral-Vectored Vaccines Expressing Plasmodium chabaudi AS Apical Membrane Antigen 1: Mechanisms of Vaccine-Induced Blood-Stage Protection', JOURNAL OF IMMUNOLOGY, 188 5041-5053 (2012)
DOI 10.4049/jimmunol.1101106
Citations Scopus - 19Web of Science - 17
2012 Lambe T, Spencer AJ, Mullarkey CE, Antrobus RD, Yu L-M, de Whalley P, et al., 'T-Cell Responses in Children to Internal Influenza Antigens, 1 Year After Immunization With Pandemic H1N1 Influenza Vaccine, and Response to Revaccination With Seasonal Trivalent-inactivated Influenza Vaccine', PEDIATRIC INFECTIOUS DISEASE JOURNAL, 31 E86-E91 (2012)
DOI 10.1097/INF.0b013e318255e443
Citations Scopus - 17Web of Science - 14
2012 Lillie PJ, Berthoud TK, Powell TJ, Lambe T, Mullarkey C, Spencer AJ, et al., 'Preliminary Assessment of the Efficacy of a T-Cell-Based Influenza Vaccine, MVA-NP+M1, in Humans', CLINICAL INFECTIOUS DISEASES, 55 19-25 (2012)
DOI 10.1093/cid/cis327
Citations Scopus - 217Web of Science - 197
2012 Sheehy SH, Duncan CJA, Elias SC, Biswas S, Collins KA, O'Hara GA, et al., 'Phase Ia Clinical Evaluation of the Safety and Immunogenicity of the Plasmodium falciparum Blood-Stage Antigen AMA1 in ChAd63 and MVA Vaccine Vectors', PLOS ONE, 7 (2012)
DOI 10.1371/journal.pone.0031208
Citations Scopus - 137Web of Science - 125
2012 Spencer AJ, Hill F, Honeycutt JD, Cottingham MG, Bregu M, Rollier CS, et al., 'Fusion of the Mycobacterium tuberculosis Antigen 85A to an Oligomerization Domain Enhances Its Immunogenicity in Both Mice and Non-Human Primates', PLOS ONE, 7 (2012)
DOI 10.1371/journal.pone.0033555
Citations Scopus - 43Web of Science - 39
2012 Dicks MDJ, Spencer AJ, Edwards NJ, Wadell G, Bojang K, Gilbert SC, et al., 'A Novel Chimpanzee Adenovirus Vector with Low Human Seroprevalence: Improved Systems for Vector Derivation and Comparative Immunogenicity', PLOS ONE, 7 (2012)
DOI 10.1371/journal.pone.0040385
Citations Scopus - 275Web of Science - 221
2012 Antrobus RD, Lillie PJ, Berthoud TK, Spencer AJ, McLaren JE, Ladell K, et al., 'A T Cell-Inducing Influenza Vaccine for the Elderly: Safety and Immunogenicity of MVA-NP+M1 in Adults Aged over 50 Years', PLOS ONE, 7 (2012)
DOI 10.1371/journal.pone.0048322
Citations Scopus - 103Web of Science - 96
2011 Sheehy SH, Duncan CJA, Elias SC, Collins KA, Ewer KJ, Spencer AJ, et al., 'Phase Ia Clinical Evaluation of the Plasmodium falciparum Blood-stage Antigen MSP1 in ChAd63 and MVA Vaccine Vectors', MOLECULAR THERAPY, 19 2269-2276 (2011)
DOI 10.1038/mt.2011.176
Citations Scopus - 142Web of Science - 122
2010 Wong SH, Vannberg FO, Spencer AJ, van der Weyden L, Hill AVS, Wyllie DH, 'Comment on "CRTAM Confers Late-Stage Activation of CD8(+) T Cells to Regulate Retention within Lymph Node"', JOURNAL OF IMMUNOLOGY, 184 4052-4053 (2010)
DOI 10.4049/jimmunol.1090017
Citations Scopus - 2Web of Science - 2
2010 Draper SJ, Biswas S, Spencer AJ, Remarque EJ, Capone S, Naddeo M, et al., 'Enhancing Blood-Stage Malaria Subunit Vaccine Immunogenicity in Rhesus Macaques by Combining Adenovirus, Poxvirus, and Protein-in-Adjuvant Vaccines', JOURNAL OF IMMUNOLOGY, 185 7583-7595 (2010)
DOI 10.4049/jimmunol.1001760
Citations Scopus - 74Web of Science - 70
2010 Alcock R, Cottingham MG, Rollier CS, Furze J, De Costa SD, Hanlon M, et al., 'Long-Term Thermostabilization of Live Poxviral and Adenoviral Vaccine Vectors at Supraphysiological Temperatures in Carbohydrate Glass', SCIENCE TRANSLATIONAL MEDICINE, 2 (2010)
DOI 10.1126/scitranslmed.3000490
Citations Scopus - 125Web of Science - 121
2009 Larsen KC, Spencer AJ, Goodman AL, Gilchrist A, Furze J, Rollier CS, et al., 'Expression of tak1 and tram induces synergistic pro-inflammatory signalling and adjuvants DNA vaccines', VACCINE, 27 5589-5598 (2009)
DOI 10.1016/j.vaccine.2009.07.025
Citations Scopus - 19Web of Science - 16
2008 Cottingham MG, Andersen RF, Spencer AJ, Saurya S, Furze J, Hill AVS, Gilbert SC, 'Recombination-Mediated Genetic Engineering of a Bacterial Artificial Chromosome Clone of Modified Vaccinia virus Ankara (MVA)', PLOS ONE, 3 (2008)
DOI 10.1371/journal.pone.0001638
Citations Scopus - 82Web of Science - 75
Show 73 more journal articles

