1
|
Migueles SA, Nettere DM, Gavil NV, Wang LT, Toulmin SA, Kelly EP, Ward AJ, Lin S, Thompson SA, Peterson BA, Abdeen CS, Sclafani CR, Pryal PF, Leach BG, Ludwig AK, Rogan DC, Przygonska PA, Cattani A, Imamichi H, Sachs A, Cafri G, Huang NN, Patamawenu A, Liang CJ, Hallahan CW, Kambach DM, Han EX, Coupet T, Chen J, Moir SL, Chun TW, Coates EE, Ledgerwood J, Schmidt J, Taillandier-Coindard M, Michaux J, Pak H, Bassani-Sternberg M, Frahm N, McElrath MJ, Connors M. HIV vaccines induce CD8 + T cells with low antigen receptor sensitivity. Science 2023; 382:1270-1276. [PMID: 38096385 DOI: 10.1126/science.adg0514] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 11/03/2023] [Indexed: 12/18/2023]
Abstract
Current HIV vaccines designed to stimulate CD8+ T cells have failed to induce immunologic control upon infection. The functions of vaccine-induced HIV-specific CD8+ T cells were investigated here in detail. Cytotoxic capacity was significantly lower than in HIV controllers and was not a consequence of low frequency or unaccumulated functional cytotoxic proteins. Low cytotoxic capacity was attributable to impaired degranulation in response to the low antigen levels present on HIV-infected targets. The vaccine-induced T cell receptor (TCR) repertoire was polyclonal and transduction of these TCRs conferred the same reduced functions. These results define a mechanism accounting for poor antiviral activity induced by these vaccines and suggest that an effective CD8+ T cell response may require a vaccination strategy that drives further TCR clonal selection.
Collapse
Affiliation(s)
- Stephen A Migueles
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Danielle M Nettere
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Noah V Gavil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence T Wang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sushila A Toulmin
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth P Kelly
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Addison J Ward
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Siying Lin
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah A Thompson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bennett A Peterson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cassidy S Abdeen
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Carina R Sclafani
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick F Pryal
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin G Leach
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amanda K Ludwig
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel C Rogan
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paulina A Przygonska
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angela Cattani
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hiromi Imamichi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Abraham Sachs
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gal Cafri
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ning-Na Huang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andy Patamawenu
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - C Jason Liang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Claire W Hallahan
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | - Susan L Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emily E Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julien Schmidt
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Marie Taillandier-Coindard
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Justine Michaux
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - HuiSong Pak
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nicole Frahm
- Vaccine and Infectious Disease Division and the HIV Vaccine Trials Network, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division and the HIV Vaccine Trials Network, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mark Connors
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Wijesundara DK, Yeow A, McMillan CL, Choo JJ, Todorovic A, Mekonnen ZA, Masavuli MG, Young PR, Gowans EJ, Grubor-Bauk B, Muller DA. Superior efficacy of a skin-applied microprojection device for delivering a novel Zika DNA vaccine. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102056. [PMID: 38028199 PMCID: PMC10630652 DOI: 10.1016/j.omtn.2023.102056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Zika virus (ZIKV) infections are spreading silently with limited global surveillance in at least 89 countries and territories. There is a pressing need to develop an effective vaccine suitable for equitable distribution globally. Consequently, we previously developed a proprietary DNA vaccine encoding secreted non-structural protein 1 of ZIKV (pVAX-tpaNS1) to elicit rapid protection in a T cell-dependent manner in mice. In the current study, we evaluated the stability, efficacy, and immunogenicity of delivering this DNA vaccine into the skin using a clinically effective and proprietary high-density microarray patch (HD-MAP). Dry-coating of pVAX-tpaNS1 on the HD-MAP device resulted in no loss of vaccine stability at 40°C storage over the course of 28 days. Vaccination of mice (BALB/c) with the HD-MAP-coated pVAX-tpaNS1 elicited a robust anti-NS1 IgG response in both the cervicovaginal mucosa and systemically and afforded protection against live ZIKV challenge. Furthermore, the vaccination elicited a significantly higher magnitude and broader NS1-specific T helper and cytotoxic T cell response in vivo compared with traditional needle and syringe intradermal vaccination. Overall, the study highlights distinctive immunological advantages coupled with an excellent thermostability profile of using the HD-MAP device to deliver a novel ZIKV DNA vaccine.
