1
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 DOI: 10.1016/j.cell.2024.07.021] [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: 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
|
2
|
Ji J, Tang T, Zhu M, Wu Z, Zhang J, Shi D, Zhu L, Zhang X, Lu X, Chen L, Yao H. Boosting the immune response in COVID-19 vaccines via an Alum:CpG complex adjuvant. Antiviral Res 2024; 229:105954. [PMID: 38964615 DOI: 10.1016/j.antiviral.2024.105954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/02/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Selecting appropriate adjuvants is crucial for developing an effective vaccine. However, studies on the immune responses triggered by different adjuvants in COVID-19 inactivated vaccines are scarce. Herein, we evaluated the efficacy of Alum, CpG HP021, Alum combined with CpG HP021 (Alum/CpG), or MF-59 adjuvants with COVID-19 inactivated vaccines in K18-hACE2 mice, and compared the different immune responses between K18-hACE2 and BALB/c mice. In K18-hACE2 mice, the Alum/CpG group produced a 6.5-fold increase in anti-receptor-binding domain (RBD) IgG antibody titers compared to the Alum group, and generated a comparable level of antibodies even when the antigen amount was reduced by two-thirds, possibly due to the significant activation of germinal center (GC) structures in the central region of the spleen. Different adjuvants induced a variety of binding antibody isotypes. CpG HP021 and Alum/CpG were biased towards Th1/IgG2c, while Alum and MF-59 were biased toward Th2/IgG1. Cytokines IFN-γ, IL-2, and TNF-α were significantly increased in the culture supernatants of splenocytes specifically stimulated in the Alum/CpG group. The antibody responses in BALB/c mice were similar to those in K18-hACE2 mice, but with lower levels of neutralizing antibodies (NAbs). Notably, the Alum/CpG-adjuvanted inactivated vaccine induced a higher number of T cells secreting IFN-γ and IL-2, increased the percentage of effector memory T (TEM) cells among CD8+ T cells, and effectively protected K18-hACE2 mice from Delta variant challenge. Our results showed that Alum/CpG complex adjuvant significantly enhanced the immune response to inactivated COVID-19 antigens and could induce a long-lasting immune response.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Mice
- Adjuvants, Immunologic/administration & dosage
- Mice, Inbred BALB C
- Alum Compounds/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Adjuvants, Vaccine/administration & dosage
- Female
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Cytokines/immunology
- Humans
Collapse
Affiliation(s)
- Jia Ji
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Taoming Tang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Miaojin Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhigang Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jiale Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Danrong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Linwei Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lei Chen
- Zhejiang Toyouvax Bio-pharmaceutical Co., Ltd., Hangzhou, 311103, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
3
|
Sangeetha Vijayan P, Xavier J, Valappil MP. A review of immune modulators and immunotherapy in infectious diseases. Mol Cell Biochem 2024; 479:1937-1955. [PMID: 37682390 DOI: 10.1007/s11010-023-04825-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/05/2023] [Indexed: 09/09/2023]
Abstract
The human immune system responds to harmful foreign invaders frequently encountered by the body and employs defense mechanisms to counteract such assaults. Various exogenous and endogenous factors play a prominent role in maintaining the balanced functioning of the immune system, which can result in immune suppression or immune stimulation. With the advent of different immune-modulatory agents, immune responses can be modulated or regulated to control infections and other health effects. Literature provides evidence on various immunomodulators from different sources and their role in modulating immune responses. Due to the limited efficacy of current drugs and the rise in drug resistance, there is a growing need for new therapies for infectious diseases. In this review, we aim to provide a comprehensive overview of different immune-modulating agents and immune therapies specifically focused on viral infectious diseases.
Collapse
Affiliation(s)
- P Sangeetha Vijayan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Joseph Xavier
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Mohanan Parayanthala Valappil
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India.
| |
Collapse
|
4
|
Sanchez PL, Andre G, Antipov A, Petrovsky N, Ross TM. Advax-SM™-Adjuvanted COBRA (H1/H3) Hemagglutinin Influenza Vaccines. Vaccines (Basel) 2024; 12:455. [PMID: 38793706 PMCID: PMC11125990 DOI: 10.3390/vaccines12050455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Adjuvants enhance immune responses stimulated by vaccines. To date, many seasonal influenza vaccines are not formulated with an adjuvant. In the present study, the adjuvant Advax-SM™ was combined with next generation, broadly reactive influenza hemagglutinin (HA) vaccines that were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. Advax-SM™ is a novel adjuvant comprising inulin polysaccharide and CpG55.2, a TLR9 agonist. COBRA HA vaccines were combined with Advax-SM™ or a comparator squalene emulsion (SE) adjuvant and administered to mice intramuscularly. Mice vaccinated with Advax-SM™ adjuvanted COBRA HA vaccines had increased serum levels of anti-influenza IgG and IgA, high hemagglutination inhibition activity against a panel of H1N1 and H3N2 influenza viruses, and increased anti-influenza antibody secreting cells isolated from spleens. COBRA HA plus Advax-SM™ immunized mice were protected against both morbidity and mortality following viral challenge and, at postmortem, had no detectable lung viral titers or lung inflammation. Overall, the Advax-SM™-adjuvanted COBRA HA formulation provided effective protection against drifted H1N1 and H3N2 influenza viruses.
