1
|
Coppola J, Parren M, Bastidas R, Saye-Francisco K, Malvin J, Jardine JG, Gilbride RM, Ojha S, Feltham S, Morrow D, Barber-Axthelm A, Bochart R, Fast R, Oswald K, Shoemaker R, Lifson JD, Picker LJ, Burton DR, Hansen SG. Combining a rhesus cytomegalovirus/SIV vaccine with neutralizing antibody to protect against SIV challenge in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644395. [PMID: 40196580 PMCID: PMC11974736 DOI: 10.1101/2025.03.20.644395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
A vaccine is widely regarded as necessary for the control of the HIV pandemic and eventual eradication of AIDS. Neutralizing antibodies and MHC-E-restricted CD8+ T cells have both been shown capable of vaccine protection against the simian counterpart of HIV, SIV, in rhesus macaques. Here we provide preliminary evidence that combining these orthogonal antiviral mechanisms can provide increased protection against SIV challenge such that replication arrest observed following vaccination with a rhesus cytomegalovirus (RhCMV/SIV)-based vaccine was enhanced in the presence of passively administered incompletely protective levels of neutralizing antibody. The report invites studies involving larger cohorts of macaques and alternate routes of providing neutralizing antibody.
Collapse
|
2
|
Zhang M, Wang C, Pan J, Cui H, Zhao X. Advancing novel veterinary vaccines: From comprehensive antigen and adjuvant design to preparation process optimization. Int Immunopharmacol 2025; 145:113784. [PMID: 39672026 DOI: 10.1016/j.intimp.2024.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
Vaccination stands as the paramount and cost-effective strategy for the prevention and management of animal infectious diseases. With the advances in biological technology, materials science and industrial optimization, substantial progress has been made in the development of innovative veterinary vaccines. A majority of the novel vaccines under current investigation tend to stimulate multiple immune pathways and to achieve long-term resistance against infectious diseases, yet it remains imperative to concentrate research efforts on the efficient utilization of vaccines, mitigating toxic side effects, and ensuring safe production processes. This article presents an overview of research progress in veterinary vaccines, encompassing comprehensive antigen design, adjuvant formulation advancements, preparation process optimization, and rigorous immune efficacy evaluation. It summarizes cutting-edge vaccines derived from in vitro synthesis and in vivo application, emphasizing immunogenic components and immune response mechanisms. It also highlights novel biological adjuvants that enhance immune efficacy, diversify raw materials, and possess targeted functions, while comprehensively exploring advancements in production methodologies and compatible vaccine products. By establishing a foundation for the integrated use of these innovative veterinary vaccines, this work facilitates future interdisciplinary cooperation in their advancement, aiming to accelerate the achievement of herd immunity through concerted efforts.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunxin Wang
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junqian Pan
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haixin Cui
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
3
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 PMCID: PMC11736809 DOI: 10.1016/j.cell.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Govindan R, Stephenson KE. HIV Vaccine Development at a Crossroads: New B and T Cell Approaches. Vaccines (Basel) 2024; 12:1043. [PMID: 39340073 PMCID: PMC11435826 DOI: 10.3390/vaccines12091043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Despite rigorous scientific efforts over the forty years since the onset of the global HIV pandemic, a safe and effective HIV-1 vaccine remains elusive. The challenges of HIV vaccine development have proven immense, in large part due to the tremendous sequence diversity of HIV and its ability to escape from antiviral adaptive immune responses. In recent years, several phase 3 efficacy trials have been conducted, testing a similar hypothesis, e.g., that non-neutralizing antibodies and classical cellular immune responses could prevent HIV-1 acquisition. These studies were not successful. As a result, the field has now pivoted to bold novel approaches, including sequential immunization strategies to drive the generation of broadly neutralizing antibodies and human CMV-vectored vaccines to elicit MHC-E-restricted CD8+ T cell responses. Many of these vaccine candidates are now in phase 1 trials, with early promising results.
Collapse
Affiliation(s)
- Ramesh Govindan
- Division of Infectious Diseases and Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Kathryn E. Stephenson
- Division of Infectious Diseases and Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Harvard Medical School, Boston, MA 02115, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
5
|
Yount KS, Darville T. Immunity to Sexually Transmitted Bacterial Infections of the Female Genital Tract: Toward Effective Vaccines. Vaccines (Basel) 2024; 12:863. [PMID: 39203989 PMCID: PMC11359697 DOI: 10.3390/vaccines12080863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Sexually transmitted infections (STIs) caused by bacterial pathogens Chlamydia trachomatis, Neisseria gonorrhoeae, and Treponema pallidum present significant public health challenges. These infections profoundly impact reproductive health, leading to pelvic inflammatory disease, infertility, and increased susceptibility to other infections. Prevention measures, including antibiotic treatments, are limited by the often-asymptomatic nature of these infections, the need for repetitive and continual screening of sexually active persons, antibiotic resistance for gonorrhea, and shortages of penicillin for syphilis. While vaccines exist for viral STIs like human papillomavirus (HPV) and hepatitis B virus (HBV), there are no vaccines available for bacterial STIs. This review examines the immune responses in the female genital tract to these bacterial pathogens and the implications for developing effective vaccines against bacterial STIs.
Collapse
Affiliation(s)
| | - Toni Darville
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
6
|
Arunachalam PS, Ha N, Dennison SM, Spreng RL, Seaton KE, Xiao P, Feng Y, Zarnitsyna VI, Kazmin D, Hu M, Santagata JM, Xie X, Rogers K, Shirreff LM, Chottin C, Spencer AJ, Dutta S, Prieto K, Julien JP, Tomai M, Fox CB, Villinger F, Hill AVS, Tomaras GD, Pulendran B. A comparative immunological assessment of multiple clinical-stage adjuvants for the R21 malaria vaccine in nonhuman primates. Sci Transl Med 2024; 16:eadn6605. [PMID: 39083589 DOI: 10.1126/scitranslmed.adn6605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Authorization of the Matrix-M (MM)-adjuvanted R21 vaccine by three countries and its subsequent endorsement by the World Health Organization for malaria prevention in children are a milestone in the fight against malaria. Yet, our understanding of the innate and adaptive immune responses elicited by this vaccine remains limited. Here, we compared three clinically relevant adjuvants [3M-052 + aluminum hydroxide (Alum) (3M), a TLR7/8 agonist formulated in Alum; GLA-LSQ, a TLR4 agonist formulated in liposomes with QS-21; and MM, the now-approved adjuvant for R21] for their capacity to induce durable immune responses to R21 in macaques. R21 adjuvanted with 3M on a 0, 8, and 23-week schedule elicited anti-circumsporozoite antibody responses comparable in magnitude to the R21/MM vaccine administered using a 0-4-8-week regimen and persisted up to 72 weeks with a half-life of 337 days. A booster dose at 72 weeks induced a recall response similar to the R21/MM vaccination. In contrast, R21/GLA-LSQ immunization induced a lower, short-lived response at the dose used. Consistent with the durable serum antibody responses, MM and 3M induced long-lived plasma cells in the bone marrow and other tissues, including the spleen. Furthermore, whereas 3M stimulated potent and persistent antiviral transcriptional and cytokine signatures after primary and booster immunizations, MM induced enhanced expression of interferon- and TH2-related signatures more highly after the booster vaccination. Collectively, these findings provide a resource on the immune responses of three clinically relevant adjuvants with R21 and highlight the promise of 3M as another adjuvant for malarial vaccines.
Collapse
Affiliation(s)
- Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - NaYoung Ha
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - S Moses Dennison
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
| | - Rachel L Spreng
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
- Duke Human Vaccine Institute, Duke University, Durham, NC 27703, USA
| | - Kelly E Seaton
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
| | - Peng Xiao
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Dmitri Kazmin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jordan M Santagata
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Xia Xie
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kenneth Rogers
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Lisa M Shirreff
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Claire Chottin
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | | | - Sheetij Dutta
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Katherine Prieto
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
- Departments of Biochemistry and Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | | | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Department of Surgery, Duke University, Durham, NC 27701, USA
- Duke Human Vaccine Institute, Duke University, Durham, NC 27703, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Maciel M, Amara RR, Bar KJ, Crotty S, Deeks SG, Duplessis C, Gaiha G, McElrath MJ, McMichael A, Palin A, Rutishauser R, Shapiro S, Smiley ST, D'Souza MP. Exploring synergies between B- and T-cell vaccine approaches to optimize immune responses against HIV-workshop report. NPJ Vaccines 2024; 9:39. [PMID: 38383616 PMCID: PMC10881492 DOI: 10.1038/s41541-024-00818-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Affiliation(s)
- Milton Maciel
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rama R Amara
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Katharine J Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
- Division of HIV, Infectious Diseases, and Global Medicine, San Francisco, CA, USA
| | - Steven G Deeks
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Christopher Duplessis
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Gaurav Gaiha
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew McMichael
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Amy Palin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rachel Rutishauser
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Stuart Shapiro
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Stephen T Smiley
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - M Patricia D'Souza
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
8
|
Nkolola JP, Barouch DH. Prophylactic HIV-1 vaccine trials: past, present, and future. Lancet HIV 2024; 11:e117-e124. [PMID: 38141639 PMCID: PMC11736820 DOI: 10.1016/s2352-3018(23)00264-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/24/2023] [Accepted: 10/11/2023] [Indexed: 12/25/2023]
Abstract
An effective HIV-1 vaccine is a global health priority but has remained elusive for more than 40 years. Key scientific hurdles that have hampered vaccine development are the unprecedented genetic variability of the virus, the rapid establishment of persistent viral latency, and the challenges associated with induction of broadly neutralising antibodies. Clinical trials have been instrumental in evaluating scientific concepts and testing vaccine strategies. This Review discusses lessons learned from clinical trials of HIV-1 vaccines, current technologies that are being explored, and future considerations in the development of a safe and effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Joseph P Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
9
|
Borgo GM, Rutishauser RL. Generating and measuring effective vaccine-elicited HIV-specific CD8 + T cell responses. Curr Opin HIV AIDS 2023; 18:331-341. [PMID: 37751362 PMCID: PMC10552829 DOI: 10.1097/coh.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW There is growing consensus that eliciting CD8 + T cells in addition to antibodies may be required for an effective HIV vaccine for both prevention and cure. Here, we review key qualities of vaccine-elicited CD8 + T cells as well as major CD8 + T cell-based delivery platforms used in recent HIV vaccine clinical trials. RECENT FINDINGS Much progress has been made in improving HIV immunogen design and delivery platforms to optimize CD8 + T cell responses. With regards to viral vectors, recent trials have tested newer chimp and human adenovirus vectors as well as a CMV vector. DNA vaccine immunogenicity has been increased by delivering the vaccines by electroporation and together with adjuvants as well as administering them as part of a heterologous regimen. In preclinical models, self-amplifying RNA vaccines can generate durable tissue-based CD8 + T cells. While it may be beneficial for HIV vaccines to recapitulate the functional and phenotypic features of HIV-specific CD8 + T cells isolated from elite controllers, most of these features are not routinely measured in HIV vaccine clinical trials. SUMMARY Identifying a vaccine capable of generating durable T cell responses that target mutationally vulnerable epitopes and that can rapidly intercept infecting or rebounding virus remains a challenge for HIV. Comprehensive assessment of HIV vaccine-elicited CD8 + T cells, as well as comparisons between different vaccine platforms, will be critical to advance our understanding of how to design better CD8 + T cell-based vaccines for HIV.