Collapse
Affiliation(s)
- Danushka K. Wijesundara
- Vaxxas Biomedical Facility, Hamilton, QLD 4007, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Arthur Yeow
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - Christopher L.D. McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jovin J.Y. Choo
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Aleksandra Todorovic
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zelalem A. Mekonnen
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - Makutiro G. Masavuli
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - Paul R. Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Eric J. Gowans
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - Branka Grubor-Bauk
- Discipline of Surgery, The University of Adelaide, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - David A. Muller
- Vaxxas Biomedical Facility, Hamilton, QLD 4007, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
3
|
Simon Davis DA, Ritchie M, Hammill D, Garrett J, Slater RO, Otoo N, Orlov A, Gosling K, Price J, Yip D, Jung K, Syed FM, Atmosukarto II, Quah BJC. Identifying cancer-associated leukocyte profiles using high-resolution flow cytometry screening and machine learning. Front Immunol 2023; 14:1211064. [PMID: 37600768 PMCID: PMC10435879 DOI: 10.3389/fimmu.2023.1211064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
Background Machine learning (ML) is a valuable tool with the potential to aid clinical decision making. Adoption of ML to this end requires data that reliably correlates with the clinical outcome of interest; the advantage of ML is that it can model these correlations from complex multiparameter data sets that can be difficult to interpret conventionally. While currently available clinical data can be used in ML for this purpose, there exists the potential to discover new "biomarkers" that will enhance the effectiveness of ML in clinical decision making. Since the interaction of the immune system and cancer is a hallmark of tumor establishment and progression, one potential area for cancer biomarker discovery is through the investigation of cancer-related immune cell signatures. Hence, we hypothesize that blood immune cell signatures can act as a biomarker for cancer progression. Methods To probe this, we have developed and tested a multiparameter cell-surface marker screening pipeline, using flow cytometry to obtain high-resolution systemic leukocyte population profiles that correlate with detection and characterization of several cancers in murine syngeneic tumor models. Results We discovered a signature of several blood leukocyte subsets, the most notable of which were monocyte subsets, that could be used to train CATboost ML models to predict the presence and type of cancer present in the animals. Conclusions Our findings highlight the potential utility of a screening approach to identify robust leukocyte biomarkers for cancer detection and characterization. This pipeline can easily be adapted to screen for cancer specific leukocyte markers from the blood of cancer patient.
Collapse
Affiliation(s)
- David A. Simon Davis
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
| | - Melissa Ritchie
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
| | - Dillon Hammill
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jessica Garrett
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Robert O. Slater
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Naomi Otoo
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Anna Orlov
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Katharine Gosling
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
| | - Jason Price
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Desmond Yip
- Australian National University, Canberra, ACT, Australia
- Department of Medical Oncology, Canberra Hospital & Health Services, Canberra, ACT, Australia
| | - Kylie Jung
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
- Radiation Oncology Department, Canberra Hospital & Health Services, Canberra, ACT, Australia
| | - Farhan M. Syed
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
- Radiation Oncology Department, Canberra Hospital & Health Services, Canberra, ACT, Australia
| | - Ines I. Atmosukarto
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
- Division of Genome Sciences & Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Ben J. C. Quah
- Irradiation Immunity Interaction Lab, Australian National University, Canberra, ACT, Australia
- Radiation Oncology Department, Canberra Hospital & Health Services, Canberra, ACT, Australia
| |
Collapse
|
4
|
Li Z, Roy S, Ranasinghe C. IL-13Rα2 Regulates the IL-13/IFN-γ Balance during Innate Lymphoid Cell and Dendritic Cell Responses to Pox Viral Vector-Based Vaccination. Vaccines (Basel) 2021; 9:440. [PMID: 34062727 PMCID: PMC8147251 DOI: 10.3390/vaccines9050440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/08/2021] [Accepted: 04/23/2021] [Indexed: 12/02/2022] Open
Abstract
We have shown that manipulation of IL-13 and STAT6 signaling at the vaccination site can lead to different innate lymphoid cell (ILC)/dendritic cell (DC) recruitment, resulting in high avidity/poly-functional T cells and effective antibody differentiation. Here we show that permanent versus transient blockage of IL-13 and STAT6 at the vaccination site can lead to unique ILC-derived IL-13 and IFN-γ profiles, and differential IL-13Rα2, type I and II IL-4 receptor regulation on ILC. Specifically, STAT6-/- BALB/c mice given fowl pox virus (FPV) expressing HIV antigens induced elevated ST2/IL-33R+ ILC2-derived IL-13 and reduced NKp46+/- ILC1/ILC3-derived IFN-γ expression, whilst the opposite (reduced IL-13 and elevated IFN-γ expression) was observed during transient inhibition of STAT6 signaling in wild type BALB/c mice given FPV-HIV-IL-4R antagonist vaccination. Interestingly, disruption/inhibition of STAT6 signaling considerably impacted IL-13Rα2 expression by ST2/IL-33R+ ILC2 and NKp46- ILC1/ILC3, unlike direct IL-13 inhibition. Consistently with our previous findings, this further indicated that inhibition of STAT6 most likely promoted IL-13 regulation via IL-13Rα2. Moreover, the elevated ST2/IL-33R+ IL-13Rα2+ lung ILC2, 24 h post FPV-HIV-IL-4R antagonist vaccination was also suggestive of an autocrine regulation of ILC2-derived IL-13 and IL-13Rα2, under certain conditions. Knowing that IL-13 can modulate IFN-γ expression, the elevated expression of IFN-γR on lung ST2/IL-33R+ ILC2 provoked the notion that there could also be inter-regulation of lung ILC2-derived IL-13 and NKp46- ILC1/ILC3-derived IFN-γ via their respective receptors (IFN-γR and IL-13Rα2) at the lung mucosae early stages of vaccination. Intriguingly, under different IL-13 conditions differential regulation of IL-13/IL-13Rα2 on lung DC was also observed. Collectively these findings further substantiated that IL-13 is the master regulator of, not only DC, but also different ILC subsets at early stages of viral vector vaccination, and responsible for shaping the downstream adaptive immune outcomes. Thus, thoughtful selection of vaccine strategies/adjuvants that can manipulate IL-13Rα2, and STAT6 signaling at the ILC/DC level may prove useful in designing more efficacious vaccines against different/chronic pathogens.