Collapse
Affiliation(s)
- Pedro L. Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Greiciely Andre
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Anna Antipov
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
5
|
Lapi F, Domnich A, Marconi E, Rossi A, Grattagliano I, Cricelli C. Examining the effectiveness and duration of adjuvanted vs. non-adjuvanted influenza vaccines in protecting older adults against symptomatic SARS-CoV-2 infection. Br J Clin Pharmacol 2024; 90:600-605. [PMID: 37876110 DOI: 10.1111/bcp.15940] [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: 05/31/2023] [Revised: 08/24/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
Annual influenza vaccination is one of the main public health measures able to drastically reduce the burden of this infectious disease. Some evidence suggests 'trained immunity' triggered by influenza vaccine might reduce the risk of SARS-CoV-2 infection. Adjuvanted influenza vaccines are known to induce a broader cross-reactive immunity. No studies investigated the effect of adjuvanted vs. non-adjuvanted influenza vaccines on the risk of symptomatic SARS-CoV-2 infection. A case-control analysis nested in a cohort of subjects aged ≥65 years and immunized with adjuvanted or non-adjuvanted influenza vaccines was conducted. Although no statistically significant (OR = 0.87; P = .082) difference between the two vaccine types was observed for the 9-month follow-up period, a 17% (OR = 0.83; P = .042) reduction in the odds of COVID-19 was observed for adjuvanted vaccines with a 6-month follow-up. Further evidence is needed, but these results might have implications given the complexity of the upcoming winter seasons, in which the co-occurrence of influenza, SARS-CoV-2 and other respiratory infections (e.g., syncytial virus) might be unpredictable.
Collapse
Affiliation(s)
- Francesco Lapi
- Health Search, Italian College of General Practitioners and Primary Care, Florence, Italy
| | - Alexander Domnich
- Hygiene Unit, San Martino Policlinico Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Ettore Marconi
- Health Search, Italian College of General Practitioners and Primary Care, Florence, Italy
| | - Alessandro Rossi
- Italian College of General Practitioners and Primary Care, Florence, Italy
| | | | - Claudio Cricelli
- Italian College of General Practitioners and Primary Care, Florence, Italy
| |
Collapse
|
6
|
Sepasi A, Ghafourian M, Taghizadeh M, Mahdavi M. Formulation of Recombinant H1N1 Hemagglutinin in MF59 and Alum Adjuvants: A Comparison of the Vaccines Potency and Efficacy in BALB/C Mice. Viral Immunol 2023; 36:401-408. [PMID: 37504965 DOI: 10.1089/vim.2023.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
In this study, we reported the expression and potency of the recombinant H1N1 hemagglutinin (HA) vaccine as our in-house vaccine in a BALB/c mouse model. Recombinant H1N1 HA was produced in SF9 cell line, purified and formulated in MF59 adjuvant. Experimental mice were injected on days 0 and 14 with MF59-formulated vaccine, alum-based vaccine, and phosphate-buffered saline (PBS). Interleukin (IL)-2, IL-4, and interferon (IFN)-γ were assessed with commercial enzyme-linked immunosorbent assay (ELISA). Antibody responses and cytotoxic T lymphocyte (CTL) activity were assessed by hemagglutination inhibition and granzyme B ELISA, respectively. Moreover, the mice were challenged to show the vaccine efficacy. A considerable rise in IFN-γ and IL-4, as well as IFN-γ/IL-4 ratio, was observed in comparison with the alum-based vaccine and PBS group. Furthermore, our candidate vaccine showed superiority in humoral immune responses and CTL activity versus the alum-based vaccine and PBS group. The challenge showed that the survival rate in the vaccinated groups revealed a significant increase as compared with that in the PBS group. In conclusion, our candidate vaccine showed a robust Th1 response and CTL activity the alum-based vaccine. Moreover, a significant humoral immune response and a higher survival rate were detected in our vaccine as compared with the alum-based vaccine. It seems that the superiority of the MF59-based vaccine is due to the type of vaccine formulation in the candidate vaccine.