Collapse
Affiliation(s)
- Gina M Borgo
- Department of Medicine, University of California, San Francisco, California, USA
| | | |
Collapse
|
10
|
Counts JA, Saunders KO. Guiding HIV-1 vaccine development with preclinical nonhuman primate research. Curr Opin HIV AIDS 2023; 18:315-322. [PMID: 37712825 PMCID: PMC10810179 DOI: 10.1097/coh.0000000000000819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
PURPOSE OF THE REVIEW Nonhuman primates (NHPs) are seen as the closest animal model to humans in terms of anatomy and immune system makeup. Here, we review how preclinical studies in this model system are teaching the field of HIV vaccinology the basic immunology that is needed to induce broadly neutralizing antibodies (bnAbs) with vaccination and elicit protective T cell responses. These lessons are being translated into clinical trials to advance towards protective active vaccination against HIV-1 infection. RECENT FINDINGS Preclinical vaccination studies in NHPs have shown that highly engineered HIV-1 immunogens can initiate bnAb precursors providing proof of concept for Phase I clinical trials. Additionally, NHP models of HIV-1 infection are elucidating the pathways for bnAb development while serving as systems to evaluate vaccine protection. Innovative immunization strategies have increased affinity maturation of HIV-1 antibodies in long-lived germinal centers. Preclinical studies in macaques have defined the protective level of neutralizing antibodies and have shown that T cell responses can synergize with antibody-mediated immunity to provide protection in the presence of lower neutralizing antibody titers. SUMMARY The NHP model provides vaccine regimens and desired antibody and T cell responses that serve as benchmarks for clinical trials, accelerating HIV vaccine design.
Collapse
Affiliation(s)
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
11
|
Diniz MO, Maini MK, Swadling L. T cell control of SARS-CoV-2: When, which, and where? Semin Immunol 2023; 70:101828. [PMID: 37651850 DOI: 10.1016/j.smim.2023.101828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Efficient immune protection against viruses such as SARS-CoV-2 requires the coordinated activity of innate immunity, B and T cells. Accumulating data point to a critical role for T cells not only in the clearance of established infection, but also for aborting viral replication independently of humoral immunity. Here we review the evidence supporting the contribution of antiviral T cells and consider which of their qualitative features favour efficient control of infection. We highlight how studies of SARS-CoV-2 and other coronaviridae in animals and humans have provided important lessons on the optimal timing (When), functionality and specificity (Which), and location (Where) of antiviral T cells. We discuss the clinical implications, particularly for the development of next-generation vaccines, and emphasise areas requiring further study.
Collapse
Affiliation(s)
- Mariana O Diniz
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| |
Collapse
|
12
|
Edwards C, Oakes RS, Jewell CM. Tuning innate immune function using microneedles containing multiple classes of toll-like receptor agonists. NANOSCALE 2023; 15:8662-8674. [PMID: 37185984 PMCID: PMC10358826 DOI: 10.1039/d3nr00333g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Microneedle arrays (MNAs) are patches displaying hundreds of micron-scale needles that can penetrate skin. As a result, these arrays efficiently and painlessly access this immune cell-rich niche, motivating significant clinical interest in MNA-based vaccines. Our lab has developed immune polyelectrolyte multilayers (iPEMs), nanostructures built entirely from immune signals employing electrostatic self-assembly. iPEMs consist of positively charged peptide antigen and negatively charged toll-like receptor agonists (TLRas) to assemble these components at ultra-high density since no carrier is needed. Here we used this technology to deliver MNAs with antigen and defined ratios of multiple classes of TLRa. Notably, this approach resulted in facile assembly and corresponding signal transduction through each respective TLR pathway. This control ultimately activated primary antigen presenting cells and drove proliferation of antigen-specific T cells. In related in vivo vaccine studies, application of MNAs resulted in distinct T cells response depending on the number of TLRa classes delivered with MNAs. These MNAs technologies create an opportunity to deliver nanostructured vaccine components at high density, and to probe integration of multiple TLRas in skin to tune immunity.
Collapse
Affiliation(s)
- Camilla Edwards
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
| | - Robert S Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
Amaya L, Grigoryan L, Li Z, Lee A, Wender PA, Pulendran B, Chang HY. Circular RNA vaccine induces potent T cell responses. Proc Natl Acad Sci U S A 2023; 120:e2302191120. [PMID: 37155869 PMCID: PMC10193964 DOI: 10.1073/pnas.2302191120] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of RNAs commonly found across eukaryotes and viruses, characterized by their resistance to exonuclease-mediated degradation. Their superior stability compared to linear RNAs, combined with previous work showing that engineered circRNAs serve as efficient protein translation templates, make circRNA a promising candidate for RNA medicine. Here, we systematically examine the adjuvant activity, route of administration, and antigen-specific immunity of circRNA vaccination in mice. Potent circRNA adjuvant activity is associated with RNA uptake and activation of myeloid cells in the draining lymph nodes and transient cytokine release. Immunization of mice with engineered circRNA encoding a protein antigen delivered by a charge-altering releasable transporter induced innate activation of dendritic cells, robust antigen-specific CD8 T cell responses in lymph nodes and tissues, and strong antitumor efficacy as a therapeutic cancer vaccine. These results highlight the potential utility of circRNA vaccines for stimulating potent innate and T cell responses in tissues.
Collapse
Affiliation(s)
- Laura Amaya
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Zhijian Li
- Department of Chemistry, Stanford University, Stanford, CA94305
| | - Audrey Lee
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA94305
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA94305
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| |
Collapse
|
14
|
Williams LD, Shen X, Sawant SS, Akapirat S, Dahora LC, Tay MZ, Stanfield-Oakley S, Wills S, Goodman D, Tenney D, Spreng RL, Zhang L, Yates NL, Montefiori DC, Eller MA, Easterhoff D, Hope TJ, Rerks-Ngarm S, Pittisuttithum P, Nitayaphan S, Excler JL, Kim JH, Michael NL, Robb ML, O’Connell RJ, Karasavvas N, Vasan S, Ferrari G, Tomaras GD, RV305 study team. Viral vector delivered immunogen focuses HIV-1 antibody specificity and increases durability of the circulating antibody recall response. PLoS Pathog 2023; 19:e1011359. [PMID: 37256916 PMCID: PMC10284421 DOI: 10.1371/journal.ppat.1011359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/21/2023] [Accepted: 04/14/2023] [Indexed: 06/02/2023] Open
Abstract
The modestly efficacious HIV-1 vaccine regimen (RV144) conferred 31% vaccine efficacy at 3 years following the four-shot immunization series, coupled with rapid waning of putative immune correlates of decreased infection risk. New strategies to increase magnitude and durability of protective immunity are critically needed. The RV305 HIV-1 clinical trial evaluated the immunological impact of a follow-up boost of HIV-1-uninfected RV144 recipients after 6-8 years with RV144 immunogens (ALVAC-HIV alone, AIDSVAX B/E gp120 alone, or ALVAC-HIV + AIDSVAX B/E gp120). Previous reports demonstrated that this regimen elicited higher binding, antibody Fc function, and cellular responses than the primary RV144 regimen. However, the impact of the canarypox viral vector in driving antibody specificity, breadth, durability and function is unknown. We performed a follow-up analysis of humoral responses elicited in RV305 to determine the impact of the different booster immunogens on HIV-1 epitope specificity, antibody subclass, isotype, and Fc effector functions. Importantly, we observed that the ALVAC vaccine component directly contributed to improved breadth, function, and durability of vaccine-elicited antibody responses. Extended boosts in RV305 increased circulating antibody concentration and coverage of heterologous HIV-1 strains by V1V2-specific antibodies above estimated protective levels observed in RV144. Antibody Fc effector functions, specifically antibody-dependent cellular cytotoxicity and phagocytosis, were boosted to higher levels than was achieved in RV144. V1V2 Env IgG3, a correlate of lower HIV-1 risk, was not increased; plasma Env IgA (specifically IgA1), a correlate of increased HIV-1 risk, was elevated. The quality of the circulating polyclonal antibody response changed with each booster immunization. Remarkably, the ALVAC-HIV booster immunogen induced antibody responses post-second boost, indicating that the viral vector immunogen can be utilized to selectively enhance immune correlates of decreased HIV-1 risk. These results reveal a complex dynamic of HIV-1 immunity post-vaccination that may require careful balancing to achieve protective immunity in the vaccinated population. Trial registration: RV305 clinical trial (ClinicalTrials.gov number, NCT01435135). ClinicalTrials.gov Identifier: NCT00223080.
Collapse
Affiliation(s)
- LaTonya D. Williams
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sheetal S. Sawant
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Siriwat Akapirat
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Lindsay C. Dahora
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Matthew Zirui Tay
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sherry Stanfield-Oakley
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Saintedym Wills
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Derrick Goodman
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - DeAnna Tenney
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Rachel L. Spreng
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lu Zhang
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nicole L. Yates
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Michael A. Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - David Easterhoff
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | | | - Punnee Pittisuttithum
- Royal Thai Army Component, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sorachai Nitayaphan
- Royal Thai Army Component, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Jean-Louis Excler
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jerome H. Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Nelson L. Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Merlin L. Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Robert J. O’Connell
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Nicos Karasavvas
- Department of Retrovirology, US Army Medical Directorate, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sandhya Vasan
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Guido Ferrari
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Molecular Genetics Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | | |
Collapse
|
15
|
Phoolcharoen W, Shanmugaraj B, Khorattanakulchai N, Sunyakumthorn P, Pichyangkul S, Taepavarapruk P, Praserthsee W, Malaivijitnond S, Manopwisedjaroen S, Thitithanyanont A, Srisutthisamphan K, Jongkaewwattana A, Tomai M, Fox CB, Taychakhoonavudh S. Preclinical evaluation of immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052-Alum adjuvant. Vaccine 2023; 41:2781-2792. [PMID: 36963999 PMCID: PMC10027959 DOI: 10.1016/j.vaccine.2023.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
Cost-effective, and accessible vaccines are needed for mass immunization to control the ongoing coronavirus disease 2019 (COVID-19), especially in low- and middle-income countries (LMIC).A plant-based vaccine is an attractive technology platform since the recombinant proteins can be easily produced at large scale and low cost. For the recombinant subunit-based vaccines, effective adjuvants are crucial to enhance the magnitude and breadth of immune responses elicited by the vaccine. In this study, we report a preclinical evaluation of the immunogenicity, efficacy and safety of a recombinant plant-based SARS-CoV-2 RBD vaccine formulated with 3M-052 (TLR7/8 agonist)-Alum adjuvant. This vaccine formulation, named Baiya SARS-CoV-2 Vax 2, induced significant levels of RBD-specific IgG and neutralizing antibody responses in mice. A viral challenge study using humanized K18-hACE2 mice has shown that animals vaccinated with two doses of Baiya SARS-CoV-2 Vax 2 established immune protection against SARS-CoV-2. A study in nonhuman primates (cynomolgus monkeys) indicated that immunization with two doses of Baiya SARS-CoV-2 Vax 2 was safe, well tolerated, and induced neutralizing antibodies against the prototype virus and other viral variants (Alpha, Beta, Gamma, Delta, and Omicron subvariants). The toxicity of Baiya SARS-CoV-2 Vax 2 was further investigated in Jcl:SD rats, which demonstrated that a single dose and repeated doses of Baiya SARS-CoV-2 Vax 2 were well tolerated and no mortality or unanticipated findings were observed. Overall, these preclinical findings support further clinical development of Baiya SARS-CoV-2 Vax 2.
Collapse
Affiliation(s)
- Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | | | - Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Sathit Pichyangkul
- US Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Pornnarin Taepavarapruk
- Center for Animal Research and Department of Physiology, Faculty of Medical Science, Naresuan University, Pitsanulok 65000, Thailand
| | | | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Mark Tomai
- 3M Healthcare, 3M Center, Bldg 270-4N-04, St. Paul, MN 55144-1000, USA
| | - Christopher B Fox
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Ste 400, Seattle, WA 98102, USA
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
16
|
Yin Q, Luo W, Mallajosyula V, Bo Y, Guo J, Xie J, Sun M, Verma R, Li C, Constantz CM, Wagar LE, Li J, Sola E, Gupta N, Wang C, Kask O, Chen X, Yuan X, Wu NC, Rao J, Chien YH, Cheng J, Pulendran B, Davis MM. A TLR7-nanoparticle adjuvant promotes a broad immune response against heterologous strains of influenza and SARS-CoV-2. NATURE MATERIALS 2023; 22:380-390. [PMID: 36717665 PMCID: PMC9981462 DOI: 10.1038/s41563-022-01464-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/12/2022] [Indexed: 06/01/2023]
Abstract
The ideal vaccine against viruses such as influenza and SARS-CoV-2 must provide a robust, durable and broad immune protection against multiple viral variants. However, antibody responses to current vaccines often lack robust cross-reactivity. Here we describe a polymeric Toll-like receptor 7 agonist nanoparticle (TLR7-NP) adjuvant, which enhances lymph node targeting, and leads to persistent activation of immune cells and broad immune responses. When mixed with alum-adsorbed antigens, this TLR7-NP adjuvant elicits cross-reactive antibodies for both dominant and subdominant epitopes and antigen-specific CD8+ T-cell responses in mice. This TLR7-NP-adjuvanted influenza subunit vaccine successfully protects mice against viral challenge of a different strain. This strategy also enhances the antibody response to a SARS-CoV-2 subunit vaccine against multiple viral variants that have emerged. Moreover, this TLR7-NP augments antigen-specific responses in human tonsil organoids. Overall, we describe a nanoparticle adjuvant to improve immune responses to viral antigens, with promising implications for developing broadly protective vaccines.