Collapse
Affiliation(s)
- Zheyi Li
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; (Z.L.); (S.R.)
| | - Sreeja Roy
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; (Z.L.); (S.R.)
- Department of Immunology & Microbial Disease, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208-3479, USA
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; (Z.L.); (S.R.)
| |
Collapse
|
5
|
Watterson D, Wijesundara DK, Modhiran N, Mordant FL, Li Z, Avumegah MS, McMillan CL, Lackenby J, Guilfoyle K, van Amerongen G, Stittelaar K, Cheung ST, Bibby S, Daleris M, Hoger K, Gillard M, Radunz E, Jones ML, Hughes K, Hughes B, Goh J, Edwards D, Scoble J, Pearce L, Kowalczyk L, Phan T, La M, Lu L, Pham T, Zhou Q, Brockman DA, Morgan SJ, Lau C, Tran MH, Tapley P, Villalón-Letelier F, Barnes J, Young A, Jaberolansar N, Scott CA, Isaacs A, Amarilla AA, Khromykh AA, van den Brand JM, Reading PC, Ranasinghe C, Subbarao K, Munro TP, Young PR, Chappell KJ. Preclinical development of a molecular clamp-stabilised subunit vaccine for severe acute respiratory syndrome coronavirus 2. Clin Transl Immunology 2021; 10:e1269. [PMID: 33841880 PMCID: PMC8021130 DOI: 10.1002/cti2.1269] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Objectives Efforts to develop and deploy effective vaccines against severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) continue at pace. Here, we describe rational antigen design through to manufacturability and vaccine efficacy of a prefusion‐stabilised spike (S) protein, Sclamp, in combination with the licensed adjuvant MF59 ‘MF59C.1’ (Seqirus, Parkville, Australia). Methods A panel recombinant Sclamp proteins were produced in Chinese hamster ovary and screened in vitro to select a lead vaccine candidate. The structure of this antigen was determined by cryo‐electron microscopy and assessed in mouse immunogenicity studies, hamster challenge studies and safety and toxicology studies in rat. Results In mice, the Sclamp vaccine elicits high levels of neutralising antibodies, as well as broadly reactive and polyfunctional S‐specific CD4+ and cytotoxic CD8+ T cells in vivo. In the Syrian hamster challenge model (n = 70), vaccination results in reduced viral load within the lung, protection from pulmonary disease and decreased viral shedding in daily throat swabs which correlated strongly with the neutralising antibody level. Conclusion The SARS‐CoV‐2 Sclamp vaccine candidate is compatible with large‐scale commercial manufacture, stable at 2–8°C. When formulated with MF59 adjuvant, it elicits neutralising antibodies and T‐cell responses and provides protection in animal challenge models.
Collapse
Affiliation(s)
- Daniel Watterson
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia.,Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence The University of Queensland Brisbane QLD Australia
| | - Danushka K Wijesundara
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Zheyi Li
- Department of Immunology and Infectious Disease The John Curtin School of Medical Research, The Australian National University Canberra ACT Australia
| | - Michael S Avumegah
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Christopher Ld McMillan
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Julia Lackenby
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | | | | | | | - Stacey Tm Cheung
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia
| | - Summa Bibby
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia
| | - Mallory Daleris
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Kym Hoger
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Marianne Gillard
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Eve Radunz
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Martina L Jones
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Karen Hughes
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Ben Hughes
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Justin Goh
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - David Edwards
- The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | | | | | | | - Tram Phan
- CSIRO Manufacturing Parkville VIC Australia
| | - Mylinh La
- CSIRO Manufacturing Parkville VIC Australia
| | - Louis Lu
- CSIRO Manufacturing Parkville VIC Australia
| | - Tam Pham
- CSIRO Manufacturing Parkville VIC Australia
| | - Qi Zhou
- CSIRO Manufacturing Parkville VIC Australia
| | | | | | - Cora Lau
- University of Queensland Biological Resources The University of Queensland St Lucia QLD Australia
| | - Mai H Tran
- TetraQ The University of Queensland St Lucia QLD Australia
| | - Peter Tapley
- TetraQ The University of Queensland St Lucia QLD Australia
| | - Fernando Villalón-Letelier
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - James Barnes
- WHO Collaborating Centre for Reference and Research on Influenza Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Andrew Young
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Noushin Jaberolansar
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Connor Ap Scott
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia
| | - Ariel Isaacs
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence The University of Queensland Brisbane QLD Australia
| | - Judith Ma van den Brand
- Division of Pathology Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Patrick C Reading