Conference (10 outputs)

Year Citation Altmetrics Link
2019 Datoo M, Jenkin D, Ramos-Lopez F, Baker M, Flaxman A, Bellamy D, et al., 'PRIME-TARGET IMMUNISATION WITH LIVER-STAGE MALARIA VACCINES: A FIRST-IN-HUMAN CHALLENGE TRIAL', AMERICAN JOURNAL OF TROPICAL MEDICINE AND HYGIENE, National Harbor, MD (2019)
2019 Noe A, Bellamy D, Flaxman A, Datoo M, Jenkin D, Husainy A, et al., 'NOVEL METHODS TO DETERMINE LIVER-STAGE MALARIA VACCINE CORRELATES OF PROTECTION: KINETICS, DEEP IMMUNE PHENOTYPING AND TRANSCRIPTOMICS', AMERICAN JOURNAL OF TROPICAL MEDICINE AND HYGIENE, National Harbor, MD (2019)
2019 Flaxman A, Sebastian S, Cha K, Ulaszewska M, Purushotham J, Gilbride C, et al., 'An effective multi-pathogen vaccine targeting filoviruses and an arenavirus', EUROPEAN JOURNAL OF IMMUNOLOGY, Beijing, PEOPLES R CHINA (2019)
2018 Ewer K, Silman D, Bowyer G, Gola A, Bellamy D, Mair C, et al., '"PRIME-TARGET" IMMUNIZATION WITH VIRAL VECTORS FOR ENHANCED EFFICACY OF LIVER-STAGE MALARIA VACCINES', AMERICAN JOURNAL OF TROPICAL MEDICINE AND HYGIENE, New Orleans, LA (2018)
2017 Gola A, Walters AA, Uderhardt S, Salman AM, Halbroth BR, Khan SM, et al., 'Prime and Target Immunization Protects Against Liver-Stage Malaria', JOURNAL OF IMMUNOLOGY, Washington, DC (2017)
2014 Spencer AJ, Longley RJ, Gola A, Lambe T, Hill AV, 'Protection from liver-stage malaria is dependent on a fine balance between the number of infected hepatocytes and effector CD8(+) T-cells', IMMUNOLOGY (2014)
2014 Gola A, Sebastian S, Hill AV, Spencer AJ, 'The role of antigen persistence and its effect on the generation of memory subsets', IMMUNOLOGY (2014)
2013 Longley RJ, Ewer K, Cottingham MG, Bauza K, Williams A, Spencer AJ, Hill AVS, 'An in vitro T cell killing assay to assess new Plasmodium falciparum vaccine candidates', CLINICAL AND EXPERIMENTAL IMMUNOLOGY, London, ENGLAND (2013)
2013 Dicks M, Spencer A, Guzman E, Charleston B, Gilbert S, Hill A, Cottingham M, 'Uncovering the mechanisms of adenovirus vector immunogenicity', HUMAN GENE THERAPY, Univ London, Royal Holloway, Egham, ENGLAND (2013)
2011 Dicks MDJ, Spencer A, Edwards N, Gilbert S, Hill S, Cottingham MG, 'Improved systems for generating and evaluating adenovirus vaccine vectors reveal differences in immunogenicity between vectors of different adenoviral species', HUMAN GENE THERAPY (2011)
Show 7 more conferences

Preprint (7 outputs)