Collapse
Affiliation(s)
- Qian Yin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Wei Luo
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vamsee Mallajosyula
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jing Guo
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jinghang Xie
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Meng Sun
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit Verma
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Christian M Constantz
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Lisa E Wagar
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Elsa Sola
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Neha Gupta
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chunlin Wang
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Oliver Kask
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Xin Chen
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Xue Yuan
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Yueh-Hsiu Chien
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, USA.
- The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
17
|
Cohen KW, Fiore-Gartland A, Walsh SR, Yusim K, Frahm N, Elizaga ML, Maenza J, Scott H, Mayer KH, Goepfert PA, Edupuganti S, Pantaleo G, Hutter J, Morris DE, De Rosa SC, Geraghty DE, Robb ML, Michael NL, Fischer W, Giorgi EE, Malhi H, Pensiero MN, Ferrari G, Tomaras GD, Montefiori DC, Gilbert PB, McElrath MJ, Haynes BF, Korber BT, Baden LR, the NIAID HVTN 106 Study Group. Trivalent mosaic or consensus HIV immunogens prime humoral and broader cellular immune responses in adults. J Clin Invest 2023; 133:e163338. [PMID: 36787249 PMCID: PMC9927951 DOI: 10.1172/jci163338] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/27/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUNDMosaic and consensus HIV-1 immunogens provide two distinct approaches to elicit greater breadth of coverage against globally circulating HIV-1 and have shown improved immunologic breadth in nonhuman primate models.METHODSThis double-blind randomized trial enrolled 105 healthy HIV-uninfected adults who received 3 doses of either a trivalent global mosaic, a group M consensus (CON-S), or a natural clade B (Nat-B) gp160 env DNA vaccine followed by 2 doses of a heterologous modified vaccinia Ankara-vectored HIV-1 vaccine or placebo. We performed prespecified blinded immunogenicity analyses at day 70 and day 238 after the first immunization. T cell responses to vaccine antigens and 5 heterologous Env variants were fully mapped.RESULTSEnv-specific CD4+ T cell responses were induced in 71% of the mosaic vaccine recipients versus 48% of the CON-S recipients and 48% of the natural Env recipients. The mean number of T cell epitopes recognized was 2.5 (95% CI, 1.2-4.2) for mosaic recipients, 1.6 (95% CI, 0.82-2.6) for CON-S recipients, and 1.1 (95% CI, 0.62-1.71) for Nat-B recipients. Mean breadth was significantly greater in the mosaic group than in the Nat-B group using overall (P = 0.014), prime-matched (P = 0.002), heterologous (P = 0.046), and boost-matched (P = 0.009) measures. Overall T cell breadth was largely due to Env-specific CD4+ T cell responses.CONCLUSIONPriming with a mosaic antigen significantly increased the number of epitopes recognized by Env-specific T cells and enabled more, albeit still limited, cross-recognition of heterologous variants. Mosaic and consensus immunogens are promising approaches to address global diversity of HIV-1.TRIAL REGISTRATIONClinicalTrials.gov NCT02296541.FUNDINGUS NIH grants UM1 AI068614, UM1 AI068635, UM1 AI068618, UM1 AI069412, UL1 RR025758, P30 AI064518, UM1 AI100645, and UM1 AI144371, and Bill & Melinda Gates Foundation grant OPP52282.
Collapse
Affiliation(s)
- Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen R. Walsh
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Karina Yusim
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hyman Scott
- San Francisco Department of Public Health, San Francisco, California, USA
| | - Kenneth H. Mayer
- Harvard Medical School, Boston, Massachusetts, USA
- The Fenway Institute, Fenway Health, Boston, Massachusetts, USA
| | | | | | | | - Julia Hutter
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Daryl E. Morris
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Daniel E. Geraghty
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Merlin L. Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Nelson L. Michael
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Will Fischer
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Elena E. Giorgi
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Harmandeep Malhi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michael N. Pensiero
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - David C. Montefiori
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Bette T. Korber
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, and New Mexico Consortium, Los Alamos, New Mexico, USA
| | - Lindsey R. Baden
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
18
|
Rosato PC, Lotfi-Emran S, Joag V, Wijeyesinghe S, Quarnstrom CF, Degefu HN, Nedellec R, Schenkel JM, Beura LK, Hangartner L, Burton DR, Masopust D. Tissue-resident memory T cells trigger rapid exudation and local antibody accumulation. Mucosal Immunol 2023; 16:17-26. [PMID: 36657662 PMCID: PMC10338064 DOI: 10.1016/j.mucimm.2022.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023]
Abstract
Adaptive immunity is didactically partitioned into humoral and cell-mediated effector mechanisms, which may imply that each arm is separate and does not function together. Here, we report that the activation of CD8+ resident memory T cells (TRM) in nonlymphoid tissues triggers vascular permeability, which facilitates rapid distribution of serum antibodies into local tissues. TRM reactivation was associated with transcriptional upregulation of antiviral signaling pathways as well as Fc receptors and components of the complement cascade. Effects were local, but evidence is presented that TRM in brain and reproductive mucosa are both competent to induce rapid antibody exudation. TRM reactivation in the mouse female genital tract increased local concentrations of virus-specific neutralizing antibodies, including anti-vesicular stomatitis virus, and passively transferred anti-HIV antibodies. We showed that this response was sufficient to increase the efficacy of ex vivo vesicular stomatitis virus neutralization. These results indicate that CD8+ TRM antigen recognition can enhance local humoral immunity.
Collapse
Affiliation(s)
- Pamela C Rosato
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA; Geisel School of Medicine at Dartmouth College, Dartmouth Cancer Center, Department of Microbiology and Immunology, Lebanon, NH, USA
| | - Sahar Lotfi-Emran
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Vineet Joag
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Sathi Wijeyesinghe
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Clare F Quarnstrom
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Hanna N Degefu
- Geisel School of Medicine at Dartmouth College, Dartmouth Cancer Center, Department of Microbiology and Immunology, Lebanon, NH, USA
| | - Rebecca Nedellec
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jason M Schenkel
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Lalit K Beura
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA; Brown University, Department of Molecular Microbiology and Immunology, Providence, RI, USA
| | - Lars Hangartner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - David Masopust
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA.
| |
Collapse
|
19
|
Harwood OE, Balgeman AJ, Weaver AJ, Ellis-Connell AL, Weiler AM, Erickson KN, Matschke LM, Golfinos AE, Vezys V, Skinner PJ, Safrit JT, Edlefsen PT, Reynolds MR, Friedrich TC, O’Connor SL. Transient T Cell Expansion, Activation, and Proliferation in Therapeutically Vaccinated Simian Immunodeficiency Virus-Positive Macaques Treated with N-803. J Virol 2022; 96:e0142422. [PMID: 36377872 PMCID: PMC9749465 DOI: 10.1128/jvi.01424-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccine strategies aimed at eliciting human immunodeficiency virus (HIV)-specific CD8+ T cells are one major target of interest in HIV functional cure strategies. We hypothesized that CD8+ T cells elicited by therapeutic vaccination during antiretroviral therapy (ART) would be recalled and boosted by treatment with the interleukin 15 (IL-15) superagonist N-803 after ART discontinuation. We intravenously immunized four simian immunodeficiency virus-positive (SIV+) Mauritian cynomolgus macaques receiving ART with vesicular stomatitis virus (VSV), modified vaccinia virus Ankara strain (MVA), and recombinant adenovirus serotype 5 (rAd-5) vectors all expressing SIVmac239 Gag. Immediately after ART cessation, these animals received three doses of N-803. Four control animals received no vaccines or N-803. The vaccine regimen generated a high-magnitude response involving Gag-specific CD8+ T cells that were proliferative and biased toward an effector memory phenotype. We then compared cells elicited by vaccination (Gag specific) to cells elicited by SIV infection and unaffected by vaccination (Nef specific). We found that N-803 treatment enhanced the frequencies of both bulk and proliferating antigen-specific CD8+ T cells elicited by vaccination and the antigen-specific CD8+ T cells elicited by SIV infection. In sum, we demonstrate that a therapeutic heterologous prime-boost-boost (HPBB) vaccine can elicit antigen-specific effector memory CD8+ T cells that are boosted by N-803. IMPORTANCE While antiretroviral therapy (ART) can suppress HIV replication, it is not a cure. It is therefore essential to develop therapeutic strategies to enhance the immune system to better become activated and recognize virus-infected cells. Here, we evaluated a novel therapeutic vaccination strategy delivered to SIV+ Mauritian cynomolgus macaques receiving ART. ART was then discontinued and we delivered an immunotherapeutic agent (N-803) after ART withdrawal with the goal of eliciting and boosting anti-SIV cellular immunity. Immunologic and virologic analysis of peripheral blood and lymph nodes collected from these animals revealed transient boosts in the frequency, activation, proliferation, and memory phenotype of CD4+ and CD8+ T cells following each intervention. Overall, these results are important in educating the field of the transient nature of the immunological responses to this particular therapeutic regimen and the similar effects of N-803 on boosting T cells elicited by vaccination or elicited naturally by infection.
Collapse
Affiliation(s)
- Olivia E. Harwood
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Abigail J. Weaver
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | | | - Lea M. Matschke
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Athena E. Golfinos
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Matthew R. Reynolds
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Thomas C. Friedrich
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| |
Collapse
|
20
|
Nazeam JA, Singab ANB. Immunostimulant plant proteins: Potential candidates as vaccine adjuvants. Phytother Res 2022; 36:4345-4360. [PMID: 36128599 PMCID: PMC9538006 DOI: 10.1002/ptr.7624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/20/2022] [Accepted: 09/03/2022] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is shaking up global scientific structures toward addressing antibiotic resistance threats and indicates an urgent need to develop more cost-effective vaccines. Vaccine adjuvants play a crucial role in boosting immunogenicity and improving vaccine efficacy. The toxicity and adversity of most adjuvant formulations are the major human immunization problems, especially in routine pediatric and immunocompromised patients. The present review focused on preclinical studies of immunoadjuvant plant proteins in use with antiparasitic, antifungal, and antiviral vaccines. Moreover, this report outlines the current perspective of immunostimulant plant protein candidates that can be used by researchers in developing new generations of vaccine-adjuvants. Future clinical studies are required to substantiate the plant proteins' safety and applicability as a vaccine adjuvant in pharmaceutical manufacturing.