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia.,WHO Collaborating Centre for Reference and Research on Influenza Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Charani Ranasinghe
- Department of Immunology and Infectious Disease The John Curtin School of Medical Research, The Australian National University Canberra ACT Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia.,WHO Collaborating Centre for Reference and Research on Influenza Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Trent P Munro
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia.,Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence The University of Queensland Brisbane QLD Australia
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences The University of Queensland St Lucia QLD Australia.,The Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD Australia.,Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence The University of Queensland Brisbane QLD Australia
| |
Collapse
|
6
|
Li Z, Khanna M, Grimley SL, Ellenberg P, Gonelli CA, Lee WS, Amarasena TH, Kelleher AD, Purcell DFJ, Kent SJ, Ranasinghe C. Mucosal IL-4R antagonist HIV vaccination with SOSIP-gp140 booster can induce high-quality cytotoxic CD4 +/CD8 + T cells and humoral responses in macaques. Sci Rep 2020; 10:22077. [PMID: 33328567 PMCID: PMC7744512 DOI: 10.1038/s41598-020-79172-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/02/2020] [Indexed: 11/09/2022] Open
Abstract
Inducing humoral, cellular and mucosal immunity is likely to improve the effectiveness of HIV-1 vaccine strategies. Here, we tested a vaccine regimen in pigtail macaques using an intranasal (i.n.) recombinant Fowl Pox Virus (FPV)-gag pol env-IL-4R antagonist prime, intramuscular (i.m.) recombinant Modified Vaccinia Ankara Virus (MVA)-gag pol-IL-4R antagonist boost followed by an i.m SOSIP-gp140 boost. The viral vector-expressed IL-4R antagonist transiently inhibited IL-4/IL-13 signalling at the vaccination site. The SOSIP booster not only induced gp140-specific IgG, ADCC (antibody-dependent cellular cytotoxicity) and some neutralisation activity, but also bolstered the HIV-specific cellular and humoral responses. Specifically, superior sustained systemic and mucosal HIV Gag-specific poly-functional/cytotoxic CD4+ and CD8+ T cells were detected with the IL-4R antagonist adjuvanted strategy compared to the unadjuvanted control. In the systemic compartment elevated Granzyme K expression was linked to CD4+ T cells, whilst Granzyme B/TIA-1 to CD8+ T cells. In contrast, the cytotoxic marker expression by mucosal CD4+ and CD8+ T cells differed according to the mucosal compartment. This vector-based mucosal IL-4R antagonist/SOSIP booster strategy, which promotes cytotoxic mucosal CD4+ T cells at the first line of defence, and cytotoxic CD4+ and CD8+ T cells plus functional antibodies in the blood, may prove valuable in combating mucosal infection with HIV-1 and warrants further investigation.
Collapse
Affiliation(s)
- Z Li
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - M Khanna
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - S L Grimley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - P Ellenberg
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - C A Gonelli
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - T H Amarasena
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - A D Kelleher
- Immunovirology and Pathogenesis Program, Kirby Institute, University of New South Wales, Sydney, NSW, 2052, Australia
| | - D F J Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - S J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - C Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
7
|
Unique IL-13Rα2/STAT3 mediated IL-13 regulation detected in lung conventional dendritic cells, 24 h post viral vector vaccination. Sci Rep 2020; 10:1017. [PMID: 31974500 PMCID: PMC6978450 DOI: 10.1038/s41598-020-57815-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022] Open
Abstract
This study demonstrates that 24 h following viral vector-based vaccination IL-13Rα2 functions as a master sensor on conventional dendritic cells (cDCs), abetted by high protein stability coupled with minimal mRNA expression, to rapidly regulate DC mediated IL-13 responses at the lung mucosae, unlike IL-13Rα1. Under low IL-13, IL-13Rα2 performs as a primary signalling receptor, whilst under high IL-13, acts to sequester IL-13 to maintain homeostasis, both in a STAT3-dependent manner. Likewise, we show that viral vector-derived IL-13 levels at the vaccination site can induce differential STAT3/STAT6 paradigms in lung cDC, that can get regulated collaboratively or independently by TGF-β1 and IFN-γ. Specifically, low IL-13 responses associated with recombinant Fowlpox virus (rFPV) is regulated by early IL-13Rα2, correlated with STAT3/TGF-β1 expression. Whilst, high IL-13 responses, associated with recombinant Modified Vaccinia Ankara (rMVA) is regulated in an IL-13Rα1/STAT6 dependent manner associated with IFN-γR expression bias. Different viral vaccine vectors have previously been shown to induce unique adaptive immune outcomes. Taken together current observations suggest that IL-13Rα2-driven STAT3/STAT6 equilibrium at the cDC level may play an important role in governing the efficacy of vector-based vaccines. These new insights have high potential to be exploited to improve recombinant viral vector-based vaccine design, according to the pathogen of interest and/or therapies against IL-13 associated disease conditions.