Collapse
Affiliation(s)
- Aref Sepasi
- Department of Immunology, School of Medicine; Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehri Ghafourian
- Department of Immunology, School of Medicine; Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Morteza Taghizadeh
- Department of Medical Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Castrodeza-Sanz J, Sanz-Muñoz I, Eiros JM. Adjuvants for COVID-19 Vaccines. Vaccines (Basel) 2023; 11:vaccines11050902. [PMID: 37243006 DOI: 10.3390/vaccines11050902] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, the improvement of traditional vaccines has meant that we have moved from inactivated whole virus vaccines, which provoke a moderate immune response but notable adverse effects, to much more processed vaccines such as protein subunit vaccines, which despite being less immunogenic have better tolerability profiles. This reduction in immunogenicity is detrimental to the prevention of people at risk. For this reason, adjuvants are a good solution to improve the immunogenicity of this type of vaccine, with much better tolerability profiles and a low prevalence of side effects. During the COVID-19 pandemic, vaccination focused on mRNA-type and viral vector vaccines. However, during the years 2022 and 2023, the first protein-based vaccines began to be approved. Adjuvanted vaccines are capable of inducing potent responses, not only humoral but also cellular, in populations whose immune systems are weak or do not respond properly, such as the elderly. Therefore, this type of vaccine should complete the portfolio of existing vaccines, and could help to complete vaccination against COVID-19 worldwide now and over the coming years. In this review we analyze the advantages and disadvantages of adjuvants, as well as their use in current and future vaccines against COVID-19.
Collapse
Affiliation(s)
- Javier Castrodeza-Sanz
- National Influenza Centre, 47005 Valladolid, Spain
- Preventive Medicine and Public Health Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
| | - Iván Sanz-Muñoz
- National Influenza Centre, 47005 Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, 42002 Soria, Spain
| | - Jose M Eiros
- National Influenza Centre, 47005 Valladolid, Spain
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain
- Microbiology Unit, Hospital Universitario Río Hortega, 47013 Valladolid, Spain
| |
Collapse
|
8
|
Ravichandran S, Erra-Diaz F, Karakaslar OE, Marches R, Kenyon-Pesce L, Rossi R, Chaussabel D, Pascual V, Palucka K, Paust S, Nahm MH, Kuchel GA, Banchereau J, Ucar D. Distinct baseline immune characteristics associated with responses to conjugated and unconjugated pneumococcal polysaccharide vaccines in older adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.16.23288531. [PMID: 37131707 PMCID: PMC10153339 DOI: 10.1101/2023.04.16.23288531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Pneumococcal infections cause serious illness and death among older adults. A capsular polysaccharide vaccine PPSV23 (Pneumovax®) and a conjugated polysaccharide vaccine PCV13 (Prevnar®) are used to prevent these infections, yet underlying responses, and baseline predictors remain unknown. We recruited and vaccinated 39 older adults (>60 years) with PPSV23 or PCV13. Both vaccines induced strong antibody responses at day 28 and similar plasmablast transcriptional signatures at day 10, however, their baseline predictors were distinct. Analyses of baseline flow cytometry and RNA-seq data (bulk and single cell) revealed a novel baseline phenotype that is specifically associated with weaker PCV13 responses, characterized by i) increased expression of cytotoxicity-associated genes and increased CD16+ NK frequency; ii) increased Th17 and decreased Th1 cell frequency. Men were more likely to display this cytotoxic phenotype and mounted weaker responses to PCV13 than women. Baseline expression levels of a distinct gene set was predictive of PPSV23 responses. This first precision vaccinology study for pneumococcal vaccine responses of older adults uncovered novel and distinct baseline predictors that might transform vaccination strategies and initiate novel interventions.