Year Citation Altmetrics Link
2023 Ni T, Mendonça L, Zhu Y, Howe A, Radecke J, Shah P, et al., 'ChAdOx1 COVID vaccines express RBD open prefusion SARS-CoV-2 spikes on the cell surface (2023)
DOI 10.1101/2023.05.22.541685
2021 Lambe T, Spencer A, Thomas K, Thomas S, White A, Humphries H, et al., 'ChAdOx1 nCoV-19 protection against SARS-CoV-2 in rhesus macaque and ferret challenge models (2021)
DOI 10.21203/rs.3.rs-133970/v1
2021 Spencer A, McKay P, Belij-Rammerstorfer S, Ulaszewska M, Bissett C, Hu K, et al., 'Heterologous vaccination regimens with self-amplifying RNA and Adenoviral COVID vaccines induce robust immune responses in mice (2021)
DOI 10.1101/2021.01.28.428665
2021 Spencer A, Morris S, Ulaszewska M, Powers C, Kailath R, Bissett C, et al., 'The ChAdOx1 vectored vaccine, AZD2816, induces strong immunogenicity against SARS-CoV-2 Beta (B.1.351) and other variants of concern in preclinical studies (2021)
DOI 10.1101/2021.06.08.447308
2020 Spencer A, Smith A, de St Groth BF, 'Antigen-specific competitive inhibition of CD4
DOI 10.1101/2020.08.31.276527
2020 Graham S, McLean R, Spencer A, Belij-Rammerstorfer S, Wright D, Ulaszewska M, et al., 'Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19 (2020)
DOI 10.1101/2020.06.20.159715
2020 van Doremalen N, Lambe T, Spencer A, Belij-Rammerstorfer S, Purushotham JN, Port JR, et al., 'ChAdOx1 nCoV-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques. (2020)
DOI 10.1101/2020.05.13.093195
Show 4 more preprints
Edit

Research Supervision

Number of supervisions

Completed11
Current3

Current Supervision

Commenced Level of Study Research Title Program Supervisor Type
2023 PhD TLR2 Agonist Boosting Immunogenicity of Tumour-Targeting RNAi-Based Nanomedicines PhD (Immunology & Microbiol), College of Health, Medicine and Wellbeing, The University of Newcastle Co-Supervisor
2021 PhD Identification of conserved antigenic regions within influenza viruses as potential universal vaccine antigens Medical Science, The University of Oxford Co-Supervisor
2020 PhD Exploring vaccine regimens against respiratory viruses Medical Science, The University of Oxford Co-Supervisor

Past Supervision

Year Level of Study Research Title Program Supervisor Type
2023 PhD Development of a Durable and Efficacious Pre-Erythrocytic Malaria Vaccine Medical Science, The University of Oxford Principal Supervisor
2021 Masters A CRISPR-Cas9 screen for hepatocyte receptors for malaria parasite invasion Medical Science, The University of Oxford Co-Supervisor
2021 PhD Evaluating New Malaria Vaccine Candidates Medical Science, The University of Oxford Principal Supervisor
2019 PhD Vaccine design for global health pathogens Medical Science, The University of Oxford Co-Supervisor
2018 PhD The role of antigen in the maintenance and localisation of CD8+ T-cells in the context of liver stage malaria Medical Science, The University of Oxford Principal Supervisor
2017 PhD Immune Responses to vaccines against malaria Medical Science, The University of Oxford Co-Supervisor
2016 PhD Development of Adjuvanted Multi-Antigen Liver-Stage Malaria vaccines Medical Science, The University of Oxford Principal Supervisor
2016 Masters An in vitro assay to measure antibody-mediated inhibition of malaria sporozoite infection Medical Science, London School of Hygiene and Tropical Medicine Principal Supervisor
2015 PhD Assessment of Novel Liver-Stage Vaccines using Transgenic Rodent Malaria Parasites Medical Science, The University of Oxford Co-Supervisor
2014 PhD Liver-stage vaccines for malaria Medical Science, The University of Oxford Co-Supervisor
2012 PhD Novel Adenovirus Vaccine Vectors Medical Science, The University of Oxford Co-Supervisor
Edit

Research Collaborations

The map is a representation of a researchers co-authorship with collaborators across the globe. The map displays the number of publications against a country, where there is at least one co-author based in that country. Data is sourced from the University of Newcastle research publication management system (NURO) and may not fully represent the authors complete body of work.

Country Count of Publications
United Kingdom 85
United States 16
Australia 15
Italy 10
Netherlands 9
More...
Edit

Dr Alex Spencer

Position

Immunology Lecturer
Vaccine and Infection Immunology Group
School of Biomedical Sciences and Pharmacy
College of Health, Medicine and Wellbeing

Contact Details

Email alex.spencer@newcastle.edu.au
Phone (02) 4042 0634
Mobile 0405281741
Link Twitter

Office

Room HMRI 2409
Building Hunter Medical Research Institute
Location HMRI

,
Edit