Collapse
|
8
|
Grubor-Bauk B, Wijesundara DK, Masavuli M, Abbink P, Peterson RL, Prow NA, Larocca RA, Mekonnen ZA, Shrestha A, Eyre NS, Beard MR, Gummow J, Carr J, Robertson SA, Hayball JD, Barouch DH, Gowans EJ. NS1 DNA vaccination protects against Zika infection through T cell-mediated immunity in immunocompetent mice. SCIENCE ADVANCES 2019; 5:eaax2388. [PMID: 31844662 PMCID: PMC6905874 DOI: 10.1126/sciadv.aax2388] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/08/2019] [Indexed: 05/08/2023]
Abstract
The causal association of Zika virus (ZIKV) with microcephaly, congenital malformations in infants, and Guillain-Barré syndrome in adults highlights the need for effective vaccines. Thus far, efforts to develop ZIKV vaccines have focused on the viral envelope. ZIKV NS1 as a vaccine immunogen has not been fully explored, although it can circumvent the risk of antibody-dependent enhancement of ZIKV infection, associated with envelope antibodies. Here, we describe a novel DNA vaccine encoding a secreted ZIKV NS1, that confers rapid protection from systemic ZIKV infection in immunocompetent mice. We identify novel NS1 T cell epitopes in vivo and show that functional NS1-specific T cell responses are critical for protection against ZIKV infection. We demonstrate that vaccine-induced anti-NS1 antibodies fail to confer protection in the absence of a functional T cell response. This highlights the importance of using NS1 as a target for T cell-based ZIKV vaccines.
Collapse
Affiliation(s)
- B. Grubor-Bauk
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
- Corresponding author.
| | - D. K. Wijesundara
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - M. Masavuli
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - P. Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - R. L. Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - N. A. Prow
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - R. A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Z. A. Mekonnen
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - A. Shrestha
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - N. S. Eyre
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - M. R. Beard
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Gummow
- Gene Silencing and Expression Core Facility, Adelaide Health and Medical Sciences, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Carr
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - S. A. Robertson
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. D. Hayball
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - D. H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - E. J. Gowans
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| |
Collapse
|
9
|
Mekonnen ZA, Grubor-Bauk B, English K, Leung P, Masavuli MG, Shrestha AC, Bertolino P, Bowen DG, Lloyd AR, Gowans EJ, Wijesundara DK. Single-Dose Vaccination with a Hepatotropic Adeno-associated Virus Efficiently Localizes T Cell Immunity in the Liver with the Potential To Confer Rapid Protection against Hepatitis C Virus. J Virol 2019; 93:e00202-19. [PMID: 31292249 PMCID: PMC6744243 DOI: 10.1128/jvi.00202-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Hepatitis C virus (HCV) is a significant contributor to the global disease burden, and development of an effective vaccine is required to eliminate HCV infections worldwide. CD4+ and CD8+ T cell immunity correlates with viral clearance in primary HCV infection, and intrahepatic CD8+ tissue-resident memory T (TRM) cells provide lifelong and rapid protection against hepatotropic pathogens. Consequently, we aimed to develop a vaccine to elicit HCV-specific CD4+ and CD8+ T cells, including CD8+ TRM cells, in the liver, given that HCV primarily infects hepatocytes. To achieve this, we vaccinated wild-type BALB/c mice with a highly immunogenic cytolytic DNA vaccine encoding a model HCV (genotype 3a) nonstructural protein (NS5B) and a mutant perforin (pVAX-NS5B-PRF), as well as a recombinant adeno-associated virus (AAV) encoding NS5B (rAAV-NS5B). A novel fluorescent target array (FTA) was used to map immunodominant CD4+ T helper (TH) cell and cytotoxic CD8+ T cell epitopes of NS5B in vivo, which were subsequently used to design a KdNS5B451-459 tetramer and analyze NS5B-specific T cell responses in vaccinated mice in vivo The data showed that intradermal prime/boost vaccination with pVAX-NS5B-PRF was effective in eliciting TH and cytotoxic CD8+ T cell responses and intrahepatic CD8+ TRM cells, but a single intravenous dose of hepatotropic rAAV-NS5B was significantly more effective. As a T-cell-based vaccine against HCV should ideally result in localized T cell responses in the liver, this study describes primary observations in the context of HCV vaccination that can be used to achieve this goal.IMPORTANCE There are currently at least 71 million individuals with chronic HCV worldwide and almost two million new infections annually. Although the advent of direct-acting antivirals (DAAs) offers highly effective therapy, considerable remaining challenges argue against reliance on DAAs for HCV elimination, including high drug cost, poorly developed health infrastructure, low screening rates, and significant reinfection rates. Accordingly, development of an effective vaccine is crucial to HCV elimination. An HCV vaccine that elicits T cell immunity in the liver will be highly protective for the following reasons: (i) T cell responses against nonstructural proteins of the virus are associated with clearance of primary infection, and (ii) long-lived liver-resident T cells alone can protect against malaria infection of hepatocytes. Thus, in this study we exploit promising vaccination platforms to highlight strategies that can be used to evoke highly functional and long-lived T cell responses in the liver for protection against HCV.