Collapse
Affiliation(s)
- Jilan A. Nazeam
- Pharmacognosy Department, Faculty of PharmacyOctober 6 UniversityGizaEgypt
| | | |
Collapse
|
21
|
Sahoo A, Jones AT, Cheedarla N, Gangadhara S, Roy V, Styles TM, Shiferaw A, Walter KL, Williams LD, Shen X, Ozorowski G, Lee WH, Burton S, Yi L, Song X, Qin ZS, Derdeyn CA, Ward AB, Clements JD, Varadarajan R, Tomaras GD, Kozlowski PA, Alter G, Amara RR. A clade C HIV-1 vaccine protects against heterologous SHIV infection by modulating IgG glycosylation and T helper response in macaques. Sci Immunol 2022; 7:eabl4102. [PMID: 35867800 PMCID: PMC9410801 DOI: 10.1126/sciimmunol.abl4102] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The rising global HIV-1 burden urgently requires vaccines capable of providing heterologous protection. Here, we developed a clade C HIV-1 vaccine consisting of priming with modified vaccinia Ankara (MVA) and boosting with cyclically permuted trimeric gp120 (CycP-gp120) protein, delivered either orally using a needle-free injector or through parenteral injection. We tested protective efficacy of the vaccine against intrarectal challenges with a pathogenic heterologous clade C SHIV infection in rhesus macaques. Both routes of vaccination induced a strong envelope-specific IgG in serum and rectal secretions directed against V1V2 scaffolds from a global panel of viruses with polyfunctional activities. Envelope-specific IgG showed lower fucosylation compared with total IgG at baseline, and most of the vaccine-induced proliferating blood CD4+ T cells did not express CCR5 and α4β7, markers associated with HIV target cells. After SHIV challenge, both routes of vaccination conferred significant and equivalent protection, with 40% of animals remaining uninfected at the end of six weekly repeated challenges with an estimated efficacy of 68% per exposure. Induction of envelope-specific IgG correlated positively with G1FB glycosylation, and G2S2F glycosylation correlated negatively with protection. Vaccine-induced TNF-α+ IFN-γ+ CD8+ T cells and TNF-α+ CD4+ T cells expressing low levels of CCR5 in the rectum at prechallenge were associated with decreased risk of SHIV acquisition. These results demonstrate that the clade C MVA/CycP-gp120 vaccine provides heterologous protection against a tier2 SHIV rectal challenge by inducing a polyfunctional antibody response with distinct Fc glycosylation profile, as well as cytotoxic CD8 T cell response and CCR5-negative T helper response in the rectum.
Collapse
Affiliation(s)
- Anusmita Sahoo
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andrew T Jones
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Narayanaiah Cheedarla
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tiffany M Styles
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Korey L Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - LaTonya D Williams
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - Samantha Burton
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Lasanajak Yi
- Department of Biochemistry, Emory Glycomics and Molecular Interactions Core (EGMIC), School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Xuezheng Song
- Department of Biochemistry, Emory Glycomics and Molecular Interactions Core (EGMIC), School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Cynthia A Derdeyn
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 8638, USA
| | - Raghavan Varadarajan
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, Karnataka 560012, India.,Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560012, India
| | - Georgia D Tomaras
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Rama Rao Amara
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
22
|
Chew K, Lee B, van Haren SD, Nanishi E, O’Meara T, Splaine JB, DeLeon M, Soni D, Seo HS, Dhe-Paganon S, Ozonoff A, Smith JA, Levy O, Dowling DJ. Adjuvant Discovery via a High Throughput Screen using Human Primary Mononuclear Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.17.496630. [PMID: 35860217 PMCID: PMC9298130 DOI: 10.1101/2022.06.17.496630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Motivation Vaccines are a key biomedical intervention to prevent the spread of infectious diseases, but their efficacy can be limited by insufficient immunogenicity and nonuniform reactogenic profiles. Adjuvants are molecules that potentiate vaccine responses by inducing innate immune activation. However, there are a limited number of adjuvants in approved vaccines, and current approaches for preclinical adjuvant discovery and development are inefficient. To enhance adjuvant identification, we developed a protocol based on in vitro screening of human primary leukocytes. Summary We describe a methodology utilizing high-throughput and high-content screening for novel adjuvant candidates that was used to screen a library of ~2,500 small molecules via a 384-well quantitative combined cytokine and flow cytometry immunoassay in primary human peripheral blood mononuclear cells (PBMCs) from 4 healthy adult study participants. Hits were identified based on their induction of soluble cytokine (TNF, IFNg and IL-10) secretion and PBMC maturation (CD 80/86, Ox40, and HLA-DR) in at least two of the four donors screened. From an initial set of 197 compounds identified using these biomarkers-an 8.6% hit rate-we downselected to five scaffolds that demonstrated robust efficacy and potency in vitro and evaluated the top hit, vinblastine sulfate, for adjuvanticity in vivo. Vinblastine sulfate significantly enhanced murine humoral responses to recombinant SARS-CoV-2 spike protein, including a four-fold enhancement of IgG titer production when compared to treatment with the spike antigen alone. Overall, we outline a methodology for discovering immunomodulators with adjuvant potential via high-throughput screening of PBMCs in vitro that yielded a lead compound with in vivo adjuvanticity.
Collapse
Affiliation(s)
- Katherine Chew
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | - Branden Lee
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | - Simon D. van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy O’Meara
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | | | - Maria DeLeon
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Jennifer A. Smith
- ICCB-Longwood Screening Facility, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - David J. Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Grigoryan L, Lee A, Walls AC, Lai L, Franco B, Arunachalam PS, Feng Y, Luo W, Vanderheiden A, Floyd K, Wrenn S, Pettie D, Miranda MC, Kepl E, Ravichandran R, Sydeman C, Brunette N, Murphy M, Fiala B, Carter L, Coffman RL, Novack D, Kleanthous H, O’Hagan DT, van der Most R, McLellan JS, Suthar M, Veesler D, King NP, Pulendran B. Adjuvanting a subunit SARS-CoV-2 vaccine with clinically relevant adjuvants induces durable protection in mice. NPJ Vaccines 2022; 7:55. [PMID: 35606518 PMCID: PMC9126867 DOI: 10.1038/s41541-022-00472-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 02/03/2022] [Indexed: 01/27/2023] Open
Abstract
Adjuvants enhance the magnitude and the durability of the immune response to vaccines. However, there is a paucity of comparative studies on the nature of the immune responses stimulated by leading adjuvant candidates. In this study, we compared five clinically relevant adjuvants in mice-alum, AS03 (a squalene-based adjuvant supplemented with α-tocopherol), AS37 (a TLR7 ligand emulsified in alum), CpG1018 (a TLR9 ligand emulsified in alum), O/W 1849101 (a squalene-based adjuvant)-for their capacity to stimulate immune responses when combined with a subunit vaccine under clinical development. We found that all four of the adjuvant candidates surpassed alum with respect to their capacity to induce enhanced and durable antigen-specific antibody responses. The TLR-agonist-based adjuvants CpG1018 (TLR9) and AS37 (TLR7) induced Th1-skewed CD4+ T cell responses, while alum, O/W, and AS03 induced a balanced Th1/Th2 response. Consistent with this, adjuvants induced distinct patterns of early innate responses. Finally, vaccines adjuvanted with AS03, AS37, and CpG1018/alum-induced durable neutralizing-antibody responses and significant protection against the B.1.351 variant 7 months following immunization. These results, together with our recent results from an identical study in non-human primates (NHPs), provide a comparative benchmarking of five clinically relevant vaccine adjuvants for their capacity to stimulate immunity to a subunit vaccine, demonstrating the capacity of adjuvanted SARS-CoV-2 subunit vaccines to provide durable protection against the B.1.351 variant. Furthermore, these results reveal differences between the widely-used C57BL/6 mouse strain and NHP animal models, highlighting the importance of species selection for future vaccine and adjuvant studies.
Collapse
Affiliation(s)
- Lilit Grigoryan
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| | - Audrey Lee
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| | - Alexandra C. Walls
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Present Address: Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195 USA
| | - Lilin Lai
- grid.189967.80000 0001 0941 6502Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA 30329 USA
| | - Benjamin Franco
- Veterinary Service Center, Department of Comparative Medicine, Stanford, CA USA
| | - Prabhu S. Arunachalam
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| | - Yupeng Feng
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| | - Wei Luo
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| | - Abigail Vanderheiden
- grid.189967.80000 0001 0941 6502Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA 30329 USA
| | - Katharine Floyd
- grid.189967.80000 0001 0941 6502Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA 30329 USA
| | - Samuel Wrenn
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Deleah Pettie
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Marcos C. Miranda
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Elizabeth Kepl
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Rashmi Ravichandran
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Claire Sydeman
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Natalie Brunette
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Michael Murphy
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Brooke Fiala
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Lauren Carter
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Robert L. Coffman
- grid.418630.80000 0004 0409 1245Dynavax Technologies Corporation, Emeryville, CA USA
| | - David Novack
- grid.418630.80000 0004 0409 1245Dynavax Technologies Corporation, Emeryville, CA USA
| | - Harry Kleanthous
- grid.418309.70000 0000 8990 8592Bill and Melinda Gates Foundation, Seattle, WA 98102 USA
| | | | | | - Jason S. McLellan
- grid.55460.320000000121548364Department of Molecular Biosciences, University of Texas, Austin, TX USA
| | - Mehul Suthar
- grid.189967.80000 0001 0941 6502Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA 30329 USA
| | - David Veesler
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Present Address: Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195 USA
| | - Neil P. King
- grid.34477.330000000122986657Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA USA
| | - Bali Pulendran
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA USA
| |
Collapse
|
24
|
Denis J, Mura M, Trignol A, Tournier JN. L’exploration de l’immunogénicité vaccinale. REVUE FRANCOPHONE DES LABORATOIRES 2022; 2022:40-52. [PMID: 35284007 PMCID: PMC8896448 DOI: 10.1016/s1773-035x(22)00098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Le développement de nouveaux vaccins était traditionnellement un travail de longue haleine, même si l’expérience récente de l’émergence de la Covid-19 a fait exploser les délais de développement et de production. Il n’en reste pas moins que le développement des vaccins dans les phases précliniques et les phases 1 et 2 de développement clinique est basé sur l’étude de la réponse immunitaire spécifique du système immunitaire adaptatif.
Collapse
|
25
|
Lee A, Scott MKD, Wimmers F, Arunachalam PS, Luo W, Fox CB, Tomai M, Khatri P, Pulendran B. A molecular atlas of innate immunity to adjuvanted and live attenuated vaccines, in mice. Nat Commun 2022; 13:549. [PMID: 35087093 PMCID: PMC8795432 DOI: 10.1038/s41467-022-28197-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 01/08/2022] [Indexed: 12/27/2022] Open
Abstract
Adjuvants hold great potential in enhancing vaccine efficacy, making the understanding and improving of adjuvants critical goals in vaccinology. The TLR7/8 agonist, 3M-052, induces long-lived humoral immunity in non-human primates and is currently being evaluated in human clinical trials. However, the innate mechanisms of 3M-052 have not been fully characterized. Here, we perform flow cytometry, single cell RNA-seq and ATAC-seq to profile the kinetics, transcriptomics and epigenomics of innate immune cells in murine draining lymph nodes following 3M-052-Alum/Ovalbumin immunization. We find that 3M-052-Alum/OVA induces a robust antiviral and interferon gene program, similar to the yellow fever vaccine, which is known to confer long-lasting protection. Activation of myeloid cells in dLNs persists through day 28 and single cell analysis reveals putative TF-gene regulatory programs in distinct myeloid cells and heterogeneity of monocytes. This study provides a comprehensive characterization of the transcriptomics and epigenomics of innate populations in the dLNs after vaccination.