Collapse
Affiliation(s)
- Zelalem A Mekonnen
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Branka Grubor-Bauk
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Kieran English
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - Preston Leung
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Makutiro G Masavuli
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Ashish C Shrestha
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Patrick Bertolino
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - David G Bowen
- Liver Immunology Group and A. W. Morrow Gastroenterology and Liver Centre, Centenary Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
- Collaborative Transplantation Research Group, Bosch Institute, Royal Prince Alfred Hospital and University of Sydney, Newtown, NSW, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Eric J Gowans
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| | - Danushka K Wijesundara
- Virology Laboratory, Basil Hetzel Institute for Translational Health Research, Discipline of Surgery, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
10
|
Khanna M, Jackson RJ, Alcantara S, Amarasena TH, Li Z, Kelleher AD, Kent SJ, Ranasinghe C. Mucosal and systemic SIV-specific cytotoxic CD4 + T cell hierarchy in protection following intranasal/intramuscular recombinant pox-viral vaccination of pigtail macaques. Sci Rep 2019; 9:5661. [PMID: 30952887 PMCID: PMC6450945 DOI: 10.1038/s41598-019-41506-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/11/2019] [Indexed: 11/09/2022] Open
Abstract
A HIV vaccine that provides mucosal immunity is urgently needed. We evaluated an intranasal recombinant Fowlpox virus (rFPV) priming vaccine followed by intramuscular Modified Vaccinia Ankara (rMVA) booster vaccine, both expressing SIV antigens. The vaccination generated mucosal and systemic SIV-specific CD4+ T cell mediated immunity and was associated with partial protection against high-dose intrarectal SIVmac251 challenge in outbred pigtail macaques. Three of 12 vaccinees were completely protected and these animals elicited sustained Gag-specific poly-functional, cytotoxic mucosal CD4+ T cells, complemented by systemic poly-functional CD4+ and CD8+ T cell immunity. Humoral immune responses, albeit absent in completely protected macaques, were associated with partial control of viremia in animals with relatively weaker mucosal/systemic T cell responses. Co-expression of an IL-4R antagonist by the rFPV vaccine further enhanced the breadth and cytotoxicity/poly-functionality of mucosal vaccine-specific CD4+ T cells. Moreover, a single FPV-gag/pol/env prime was able to induce rapid anamnestic gp140 antibody response upon SIV encounter. Collectively, our data indicated that nasal vaccination was effective at inducing robust cervico-vaginal and rectal immunity, although cytotoxic CD4+ T cell mediated mucosal and systemic immunity correlated strongly with 'complete protection', the different degrees of protection observed was multi-factorial.
Collapse
Affiliation(s)
- Mayank Khanna
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, 2601, Australia
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, 2601, Australia
| | - Sheilajen Alcantara
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Thakshila H Amarasena
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Zheyi Li
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, 2601, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, Kirby Institute, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, 2601, Australia.
| |
Collapse
|
11
|
Roy S, Jaeson MI, Li Z, Mahboob S, Jackson RJ, Grubor-Bauk B, Wijesundara DK, Gowans EJ, Ranasinghe C. Viral vector and route of administration determine the ILC and DC profiles responsible for downstream vaccine-specific immune outcomes. Vaccine 2019; 37:1266-1276. [PMID: 30733092 DOI: 10.1016/j.vaccine.2019.01.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/08/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022]
Abstract
This study demonstrates that route and viral vector can significantly influence the innate lymphoid cells (ILC) and dendritic cells (DC) recruited to the vaccination site, 24 h post delivery. Intranasal (i.n.) vaccination induced ST2/IL-33R+ ILC2, whilst intramuscular (i.m.) induced IL-25R+ and TSLPR+ (Thymic stromal lymphopoietin protein receptor) ILC2 subsets. However, in muscle a novel ILC subset devoid of the known ILC2 markers (IL-25R- IL-33R- TSLPR-) were found to express IL-13, unlike in lung. Different viral vectors also influenced the ILC-derived cytokines and the DC profiles at the respective vaccination sites. Both i.n. and i.m. recombinant fowlpox virus (rFPV) priming, which has been associated with induction of high avidity T cells and effective antibody differentiation exhibited low ILC2-derived IL-13, high NKp46+ ILC1/ILC3 derived IFN-γ and low IL-17A, together with enhanced CD11b+ CD103- conventional DCs (cDC). In contrast, recombinant Modified Vaccinia Ankara (rMVA) and Influenza A vector priming, which has been linked to low avidity T cells, induced opposing ILC derived-cytokine profiles and enhanced cross-presenting DCs. These observations suggested that the former ILC/DC profiles could be a predictor of a balanced cellular and humoral immune outcome. In addition, following i.n. delivery Rhinovirus (RV) and Adenovius type 5 (Ad5) vectors that induced elevated ILC2-derived IL-13, NKp46+ ILC1/ILC3-derived-IFN-γ and no IL-17A, predominantly recruited CD11b- B220+ plasmacytoid DCs (pDC). Knowing that pDC are involved in antibody differentiation, we postulate that i.n. priming with these vectors may favour induction of effective humoral immunity. Our data also revealed that vector-specific replication status and/or presence or absence of immune evasive genes can significantly alter the ILC and DC activity. Collectively, our findings suggest that understanding the route- and vector-specific ILC and DC profiles at the vaccination site may help tailor/design more efficacious viral vector-based vaccines, according to the pathogen of interest.
Collapse
Affiliation(s)
- S Roy
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - M I Jaeson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Z Li
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - S Mahboob
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - R J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - B Grubor-Bauk
- Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - D K Wijesundara
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia; Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - E J Gowans
- Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - C Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia.
| |
Collapse
|
12
|
Induction of Genotype Cross-Reactive, Hepatitis C Virus-Specific, Cell-Mediated Immunity in DNA-Vaccinated Mice. J Virol 2018; 92:JVI.02133-17. [PMID: 29437963 DOI: 10.1128/jvi.02133-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/11/2018] [Indexed: 12/24/2022] Open
Abstract
A universal hepatitis C virus (HCV) vaccine should elicit multiantigenic, multigenotypic responses, which are more likely to protect against challenge with the range of genotypes and subtypes circulating in the community. A vaccine cocktail and vaccines encoding consensus HCV sequences are attractive approaches to achieve this goal. Consequently, in a series of mouse vaccination studies, we compared the immunogenicity of a DNA vaccine encoding a consensus HCV nonstructural 5B (NS5B) protein to that of a cocktail of DNA plasmids encoding the genotype 1b (Gt1b) and Gt3a NS5B proteins. To complement this study, we assessed responses to a multiantigenic cocktail regimen by comparing a DNA vaccine cocktail encoding Gt1b and Gt3a NS3, NS4, and NS5B proteins to a single-genotype NS3/4/5B DNA vaccine. To thoroughly evaluate in vivo cytotoxic T lymphocyte (CTL) and T helper (Th) cell responses against Gt1b and Gt3a HCV peptide-pulsed target cells, we exploited a novel fluorescent-target array (FTA). FTA and enzyme-linked immunosorbent spot (ELISpot) analyses collectively indicated that the cocktail regimens elicited higher responses to Gt1b and Gt3a NS5B proteins than those with the consensus vaccine, while the multiantigenic DNA cocktail significantly increased the responses to NS3 and NS5B compared to those elicited by the single-genotype vaccines. Thus, a DNA cocktail vaccination regimen is more effective than a consensus vaccine or a monovalent vaccine at increasing the breadth of multigenotypic T cell responses, which has implications for the development of vaccines for communities where multiple HCV genotypes circulate.IMPORTANCE Despite the development of highly effective direct-acting antivirals (DAA), infections with hepatitis C virus (HCV) continue, particularly in countries where the supply of DAA is limited. Furthermore, patients who eliminate the virus as a result of DAA therapy can still be reinfected. Thus, a vaccine for HCV is urgently required, but the heterogeneity of HCV strains makes the development of a universal vaccine difficult. To address this, we developed a novel cytolytic DNA vaccine which elicits robust cell-mediated immunity (CMI) to the nonstructural (NS) proteins in vaccinated animals. We compared the immune responses against genotypes 1 and 3 that were elicited by a consensus DNA vaccine or a DNA vaccine cocktail and showed that the cocktail induced higher levels of CMI to the NS proteins of both genotypes. This study suggests that a universal HCV vaccine can most readily be achieved by use of a DNA vaccine cocktail.
Collapse
|
13
|
Townsend DG, Trivedi S, Jackson RJ, Ranasinghe C. Recombinant fowlpox virus vector-based vaccines: expression kinetics, dissemination and safety profile following intranasal delivery. J Gen Virol 2017; 98:496-505. [PMID: 28056224 PMCID: PMC5797952 DOI: 10.1099/jgv.0.000702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/03/2017] [Indexed: 12/23/2022] Open
Abstract
We have previously established that mucosal uptake of recombinant fowlpox virus (rFPV) vaccines is far superior to other vector-based vaccines. Specifically, intranasal priming with rFPV vaccines can recruit unique antigen-presenting cells, which induce excellent mucosal and systemic HIV-specific CD8+ T-cell immunity. In this study, we have for the first time investigated the in vivo dissemination, safety and expression kinetics of rFPV post intranasal delivery using recombinant viruses expressing green fluorescent protein or mCherry. Both confocal microscopy of tissue sections using green fluorescent protein and in vivo Imaging System (IVIS) spectrum live animal and whole organ imaging studies using mCherry revealed that (i) the peak antigen expression occurs 12 to 24 h post vaccination and no active viral gene expression is detected 96 h post vaccination. (ii) The virus only infects the initial vaccination site (lung and nasal cavity) and does not disseminate to distal sites such as the spleen or gut. (iii) More importantly, rFPV does not cross the olfactory receptor neuron pathway. Collectively, our findings indicate that rFPV vector-based vaccines have all the hallmarks of a safe and effective mucosal delivery vector, suitable for clinical evaluation.