Collapse
Affiliation(s)
- Audrey Lee
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Madeleine K D Scott
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Center for Biomedical Informatics, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Wei Luo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Mark Tomai
- 3M Corporate Research and Materials Lab, St. Paul, MN, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Center for Biomedical Informatics, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
27
|
Silva M, Kato Y, Melo MB, Phung I, Freeman BL, Li Z, Roh K, Van Wijnbergen JW, Watkins H, Enemuo CA, Hartwell BL, Chang JYH, Xiao S, Rodrigues KA, Cirelli KM, Li N, Haupt S, Aung A, Cossette B, Abraham W, Kataria S, Bastidas R, Bhiman J, Linde C, Bloom NI, Groschel B, Georgeson E, Phelps N, Thomas A, Bals J, Carnathan DG, Lingwood D, Burton DR, Alter G, Padera TP, Belcher AM, Schief WR, Silvestri G, Ruprecht RM, Crotty S, Irvine DJ. A particulate saponin/TLR agonist vaccine adjuvant alters lymph flow and modulates adaptive immunity. Sci Immunol 2021; 6:eabf1152. [PMID: 34860581 DOI: 10.1126/sciimmunol.abf1152] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Kato
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Mariane B Melo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Ivy Phung
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.,Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Brian L Freeman
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Zhongming Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kangsan Roh
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jan W Van Wijnbergen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hannah Watkins
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chiamaka A Enemuo
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brittany L Hartwell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason Y H Chang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shuhao Xiao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kristen A Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Harvard-MIT Health Sciences and Technology Program, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kimberly M Cirelli
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Na Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sonya Haupt
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.,Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin Cossette
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wuhbet Abraham
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Swati Kataria
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Raiza Bastidas
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jinal Bhiman
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Caitlyn Linde
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Nathaniel I Bloom
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Bettina Groschel
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA.,IAVI Neutralizing Antibody Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Erik Georgeson
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA.,IAVI Neutralizing Antibody Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole Phelps
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA.,IAVI Neutralizing Antibody Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ayush Thomas
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Julia Bals
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Diane G Carnathan
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Daniel Lingwood
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Dennis R Burton
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Timothy P Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, MGH Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Angela M Belcher
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William R Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Guido Silvestri
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ruth M Ruprecht
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Shane Crotty
- Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.,Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Consortium for HIV/AIDS Vaccine Development (CHAVD), Scripps Research Institute, La Jolla, CA 92037, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
28
|
Park SL, Mackay LK. Decoding Tissue-Residency: Programming and Potential of Frontline Memory T Cells. Cold Spring Harb Perspect Biol 2021; 13:a037960. [PMID: 33753406 PMCID: PMC8485744 DOI: 10.1101/cshperspect.a037960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Memory T-cell responses are partitioned between the blood, secondary lymphoid organs, and nonlymphoid tissues. Tissue-resident memory T (Trm) cells are a population of immune cells that remain permanently in tissues without recirculating in blood. These nonrecirculating cells serve as a principal node in the anamnestic response to invading pathogens and developing malignancies. Here, we contemplate how T-cell tissue residency is defined and shapes protective immunity in the steady state and in the context of disease. We review the properties and heterogeneity of Trm cells, highlight the critical roles these cells play in maintaining tissue homeostasis and eliciting immune pathology, and explore how they might be exploited to treat disease.
Collapse
Affiliation(s)
- Simone L Park
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Laura K Mackay
- Department of Microbiology & Immunology at The Peter Doherty Institute for Infection & Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
29
|
Najafabadi AH, Abadi ZIN, Aikins ME, Foulds KE, Donaldson MM, Yuan W, Okeke EB, Nam J, Xu Y, Weerappuli P, Hetrick T, Adams D, Lester PA, Salazar AM, Barouch DH, Schwendeman A, Seder RA, Moon JJ. Vaccine nanodiscs plus polyICLC elicit robust CD8+ T cell responses in mice and non-human primates. J Control Release 2021; 337:168-178. [PMID: 34280415 PMCID: PMC8440392 DOI: 10.1016/j.jconrel.2021.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
Conventional cancer vaccines based on soluble vaccines and traditional adjuvants have produced suboptimal therapeutic efficacy in clinical trials. Thus, there is an urgent need for vaccine technologies that can generate potent T cell responses with strong anti-tumor efficacy. We have previously reported the development of synthetic high-density protein (sHDL) nanodiscs for efficient lymph node (LN)-targeted co-delivery of antigen peptides and CpG oligonucleotides (a Toll-like receptor-9 agonist). Here, we performed a comparative study in mice and non-human primates (NHPs) to identify an ideal vaccine platform for induction of CD8+ T cell responses. In particular, we compared the efficacy of CpG class B, CpG class C, and polyICLC (a synthetic double-stranded RNA analog, a TLR-3 agonist), each formulated with antigen-carrying sHDL nanodiscs. Here, we report that sHDL-Ag admixed with polyICLC elicited robust Ag-specific CD8+ T cell responses in mice, and when used in combination with α-PD-1 immune checkpoint inhibitor, sHDL-Ag + polyICLC eliminated large established (~100 mm3) MC-38 tumors in mice. Moreover, sHDL-Gag + polyICLC induced robust Simian immunodeficiency virus Gag-specific, polyfunctional CD8+ T cell responses in rhesus macaques and could further amplify the efficacy of recombinant adenovirus-based vaccine. Notably, while both sHDL-Ag-CpG-B and sHDL-Ag-CpG-C generated strong Ag-specific CD8+ T cell responses in mice, their results were mixed in NHPs. Overall, sHDL combined with polyICLC offers a strong platform to induce CD8+ T cells for vaccine applications.
Collapse
Affiliation(s)
- Alireza Hassani Najafabadi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Zeynab Izadi Najaf Abadi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marisa E Aikins
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn E Foulds
- The Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mitzi M Donaldson
- The Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenmin Yuan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emeka B Okeke
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biology, State University of New York, Fredonia, NY 14063, USA
| | - Jutaek Nam
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yao Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Priyan Weerappuli
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Taryn Hetrick
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - David Adams
- Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick A Lester
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robert A Seder
- The Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA..
| |
Collapse
|
30
|
Chen XL, Wang JH, Zhao W, Shi CW, Yang KD, Niu TM, Yang GL, Cao X, Jiang YL, Wang JZ, Huang HB, Zeng Y, Wang N, Yang WT, Wang CF. Lactobacillus plantarum surface-displayed ASFV (p54) with porcine IL-21 generally stimulates protective immune responses in mice. AMB Express 2021; 11:114. [PMID: 34383171 PMCID: PMC8360262 DOI: 10.1186/s13568-021-01275-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
African classical swine fever virus (ASFV) has spread seriously around the world and has dealt with a heavy blow to the pig breeding industry due to the lack of vaccines. In this study, we produced recombinant Lactobacillus plantarum (L. plantarum) expressing an ASFV p54 and porcine IL-21 (pIL-21) fusion protein and evaluated the immune effect of NC8-pSIP409-pgsA'-p54-pIL-21 in a mouse model. First, we verified that the ASFV p54 protein and p54-pIL-21 fusion protein were anchored on the surface of L. plantarum NC8 by flow cytometry, immunofluorescence and Western blotting. Then, the results were verified by flow cytometry, ELISA and MTT assays. Mouse-specific humoral immunity and mucosal and T cell-mediated immune responses were induced by recombinant L. plantarum. The results of feeding mice recombinant L. plantarum showed that the levels of serum IgG and mucosal secreted IgA (SIgA), the number of CD4 and CD8 T cells, and the expression of IFN-γ in CD4 and CD8 T cells increased significantly, and lymphocyte proliferation occurred under stimulation with the ASFV p54 protein. Our data lay a foundation for the development of oral vaccines against ASFV in the future.
Collapse
|
31
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Pino M, Abid T, Pereira Ribeiro S, Edara VV, Floyd K, Smith JC, Latif MB, Pacheco-Sanchez G, Dutta D, Wang S, Gumber S, Kirejczyk S, Cohen J, Stammen RL, Jean SM, Wood JS, Connor-Stroud F, Pollet J, Chen WH, Wei J, Zhan B, Lee J, Liu Z, Strych U, Shenvi N, Easley K, Weiskopf D, Sette A, Pollara J, Mielke D, Gao H, Eisel N, LaBranche CC, Shen X, Ferrari G, Tomaras GD, Montefiori DC, Sekaly RP, Vanderford TH, Tomai MA, Fox CB, Suthar MS, Kozlowski PA, Hotez PJ, Paiardini M, Bottazzi ME, Kasturi SP. A yeast expressed RBD-based SARS-CoV-2 vaccine formulated with 3M-052-alum adjuvant promotes protective efficacy in non-human primates. Sci Immunol 2021; 6:eabh3634. [PMID: 34266981 PMCID: PMC9119307 DOI: 10.1126/sciimmunol.abh3634] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/26/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Ongoing SARS-CoV-2 vaccine development is focused on identifying stable, cost-effective, and accessible candidates for global use, specifically in low and middle-income countries. Here, we report the efficacy of a rapidly scalable, novel yeast expressed SARS-CoV-2 specific receptor-binding domain (RBD) based vaccine in rhesus macaques. We formulated the RBD immunogen in alum, a licensed and an emerging alum adsorbed TLR-7/8 targeted, 3M-052-alum adjuvants. The RBD+3M-052-alum adjuvanted vaccine promoted better RBD binding and effector antibodies, higher CoV-2 neutralizing antibodies, improved Th1 biased CD4+T cell reactions, and increased CD8+ T cell responses when compared to the alum-alone adjuvanted vaccine. RBD+3M-052-alum induced a significant reduction of SARS-CoV-2 virus in respiratory tract upon challenge, accompanied by reduced lung inflammation when compared with unvaccinated controls. Anti-RBD antibody responses in vaccinated animals inversely correlated with viral load in nasal secretions and BAL. RBD+3M-052-alum blocked a post SARS-CoV-2 challenge increase in CD14+CD16++ intermediate blood monocytes, and Fractalkine, MCP-1, and TRAIL in the plasma. Decreased plasma analytes and intermediate monocyte frequencies correlated with reduced nasal and BAL viral loads. Lastly, RBD-specific plasma cells accumulated in the draining lymph nodes and not in the bone marrow, contrary to previous findings. Together, these data show that a yeast expressed, RBD-based vaccine+3M-052-alum provides robust immune responses and protection against SARS-CoV-2, making it a strong and scalable vaccine candidate.
Collapse
Affiliation(s)
- Maria Pino
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Talha Abid
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Susan Pereira Ribeiro
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Venkata Viswanadh Edara
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Katharine Floyd
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, U.S.A
| | - Muhammad Bilal Latif
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Gabriela Pacheco-Sanchez
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Debashis Dutta
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Shelly Wang
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Sanjeev Gumber
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Shannon Kirejczyk
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce Cohen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rachelle L Stammen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sherrie M Jean
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Fawn Connor-Stroud
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jeroen Pollet
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Wen-Hsiang Chen
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Junfei Wei
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Bin Zhan
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Jungsoon Lee
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Zhuyun Liu
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Ulrich Strych
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Neeta Shenvi
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, U.S.A
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, U.S.A
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Justin Pollara
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Dieter Mielke
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Hongmei Gao
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Nathan Eisel
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - David C Montefiori
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Rafick P Sekaly
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Thomas H Vanderford
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Mark A Tomai
- 3M Corporate Research Materials Laboratory, St. Paul, MN, USA
| | | | - Mehul S Suthar
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, U.S.A
| | - Peter J Hotez
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Maria Elena Bottazzi
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A.
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Sudhir Pai Kasturi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| |
Collapse
|
33
|
Wang JH, Shi CW, Lu YY, Zeng Y, Cheng MY, Wang RY, Sun Y, Jiang YL, Yang WT, Zhao DD, Huang HB, Ye LP, Cao X, Yang GL, Wang CF. MicroRNA and circRNA Expression Analysis in a Zbtb1 Gene Knockout Monoclonal EL4 Cell Line. Front Cell Infect Microbiol 2021; 11:706919. [PMID: 34290994 PMCID: PMC8287301 DOI: 10.3389/fcimb.2021.706919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022] Open
Abstract
Zinc finger and BTB domain containing 1(Zbtb1) is a transcriptional suppressor protein, and a member of the mammalian Zbtb gene family. Previous studies have shown that Zbtb1 is essential for T-cell development. However, the role of Zbtb1 in T-cell lymphoma is undetermined. In this study, an EL4 cell line with Zbtb1 deletion was constructed using the CRISPR-Cas9 technique. The expression profiles of microRNA and circRNA produced by the control and gene deletion groups were determined by RNA-seq. In general, 24 differentially expressed microRNA and 16 differentially expressed circRNA were found between normal group and gene deletion group. Through further analysis of differentially expressed genes, GO term histogram and KEGG scatter plot were drawn, and three pairs of miRNA and circRNA regulatory relationships were found. This study describes the differentially expressed microRNA and circRNA in normal and Zbtb1-deficient EL4 cell lines, thus providing potential targets for drug development and clinical treatment of T-cell lymphoma.
Collapse
Affiliation(s)
- Jun-Hong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yi-Yuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming-Yang Cheng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ru-Yu Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Dan-Dan Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Li-Ping Ye
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
34
|
Pulendran B, S Arunachalam P, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454-475. [PMID: 33824489 PMCID: PMC8023785 DOI: 10.1038/s41573-021-00163-y] [Citation(s) in RCA: 809] [Impact Index Per Article: 202.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Adjuvants are vaccine components that enhance the magnitude, breadth and durability of the immune response. Following its introduction in the 1920s, alum remained the only adjuvant licensed for human use for the next 70 years. Since the 1990s, a further five adjuvants have been included in licensed vaccines, but the molecular mechanisms by which these adjuvants work remain only partially understood. However, a revolution in our understanding of the activation of the innate immune system through pattern recognition receptors (PRRs) is improving the mechanistic understanding of adjuvants, and recent conceptual advances highlight the notion that tissue damage, different forms of cell death, and metabolic and nutrient sensors can all modulate the innate immune system to activate adaptive immunity. Furthermore, recent advances in the use of systems biology to probe the molecular networks driving immune response to vaccines ('systems vaccinology') are revealing mechanistic insights and providing a new paradigm for the vaccine discovery and development process. Here, we review the 'known knowns' and 'known unknowns' of adjuvants, discuss these emerging concepts and highlight how our expanding knowledge about innate immunity and systems vaccinology are revitalizing the science and development of novel adjuvants for use in vaccines against COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering & Medicine for Human Health, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
35
|
Seaton KE, Deal A, Han X, Li SS, Clayton A, Heptinstall J, Duerr A, Allen MA, Shen X, Sawant S, Yates NL, Spearman P, Churchyard G, Goepfert PA, Maenza J, Gray G, Pantaleo G, Polakowski L, Robinson HL, Grant S, Randhawa AK, Huang Y, Morgan C, Grunenberg N, Karuna S, Gilbert PB, McElrath MJ, Huang Y, Tomaras GD. Meta-analysis of HIV-1 vaccine elicited mucosal antibodies in humans. NPJ Vaccines 2021; 6:56. [PMID: 33859204 PMCID: PMC8050318 DOI: 10.1038/s41541-021-00305-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
We studied mucosal immune responses in six HIV-1 vaccine trials investigating different envelope (Env)-containing immunogens. Regimens were classified into four categories: DNA/vector, DNA/vector plus protein, protein alone, and vector alone. We measured HIV-1-specific IgG and IgA in secretions from cervical (n = 111) and rectal swabs (n = 154), saliva (n = 141), and seminal plasma (n = 124) and compared to corresponding blood levels. Protein-containing regimens had up to 100% response rates and the highest Env-specific IgG response rates. DNA/vector groups elicited mucosal Env-specific IgG response rates of up to 67% that varied across specimen types. Little to no mucosal IgA responses were observed. Overall, gp41- and gp140-specific antibodies dominated gp120 mucosal responses. In one trial, prior vaccination with a protein-containing immunogen maintained durability of cervical and rectal IgG for up to 17 years. Mucosal IgG responses were boosted after revaccination. These findings highlight a role for protein immunization in eliciting HIV-1-specific mucosal antibodies and the ability of HIV-1 vaccines to elicit durable HIV-1-specific mucosal IgG.
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Human Vaccine Institute, Durham, NC, USA.
- Department of Surgery, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.
| | - Aaron Deal
- Duke Human Vaccine Institute, Durham, NC, USA
| | - Xue Han
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shuying S Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ashley Clayton
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jack Heptinstall
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Ann Duerr
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Sheetal Sawant
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Nicole L Yates
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Paul Spearman
- Division of Infectious Diseases, Cincinnati Children's Hospital, Cincinnatti, OH, USA
| | - Gavin Churchyard
- Aurum Institute, Johannesburg, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Glenda Gray
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- South African Medical Research Council, Cape Town, South Africa
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, and Swiss Vaccine Research Institute, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | - Shannon Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Cecilia Morgan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC, USA.
- Department of Surgery, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.
| |
Collapse
|
36
|
Knight FC, Wilson JT. Engineering Vaccines for Tissue-Resident Memory T Cells. ADVANCED THERAPEUTICS 2021; 4:2000230. [PMID: 33997268 PMCID: PMC8114897 DOI: 10.1002/adtp.202000230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Indexed: 01/01/2023]
Abstract
In recent years, tissue-resident memory T cells (TRM) have attracted significant attention in the field of vaccine development. Distinct from central and effector memory T cells, TRM cells take up residence in home tissues such as the lung or urogenital tract and are ideally positioned to respond quickly to pathogen encounter. TRM have been found to play a role in the immune response against many globally important infectious diseases for which new or improved vaccines are needed, including influenza and tuberculosis. It is also increasingly clear that TRM play a pivotal role in cancer immunity. Thus, vaccines that can generate this memory T cell population are highly desirable. The field of immunoengineering-that is, the application of engineering principles to study the immune system and design new and improved therapies that harness or modulate immune responses-is ideally poised to provide solutions to this need for next-generation TRM vaccines. This review covers recent developments in vaccine technologies for generating TRM and protecting against infection and cancer, including viral vectors, virus-like particles, and synthetic and natural biomaterials. In addition, it offers critical insights on the future of engineering vaccines for tissue-resident memory T cells.
Collapse
Affiliation(s)
- Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
37
|
Saunders KO, Lee E, Parks R, Martinez DR, Li D, Chen H, Edwards RJ, Gobeil S, Barr M, Mansouri K, Alam SM, Sutherland LL, Cai F, Sanzone AM, Berry M, Manne K, Kapingidza AB, Azoitei M, Tse LV, Scobey TD, Spreng RL, Rountree RW, DeMarco CT, Denny TN, Woods CW, Petzold EW, Oguin TH, Sempowski GD, Gagne M, Douek DC, Tomai MA, Fox CB, Seder R, Wiehe K, Weissman D, Pardi N, Acharya P, Andersen H, Lewis MG, Moore IN, Montefiori DC, Baric RS, Haynes BF. SARS-CoV-2 vaccination induces neutralizing antibodies against pandemic and pre-emergent SARS-related coronaviruses in monkeys. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.02.17.431492. [PMID: 33619494 PMCID: PMC7899458 DOI: 10.1101/2021.02.17.431492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Betacoronaviruses (betaCoVs) caused the severe acute respiratory syndrome (SARS) and Middle East Respiratory Syndrome (MERS) outbreaks, and now the SARS-CoV-2 pandemic. Vaccines that elicit protective immune responses against SARS-CoV-2 and betaCoVs circulating in animals have the potential to prevent future betaCoV pandemics. Here, we show that immunization of macaques with a multimeric SARS-CoV-2 receptor binding domain (RBD) nanoparticle adjuvanted with 3M-052-Alum elicited cross-neutralizing antibody responses against SARS-CoV-1, SARS-CoV-2, batCoVs and the UK B.1.1.7 SARS-CoV-2 mutant virus. Nanoparticle vaccination resulted in a SARS-CoV-2 reciprocal geometric mean neutralization titer of 47,216, and robust protection against SARS-CoV-2 in macaque upper and lower respiratory tracts. Importantly, nucleoside-modified mRNA encoding a stabilized transmembrane spike or monomeric RBD protein also induced SARS-CoV-1 and batCoV cross-neutralizing antibodies, albeit at lower titers. These results demonstrate current mRNA vaccines may provide some protection from future zoonotic betaCoV outbreaks, and provide a platform for further development of pan-betaCoV nanoparticle vaccines.
Collapse
|
38
|
Kasturi SP, Rasheed MAU, Havenar-Daughton C, Pham M, Legere T, Sher ZJ, Kovalenkov Y, Gumber S, Huang JY, Gottardo R, Fulp W, Sato A, Sawant S, Stanfield-Oakley S, Yates N, LaBranche C, Alam SM, Tomaras G, Ferrari G, Montefiori D, Wrammert J, Villinger F, Tomai M, Vasilakos J, Fox CB, Reed SG, Haynes BF, Crotty S, Ahmed R, Pulendran B. 3M-052, a synthetic TLR-7/8 agonist, induces durable HIV-1 envelope-specific plasma cells and humoral immunity in nonhuman primates. Sci Immunol 2021; 5:5/48/eabb1025. [PMID: 32561559 DOI: 10.1126/sciimmunol.abb1025] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022]
Abstract
A fundamental challenge in vaccinology is learning how to induce durable antibody responses. Live viral vaccines induce antibody responses that last a lifetime, but those induced with subunit vaccines wane rapidly. Studies in mice and humans have established that long-lived plasma cells (LLPCs) in the bone marrow (BM) are critical mediators of durable antibody responses. Here, we present data that adjuvanting an HIV-1 clade C 1086.C-derived gp140 immunogen (Env) with a novel synthetic Toll-like receptor (TLR)-7/8 agonist named 3M-052 formulated in poly(lactic-co-glycolic)acid or PLGA nanoparticles (NPs) or with alum, either alone or in combination with a TLR-4 agonist GLA, induces notably high and persistent (up to ~1 year) frequencies of Env-specific LLPCs in the BM and serum antibody responses in rhesus macaques. Up to 36 and 18% of Env-specific cells among total IgG-secreting BM-resident plasma cells were detected at peak and termination, respectively. In contrast, adjuvanting Env with alum or GLA in NP induced significantly lower (~<100-fold) LLPC and antibody responses. Immune responses induced by 3M-052 were also significantly higher than those induced by a combination of TLR-7/8 (R848) and TLR-4 (MPL) agonists. Adjuvanting Env with 3M-052 also induced robust activation of blood monocytes, strong plasmablast responses in blood, germinal center B cells, T follicular helper (TFH) cells, and persistent Env-specific plasma cells in draining lymph nodes. Overall, these results demonstrate efficacy of 3M-052 in promoting high magnitude and durability of antibody responses via robust stimulation of innate immunity and BM-resident LLPCs.