Collapse
Affiliation(s)
- David G Townsend
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
- Present address: Division of Infectious Diseases, Department of Internal Medicine, The University of Utah, Salt Lake City, UT, USA
| | - Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| |
Collapse
|
14
|
Hu Z, Zhu L, Wang J, Wan Y, Yuan S, Chen J, Ding X, Qiu C, Zhang X, Qiu C, Xu J. Immune Signature of Enhanced Functional Avidity CD8 + T Cells in vivo Induced by Vaccinia Vectored Vaccine. Sci Rep 2017; 7:41558. [PMID: 28155878 PMCID: PMC5290741 DOI: 10.1038/srep41558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/21/2016] [Indexed: 11/09/2022] Open
Abstract
Functional avidity of T cells is a critical determinant for clearing viral infection and eliminating tumor. Understanding how functional avidity is maintained in T cells is imperative for immunotherapy. However, studies systematically characterize T cell with high functional avidity induced in vivo are still lacking. Previously, we and others found vaccinia vectored vaccine (VACV) induced antigen-specific CD8+ T cells with relatively high functional avidity to those from DNA vaccine. Herein, we used functional, immune phenotyping and transcriptomic studies to define the immune signature of these CD8+ T cells with high functional avidity. Antigen-specific CD8+ T cells induced by VACV executed superior in vivo killing activity and displayed a distinct transcriptional profile, whereas no significantly differences were found in composition of memory sub-populations and cytokine poly-functionality. Transcriptional analyses revealed unique features of VACV induced CD8+ T cells in several biological processes, including transport, cell cycle, cell communication and metabolic processes. In summary, we characterize CD8+ T cells of high functional avidity induced in vivo by VACV, which not only improves our understanding of adaptive T cell immunity in VACV vaccination, but also provides clues to modulate functional avidity of CD8+ T cells for T cell based immunotherapy.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lingyan Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jing Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yanmin Wan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Songhua Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jian Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiangqing Ding
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Chenli Qiu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of MOE/ MOH, Fudan University, Shanghai, China
| | - Chao Qiu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of MOE/ MOH, Fudan University, Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology of MOE/ MOH, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Trivedi S, Ranasinghe C. The Influence of Immunization Route, Tissue Microenvironment, and Cytokine Cell Milieu on HIV-Specific CD8+ T Cells Measured Using Fluidigm Dynamic Arrays. PLoS One 2015; 10:e0126487. [PMID: 25946028 PMCID: PMC4422706 DOI: 10.1371/journal.pone.0126487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/03/2015] [Indexed: 01/01/2023] Open
Abstract
Thirty different genes including cytokines, chemokines, granzymes, perforin and specifically integrins were evaluated in Peyer's patch-KdGag197–205-specific CD8+ T cells (pools of 100 cells) using Fluidigm 48.48 Dynamic arrays following three different prime-boost immunization strategies. Data revealed that the route of prime or the booster immunization differentially influenced the integrin expression profile on gut KdGag197–205-specific CD8+ T cells. Specifically, elevated numbers of integrin αE and αD expressing gut KdGag197–205-specific CD8+ T cells were detected following mucosal but not systemic priming. Also, αE/β7 and αD/β2 heterodimerization were more noticeable in an intranasal (i.n.)/i.n. vaccination setting compared to i.n./intramuscular (i.m) or i.m./i.m. vaccinations. Moreover, in all vaccine groups tested α4 appeared to heterodimerize more closely with β7 then β1. Also MIP-1β, RANTES, CCR5, perforin and integrin α4 bio-markers were significantly elevated in i.n./i.m. and i.m./i.m. immunization groups compared to purely mucosal i.n./i.n. delivery. Furthermore, when wild type (WT) BALB/c and IL-13 knockout (KO) mice were immunized using i.n./i.m. strategy, MIP-1α, MIP-1β, RANTES, integrins α4, β1 and β7 mRNA expression levels were found to be significantly different, in mucosal verses systemic KdGag197–205-specific CD8+ T cells. Interestingly, the numbers of gut KdGag197–205-specific CD8+ T cells expressing gut-homing markers α4β7 and CCR9 protein were also significantly elevated in IL-13 KO compared to WT control. Collectively, our findings further corroborate that the route of vaccine delivery, tissue microenvironment and IL-13 depleted cytokine milieu can significantly alter the antigen-specific CD8+ T cell gene expression profiles and in turn modulate their functional avidities as well as homing capabilities.
Collapse
Affiliation(s)
- Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT, Australia
- * E-mail:
| |
Collapse
|
16
|
Trivedi S, Jackson RJ, Ranasinghe C. Different HIV pox viral vector-based vaccines and adjuvants can induce unique antigen presenting cells that modulate CD8 T cell avidity. Virology 2014; 468-470:479-489. [PMID: 25261870 DOI: 10.1016/j.virol.2014.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/04/2014] [Accepted: 09/06/2014] [Indexed: 11/25/2022]
Abstract
The lung-derived dendritic cell (LDC) recruitment following intranasal (i.n.) vaccination of different poxviral vector-based vaccines/adjuvants were evaluated to decipher how these factors influenced CD8(+) T cell avidity. Compared to the standard i.n. recombinant fowlpox virus (FPV)-HIV vaccination, the FPV-HIV IL-13Rα2 or IL-4Rα antagonist adjuvanted vaccines that induced higher avidity CD8(+) T cells, also recruited significantly elevated MHCII(+) CD11c(+) CD11b(+) CD103(-) CD64(-) MAR-1(-) conventional DC (cDCs) to the lung mucosae (hierarchy: IL-4R antagonist>IL-13Rα2>unadjuvanted). In contrast, elevated CD11b(-) CD103(+) LDCs were detected in animals that received recombinant HIV vaccinia virus (rVV) or Modified Vaccinia Ankara virus (MVA) vector-based vaccines. Adoptive transfer studies indicated that CD11b(-) CD103(+) LDCs significantly dampened HIV-specific CD8(+) T cell avidity compared to CD11b(+) CD103(-) LDCs. Collectively; our observations revealed that rFPV vector prime and transient inhibition of IL-4/IL-13 at the vaccination site favoured the recruitment of unique LDCs, associated with the induction of high quality immunity.
Collapse
Affiliation(s)
- Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia.
| | - Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| |
Collapse
|