Collapse
Affiliation(s)
- Sudhir Pai Kasturi
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Mohammed Ata Ur Rasheed
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA.,Division of Microbiology and Immunology and Rollins Research Center, Emory University, 1510 Clifton Road, Atlanta, GA, USA
| | | | - Mathew Pham
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Traci Legere
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Zarpheen Jinnah Sher
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Yevgeny Kovalenkov
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Sanjeev Gumber
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Jessica Y Huang
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - William Fulp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alicia Sato
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sheetal Sawant
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA.,Department of Molecular Genetics and Microbiology and Department of Immunology, Duke University, NC, USA
| | - Sherry Stanfield-Oakley
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Nicole Yates
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA.,Department of Molecular Genetics and Microbiology and Department of Immunology, Duke University, NC, USA
| | - Celia LaBranche
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Georgia Tomaras
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA.,Department of Molecular Genetics and Microbiology and Department of Immunology, Duke University, NC, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA.,Department of Molecular Genetics and Microbiology and Department of Immunology, Duke University, NC, USA
| | - David Montefiori
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Jens Wrammert
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA
| | - Francois Villinger
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA.,New Iberia Research Center, University of Louisiana Lafayette, New Iberia, LA, USA
| | - Mark Tomai
- 3M Drug Delivery Systems, St. Paul, MN, USA
| | | | - Christopher B Fox
- Infectious Disease Research Institute, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA, USA.,HDT Bio, Seattle, WA, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute of Immunology, La Jolla, CA, USA.,Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA. .,Division of Microbiology and Immunology and Rollins Research Center, Emory University, 1510 Clifton Road, Atlanta, GA, USA
| | - Bali Pulendran
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, 954, Gatewood Road, Atlanta, GA, USA. .,Departments of Pathology and Microbiology & Immunology, Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| |
Collapse
|
39
|
Abstract
The way to a successful vaccine against human cytomegalovirus is hampered by the peculiar biology of this infection. However, some candidate vaccines have been shown to protect seronegative women and transplant recipients, and we should know soon whether they can prevent congenital infection.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania, Vaxconsult, Doylestown, Pennsylvania, USA
| |
Collapse
|
40
|
Kusnadi A, Ramírez-Suástegui C, Fajardo V, Chee SJ, Meckiff BJ, Simon H, Pelosi E, Seumois G, Ay F, Vijayanand P, Ottensmeier CH. Severely ill COVID-19 patients display impaired exhaustion features in SARS-CoV-2-reactive CD8 + T cells. Sci Immunol 2021; 6:eabe4782. [PMID: 33478949 PMCID: PMC8101257 DOI: 10.1126/sciimmunol.abe4782] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
The molecular properties of CD8+ T cells that respond to SARS-CoV-2 infection are not fully known. Here, we report on the single-cell transcriptomes of >80,000 virus-reactive CD8+ T cells, obtained using a modified Antigen-Reactive T cell Enrichment (ARTE) assay, from 39 COVID-19 patients and 10 healthy subjects. COVID-19 patients segregated into two groups based on whether the dominant CD8+ T cell response to SARS-CoV-2 was 'exhausted' or not. SARS-CoV-2-reactive cells in the exhausted subset were increased in frequency and displayed lesser cytotoxicity and inflammatory features in COVID-19 patients with mild compared to severe illness. In contrast, SARS-CoV-2-reactive cells in the dominant non-exhausted subset from patients with severe disease showed enrichment of transcripts linked to co-stimulation, pro-survival NF-κB signaling, and anti-apoptotic pathways, suggesting the generation of robust CD8+ T cell memory responses in patients with severe COVID-19 illness. CD8+ T cells reactive to influenza and respiratory syncytial virus from healthy subjects displayed polyfunctional features and enhanced glycolysis. Cells with such features were largely absent in SARS-CoV-2-reactive cells from both COVID-19 patients and healthy controls non-exposed to SARS-CoV-2. Overall, our single-cell analysis revealed substantial diversity in the nature of CD8+ T cells responding to SARS-CoV-2.
Collapse
Affiliation(s)
| | | | | | - Serena J Chee
- NIHR and CRUK Southampton Experimental Cancer Medicine Center, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Hayley Simon
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Emanuela Pelosi
- Southampton Specialist Virology Centre, Department of Infection, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Ferhat Ay
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA 92037.
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, UK
- Department of Medicine, University of California San Diego, La Jolla, CA 92037
| | - Christian H Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA 92037.
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
41
|
Koguchi-Yoshioka H, Hoffer E, Cheuk S, Matsumura Y, Vo S, Kjellman P, Grema L, Ishitsuka Y, Nakamura Y, Okiyama N, Fujisawa Y, Fujimoto M, Eidsmo L, Clark RA, Watanabe R. Skin T cells maintain their diversity and functionality in the elderly. Commun Biol 2021; 4:13. [PMID: 33398080 PMCID: PMC7782613 DOI: 10.1038/s42003-020-01551-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/27/2020] [Indexed: 01/23/2023] Open
Abstract
Recent studies have highlighted that human resident memory T cells (TRM) are functionally distinct from circulating T cells. Thus, it can be postulated that skin T cells age differently from blood-circulating T cells. We assessed T-cell density, diversity, and function in individuals of various ages to study the immunologic effects of aging on human skin from two different countries. No decline in the density of T cells was noted with advancing age, and the frequency of epidermal CD49a+ CD8 TRM was increased in elderly individuals regardless of ethnicity. T-cell diversity and antipathogen responses were maintained in the skin of elderly individuals but declined in the blood. Our findings demonstrate that in elderly individuals, skin T cells maintain their density, diversity, and protective cytokine production despite the reduced T-cell diversity and function in blood. Skin resident T cells may represent a long-lived, highly protective reservoir of immunity in elderly people.
Collapse
Affiliation(s)
- Hanako Koguchi-Yoshioka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.,Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Elena Hoffer
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Visionsgatan 18, L8, 171 76, Solna, Sweden
| | - Stanley Cheuk
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Visionsgatan 18, L8, 171 76, Solna, Sweden
| | - Yutaka Matsumura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Sa Vo
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Petra Kjellman
- Diagnostiskt Centrum Hud, Apelbergsgatan 60, 111 37, Stockholm, Sweden
| | - Lucian Grema
- Diagnostiskt Centrum Hud, Apelbergsgatan 60, 111 37, Stockholm, Sweden
| | - Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.,Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Liv Eidsmo
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Visionsgatan 18, L8, 171 76, Solna, Sweden.,Diagnostiskt Centrum Hud, Apelbergsgatan 60, 111 37, Stockholm, Sweden
| | - Rachael A Clark
- Department of Dermatology, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Rei Watanabe
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan. .,Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
42
|
|
43
|
Om K, Paquin-Proulx D, Montero M, Peachman K, Shen X, Wieczorek L, Beck Z, Weiner JA, Kim D, Li Y, Mdluli T, Shubin Z, Bryant C, Sharma V, Tokarev A, Dawson P, White Y, Appelbe O, Klatt NR, Tovanabutra S, Estes JD, Matyas GR, Ferrari G, Alving CR, Tomaras GD, Ackerman ME, Michael NL, Robb ML, Polonis V, Rolland M, Eller MA, Rao M, Bolton DL. Adjuvanted HIV-1 vaccine promotes antibody-dependent phagocytic responses and protects against heterologous SHIV challenge. PLoS Pathog 2020; 16:e1008764. [PMID: 32881968 PMCID: PMC7505435 DOI: 10.1371/journal.ppat.1008764] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/21/2020] [Accepted: 06/30/2020] [Indexed: 01/29/2023] Open
Abstract
To augment HIV-1 pox-protein vaccine immunogenicity using a next generation adjuvant, a prime-boost strategy of recombinant modified vaccinia virus Ankara and multimeric Env gp145 was evaluated in macaques with either aluminum (alum) or a novel liposomal monophosphoryl lipid A (MPLA) formulation adsorbed to alum, ALFA. Binding antibody responses were robust and comparable between arms, while antibody-dependent neutrophil and monocyte phagocytotic responses were greatly enhanced by ALFA. Per-exposure vaccine efficacy against heterologous tier 2 SHIV mucosal challenge was 90% in ALFA-adjuvanted males (P = 0.002), while alum conferred no protection. Half of the ALFA-adjuvanted males remained uninfected after the full challenge series, which spanned seven months after the last vaccination. Antibody-dependent monocyte and neutrophil phagocytic responses both strongly correlated with protection. Significant sex differences in infection risk were observed, with much lower infection rates in females than males. In humans, MPLA-liposome-alum adjuvanted gp120 also increased HIV-1-specific phagocytic responses relative to alum. Thus, next-generation liposome-based adjuvants can drive vaccine elicited antibody effector activity towards potent phagocytic responses in both macaques and humans and these responses correlate with protection. Future protein vaccination strategies aiming to improve functional humoral responses may benefit from such adjuvants.
Collapse
Affiliation(s)
- Kier Om
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Dominic Paquin-Proulx
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Maria Montero
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Kristina Peachman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lindsay Wieczorek
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Zoltan Beck
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Joshua A. Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Dohoon Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Yifan Li
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Thembi Mdluli
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Zhanna Shubin
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | | | - Vishakha Sharma
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Andrey Tokarev
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Peter Dawson
- EMMES, Rockville, Maryland, United States of America
| | - Yohann White
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Oliver Appelbe
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Nichole R. Klatt
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Sodsai Tovanabutra
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, United States of America
| | - Gary R. Matyas
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Carl R. Alving
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Nelson L. Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Merlin L. Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Victoria Polonis
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Morgane Rolland
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Michael A. Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Mangala Rao
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Diane L. Bolton
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| |
Collapse
|
44
|
Kusnadi A, Ramírez-Suástegui C, Fajardo V, Chee SJ, Meckiff BJ, Simon H, Pelosi E, Seumois G, Ay F, Vijayanand P, Ottensmeier CH. Severely ill COVID-19 patients display augmented functional properties in SARS-CoV-2-reactive CD8 + T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.09.194027. [PMID: 32676602 PMCID: PMC7359524 DOI: 10.1101/2020.07.09.194027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The molecular properties of CD8 + T cells that respond to SARS-CoV-2 infection are not fully known. Here, we report on the single-cell transcriptomes of >80,000 virus-reactive CD8 + T cells from 39 COVID-19 patients and 10 healthy subjects. COVID-19 patients segregated into two groups based on whether the dominant CD8 + T cell response to SARS-CoV-2 was 'exhausted' or not. SARS-CoV-2-reactive cells in the exhausted subset were increased in frequency and displayed lesser cytotoxicity and inflammatory features in COVID-19 patients with mild compared to severe illness. In contrast, SARS-CoV-2-reactive cells in the non-exhausted subsets from patients with severe disease showed enrichment of transcripts linked to co-stimulation, pro-survival NF-κB signaling, and anti-apoptotic pathways, suggesting the generation of robust CD8 + T cell memory responses in patients with severe COVID-19 illness. CD8 + T cells reactive to influenza and respiratory syncytial virus from healthy subjects displayed polyfunctional features. Cells with such features were mostly absent in SARS-CoV-2 responsive cells from both COVID-19 patients and healthy controls non-exposed to SARS-CoV-2. Overall, our single-cell analysis revealed substantial diversity in the nature of CD8 + T cells responding to SARS-CoV-2.
Collapse
Affiliation(s)
- Anthony Kusnadi
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Ciro Ramírez-Suástegui
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Vicente Fajardo
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Serena J Chee
- NIHR and CRUK Southampton Experimental Cancer Medicine Center, Faculty of Medicine, University of Southampton, Southampton, UK
- These authors jointly contributed to the work
| | | | - Hayley Simon
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Emanuela Pelosi
- Southampton Specialist Virology Centre, Department of Infection, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Ferhat Ay
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, United Kingdom
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- These authors jointly directed the work
| | - Christian H Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, United Kingdom
- These authors jointly directed the work
| |
Collapse
|
45
|
Kim EH, Woodruff MC, Grigoryan L, Maier B, Lee SH, Mandal P, Cortese M, Natrajan MS, Ravindran R, Ma H, Merad M, Gitlin AD, Mocarski ES, Jacob J, Pulendran B. Squalene emulsion-based vaccine adjuvants stimulate CD8 T cell, but not antibody responses, through a RIPK3-dependent pathway. eLife 2020; 9:52687. [PMID: 32515732 PMCID: PMC7314549 DOI: 10.7554/elife.52687] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/08/2020] [Indexed: 12/26/2022] Open
Abstract
The squalene-based oil-in-water emulsion (SE) vaccine adjuvant MF59 has been administered to more than 100 million people in more than 30 countries, in both seasonal and pandemic influenza vaccines. Despite its wide use and efficacy, its mechanisms of action remain unclear. In this study we demonstrate that immunization of mice with MF59 or its mimetic AddaVax (AV) plus soluble antigen results in robust antigen-specific antibody and CD8 T cell responses in lymph nodes and non-lymphoid tissues. Immunization triggered rapid RIPK3-kinase dependent necroptosis in the lymph node which peaked at 6 hr, followed by a sequential wave of apoptosis. Immunization with alum plus antigen did not induce RIPK3-dependent signaling. RIPK3-dependent signaling induced by MF59 or AV was essential for cross-presentation of antigen to CD8 T cells by Batf3-dependent CD8+ DCs. Consistent with this, RIPK3 deficient or Batf3 deficient mice were impaired in their ability to mount adjuvant-enhanced CD8 T cell responses. However, CD8 T cell responses were unaffected in mice deficient in MLKL, a downstream mediator of necroptosis. Surprisingly, antibody responses were unaffected in RIPK3-kinase or Batf3 deficient mice. In contrast, antibody responses were impaired by in vivo administration of the pan-caspase inhibitor Z-VAD-FMK, but normal in caspase-1 deficient mice, suggesting a contribution from apoptotic caspases, in the induction of antibody responses. These results demonstrate that squalene emulsion-based vaccine adjuvants induce antigen-specific CD8 T cell and antibody responses, through RIPK3-dependent and-independent pathways, respectively.
Collapse
Affiliation(s)
- Eui Ho Kim
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States.,Viral Immunology Laboratory, Institut Pasteur Korea, Seongnam, Republic of Korea
| | - Matthew C Woodruff
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Barbara Maier
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, United States
| | - Song Hee Lee
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Pratyusha Mandal
- Emory Vaccine Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | | | - Rajesh Ravindran
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States
| | - Huailiang Ma
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, United States
| | - Alexander D Gitlin
- Department of Physiological Chemistry, Genentech, South San Francisco, United States.,Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, United States
| | - Edward S Mocarski
- Emory Vaccine Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Joshy Jacob
- Emory Vaccine Center, Emory University, Atlanta, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, United States.,Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, United States
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, United States.,Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, United States
| |
Collapse
|
46
|
Arunachalam PS, Charles TP, Joag V, Bollimpelli VS, Scott MKD, Wimmers F, Burton SL, Labranche CC, Petitdemange C, Gangadhara S, Styles TM, Quarnstrom CF, Walter KA, Ketas TJ, Legere T, Jagadeesh Reddy PB, Kasturi SP, Tsai A, Yeung BZ, Gupta S, Tomai M, Vasilakos J, Shaw GM, Kang CY, Moore JP, Subramaniam S, Khatri P, Montefiori D, Kozlowski PA, Derdeyn CA, Hunter E, Masopust D, Amara RR, Pulendran B. T cell-inducing vaccine durably prevents mucosal SHIV infection even with lower neutralizing antibody titers. Nat Med 2020; 26:932-940. [PMID: 32393800 PMCID: PMC7303014 DOI: 10.1038/s41591-020-0858-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/27/2020] [Indexed: 01/05/2023]
Abstract
Recent efforts toward an HIV vaccine focus on inducing broadly neutralizing antibodies, but eliciting both neutralizing antibodies (nAbs) and cellular responses may be superior. Here, we immunized macaques with an HIV envelope trimer, either alone to induce nAbs, or together with a heterologous viral vector regimen to elicit nAbs and cellular immunity, including CD8+ tissue-resident memory T cells. After ten vaginal challenges with autologous virus, protection was observed in both vaccine groups at 53.3% and 66.7%, respectively. A nAb titer >300 was generally associated with protection but in the heterologous viral vector + nAb group, titers <300 were sufficient. In this group, protection was durable as the animals resisted six more challenges 5 months later. Antigen stimulation of T cells in ex vivo vaginal tissue cultures triggered antiviral responses in myeloid and CD4+ T cells. We propose that cellular immune responses reduce the threshold of nAbs required to confer superior and durable protection.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/drug effects
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/drug effects
- Antibodies, Viral/immunology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Genetic Vectors
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Heterologous
- Immunogenicity, Vaccine
- Immunologic Memory/immunology
- Macaca mulatta
- Mucous Membrane
- SAIDS Vaccines/pharmacology
- Simian Acquired Immunodeficiency Syndrome/prevention & control
- Simian Immunodeficiency Virus/immunology
- Vagina
Collapse
Affiliation(s)
- Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Tysheena P Charles
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Vineet Joag
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Venkata S Bollimpelli
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
| | - Madeleine K D Scott
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Center for Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Samantha L Burton
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Celia C Labranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Caroline Petitdemange
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
- HIV Inflammation and Persistence Unit, Institut Pasteur, Paris, France
| | - Sailaja Gangadhara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
| | - Tiffany M Styles
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
| | - Clare F Quarnstrom
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Korey A Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Thomas J Ketas
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Traci Legere
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
| | - Pradeep Babu Jagadeesh Reddy
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
- Pfizer, Andover, MA, USA
| | - Sudhir Pai Kasturi
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA
| | | | | | - Shakti Gupta
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Mark Tomai
- 3M Corporate Research and Materials Lab, Saint Paul, MN, USA
| | | | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chil-Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - John P Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Center for Biomedical Informatics, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Cynthia A Derdeyn
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA.
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA.
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center at Emory University, Atlanta, GA, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
47
|
Helmold Hait S, Hogge CJ, Rahman MA, Ko EJ, Hunegnaw R, Mushtaq Z, Enyindah-Asonye G, Hoang T, Miller Jenkins LM, Appella E, Appella DH, Robert-Guroff M. An SAMT-247 Microbicide Provides Potent Protection against Intravaginal Simian Immunodeficiency Virus Infection of Rhesus Macaques, whereas an Added Vaccine Component Elicits Mixed Outcomes. THE JOURNAL OF IMMUNOLOGY 2020; 204:3315-3328. [PMID: 32393514 DOI: 10.4049/jimmunol.2000165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/17/2020] [Indexed: 12/19/2022]
Abstract
Because of microbicide noncompliance and lack of a durable, highly effective vaccine, a combined approach might improve HIV prophylaxis. We tested whether a vaccine-microbicide combination would enhance protection against SIV infection in rhesus macaques. Four macaque groups included vaccine only, vaccine-microbicide, microbicide only, and controls. Vaccine groups were primed twice mucosally with replicating adenovirus type 5 host range mutant SIV env/rev, gag, and nef recombinants and boosted twice i.m. with SIV gp120 proteins in alum. Controls and the microbicide-only group received adenovirus type 5 host range mutant empty vector and alum. The microbicide was SAMT-247, a 2-mercaptobenzamide thioester that targets the viral nucleocapsid protein NCp7, causing zinc ejection and preventing RNA encapsidation. Following vaccination, macaques were challenged intravaginally with repeated weekly low doses of SIVmac251 administered 3 h after application of 0.8% SAMT-247 gel (vaccine-microbicide and microbicide groups) or placebo gel (vaccine-only and control groups). The microbicide-only group exhibited potent protection; 10 of 12 macaques remained uninfected following 15 SIV challenges. The vaccine-only group developed strong mucosal and systemic humoral and cellular immunity but did not exhibit delayed acquisition compared with adjuvant controls. However, the vaccine-microbicide group exhibited significant acquisition delay compared with both control and vaccine-only groups, indicating further exploration of the combination strategy is warranted. Impaired protection in the vaccine-microbicide group compared with the microbicide-only group was not attributed to a vaccine-induced increase in SIV target cells. Possible Ab-dependent enhancement will be further investigated. The potent protection provided by SAMT-247 encourages its movement into human clinical trials.
Collapse
Affiliation(s)
- Sabrina Helmold Hait
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Christopher James Hogge
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Mohammad Arif Rahman
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Eun-Ju Ko
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Ruth Hunegnaw
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Zuena Mushtaq
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Gospel Enyindah-Asonye
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Tanya Hoang
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4256; and
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4256; and
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, Synthetic Bioactive Molecules Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0820
| | - Marjorie Robert-Guroff
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5065;
| |
Collapse
|
48
|
Shacklett BL, Ferre AL, Kiniry BE. Defining T Cell Tissue Residency in Humans: Implications for HIV Pathogenesis and Vaccine Design. Curr HIV/AIDS Rep 2020; 17:109-117. [PMID: 32052270 PMCID: PMC7072053 DOI: 10.1007/s11904-020-00481-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW This review summarizes recent literature defining tissue-resident memory T cells (TRM) and discusses implications for HIV pathogenesis, vaccines, and eradication efforts. RECENT FINDINGS Investigations using animal models and human tissues have identified a TRM transcriptional profile and elucidated signals within the tissue microenvironment leading to TRM development and maintenance. TRM are major contributors to host response in infectious diseases and cancer; in addition, TRM contribute to pathogenic inflammation in a variety of settings. Although TRM are daunting to study in HIV infection, recent work has helped define their molecular signatures and effector functions and tested strategies for their mobilization. Exclusive reliance on blood sampling to gain an understanding of host immunity overlooks the contribution of TRM, which differ in significant ways from their counterparts in circulation. It is hoped that greater understanding of these cells will lead to novel approaches to prevent and/or eradicate HIV infection.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA.
- Division of Infectious Disease, Department of Medicine, School of Medicine, University of California, Davis, CA, 95616, USA.
| | - April L Ferre
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Brenna E Kiniry
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, 95616, USA
| |
Collapse
|
49
|
Salinas E, Grakoui A. Monkey in the Middle: Dynamics of Interspecies Antibody Responses. Gastroenterology 2020; 158:820-822. [PMID: 31926168 DOI: 10.1053/j.gastro.2020.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/03/2020] [Indexed: 12/02/2022]
Affiliation(s)
- Eduardo Salinas
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University, Atlanta, Georgia
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University and, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
50
|
Verma A, Schmidt BA, Elizaldi SR, Nguyen NK, Walter KA, Beck Z, Trinh HV, Dinasarapu AR, Lakshmanappa YS, Rane NN, Matyas GR, Rao M, Shen X, Tomaras GD, LaBranche CC, Reimann KA, Foehl DH, Gach JS, Forthal DN, Kozlowski PA, Amara RR, Iyer SS. Impact of T h1 CD4 Follicular Helper T Cell Skewing on Antibody Responses to an HIV-1 Vaccine in Rhesus Macaques. J Virol 2020; 94:e01737-19. [PMID: 31827000 PMCID: PMC7158739 DOI: 10.1128/jvi.01737-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Generating durable humoral immunity through vaccination depends upon effective interactions of follicular helper T (Tfh) cells with germinal center (GC) B cells. Th1 polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing costimulatory and cytokine-dependent Tfh help to B cells. However, the question remains as to whether adjuvant-dependent modulation of Tfh cells enhances HIV-1 vaccine-induced antienvelope (anti-Env) antibody responses. We investigated whether an HIV-1 vaccine platform designed to increase the number of Th1-polarized Tfh cells enhances the magnitude and quality of anti-Env antibodies. Utilizing a novel interferon-induced protein 10 (IP-10)-adjuvanted HIV-1 DNA prime followed by a monophosphoryl lipid A and QS-21 (MPLA+QS-21)-adjuvanted Env protein boost (DIP-10 PALFQ) in macaques, we observed higher anti-Env serum IgG titers with greater cross-clade reactivity, specificity for V1V2, and effector functions than in macaques primed with DNA lacking IP-10 and boosted with MPLA-plus-alum-adjuvanted Env protein (DPALFA) The DIP-10 PALFQ vaccine regimen elicited higher anti-Env IgG1 and lower IgG4 antibody levels in serum, showing for the first time that adjuvants can dramatically impact the IgG subclass profile in macaques. The DIP-10 PALFQ regimen also increased vaginal and rectal IgA antibodies to a greater extent. Within lymph nodes, we observed augmented GC B cell responses and the promotion of Th1 gene expression profiles in GC Tfh cells. The frequency of GC Tfh cells correlated with both the magnitude and avidity of anti-Env serum IgG. Together, these data suggest that adjuvant-induced stimulation of Th1-Tfh cells is an effective strategy for enhancing the magnitude and quality of anti-Env antibody responses.IMPORTANCE The results of the RV144 trial demonstrated that vaccination could prevent HIV transmission in humans and that longevity of anti-Env antibodies may be key to this protection. Efforts to improve upon the prime-boost vaccine regimen used in RV144 have indicated that booster immunizations can increase serum anti-Env antibody titers but only transiently. Poor antibody durability hampers efforts to develop an effective HIV-1 vaccine. This study was designed to identify the specific elements involved in the immunological mechanism necessary to produce robust HIV-1-specific antibodies in rhesus macaques. By clearly defining immune-mediated pathways that improve the magnitude and functionality of the anti-HIV-1 antibody response, we will have the foundation necessary for the rational development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Anil Verma
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Brian A Schmidt
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Sonny R Elizaldi
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- Graduate Group in Immunology, UC Davis, Davis, California, USA
| | - Nancy K Nguyen
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Korey A Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zoltan Beck
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ashok R Dinasarapu
- Emory Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | | | - Niharika N Rane
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Keith A Reimann
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - David H Foehl
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Johannes S Gach
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Smita S Iyer
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- California National Primate Research Center, School of Veterinary Medicine, UC Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|