Collapse
Affiliation(s)
| | - Fernando Erra-Diaz
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- University of Buenos Aires, School of Medicine, Buenos Aires, Argentina #Current Address
| | - Onur E Karakaslar
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Leiden University Medical Center (LUMC), Leiden, Netherlands #Current Address
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Lisa Kenyon-Pesce
- UConn Center on Aging, University of Connecticut, Farmington, Connecticut, USA
| | - Robert Rossi
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Damien Chaussabel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Virginia Pascual
- Weill Cornell Medical College, Department of Pediatrics, NY, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Moon H Nahm
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut, Farmington, Connecticut, USA
| | - Jacques Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Immunai, New York, NY, USA, #Current Address
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut Health Center, Farmington, Connecticut, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
9
|
Ustyugova IV, Pougatcheva S, Farrell T, Strugnell T, Ganesh V, Zeldovich KB, Chivukula S, Goncalvez AP, Barro M. AF03 adjuvant improves anti-hemagglutinin and anti-neuraminidase immune responses induced by licensed seasonal quadrivalent influenza vaccines in mice. Vaccine 2023; 41:2022-2034. [PMID: 36803901 DOI: 10.1016/j.vaccine.2023.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/21/2023]
Abstract
Seasonal influenza remains a serious public health concern as the viral infection spreads easily from person to person and due to antigenic drift of neutralizing epitopes. Vaccination is the best method for disease prevention, however current seasonal influenza vaccines stimulate antibodies which are often effective against only antigenically similar strains. To boost the immune responses and increase vaccine effectiveness, adjuvants have been used for the past 20 years. The current study explores the use of oil-in-water adjuvant, AF03 to improve an immunogenicity of 2 licensed vaccines. A standard-dose inactivated quadrivalent influenza vaccine (IIV4-SD), containing both hemagglutinin (HA) and neuraminidase (NA) antigens, and recombinant quadrivalent influenza vaccine (RIV4), containing only HA-antigen were adjuvanted with AF03 in naïve BALB/c mouse model. Functional HA-specific antibody titers against all four homologous vaccine strains were enhanced by AF03, indicating potential increase in protective immunity. An increase in HA-specific total immunoglobulin G (IgG) binding titers were detected against homologous HAs, heterologous panel of 30 H3 HAs and seven Influenza B HAs. The neuraminidase inhibition (NAI) activity was significantly higher in IIV4-SD-AF03 group. Use of AF03 adjuvant improved the immune response to two influenza vaccines in a mouse model via an increase in functional and total antibodies against NA and a broad panel of HA-antigens.
Collapse
|
10
|
Surveillance of Severe Acute Respiratory Infection and Influenza Vaccine Effectiveness among Hospitalized Italian Adults, 2021/22 Season. Vaccines (Basel) 2022; 11:vaccines11010083. [PMID: 36679928 PMCID: PMC9861626 DOI: 10.3390/vaccines11010083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Following an extremely low incidence of influenza during the first waves of the ongoing COVID-19 pandemic, the 2021/22 Northern Hemisphere winter season saw a resurgence of influenza virus circulation. The aim of this study was to describe epidemiology of severe acute respiratory infections (SARIs) among Italian adults and estimate the 2021/22 season influenza vaccine effectiveness. For this purpose, a test-negative case-control study was conducted in a geographically representative sample of Italian hospitals. Of 753 SARI patients analyzed, 2.5% (N = 19) tested positive for influenza, most of which belonged to the A(H3N2) subtype. Phylogenetic analysis showed that these belonged to the subclade 3C.2a1b.2a.2, which was antigenically different from the 2021/22 A(H3N2) vaccine component. Most (89.5%) cases were registered among non-vaccinated individuals, suggesting a protective effect of influenza vaccination. Due to a limited number of cases, vaccine effectiveness estimated through the Firth's penalized logistic regression was highly imprecise, being 83.4% (95% CI: 25.8-97.4%) and 83.1% (95% CI: 22.2-97.3%) against any influenza type A and A(H3N2), respectively. Exclusion of SARS-CoV-2-positive controls from the model did not significantly change the base-case estimates. Within the study limitations, influenza vaccination appeared to be effective against laboratory-confirmed SARI.
Collapse
|
11
|
Prada LSD, Sanz-Muñoz I, de Lejarazu RO, Eiros JM, García-Sastre A, Aydillo T. Immunodominance hierarchy after seasonal influenza vaccination. Emerg Microbes Infect 2022; 11:2670-2679. [PMID: 36219456 DOI: 10.1080/22221751.2022.2135460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Current influenza vaccines elicit humoral immune responses against the hemagglutinin (HA) protein of influenza viruses. Different antigenic sites have been identified in the HA head as the main target of hemagglutination inhibition (HAI) antibodies (Sb, Sa, Cb, Ca1 and Ca2). To determine immunodominance (ID) of each site, we performed HAI assays against a panel of mutant viruses, each one lacking one of the classically defined antigenic sites and compared it to wild type (Wt). Agglutinating antibodies were measured before and after vaccination in two different regimens: Quadrivalent Influenza Vaccine (QIV) in young adults; or Adjuvanted Trivalent influenza Vaccine (ATIV) in elderly. Our results showed abs before vaccination were significantly reduced against all antigenic sites in the elderly and only against Sb and Ca2 in young adults compared to the Wt. Humoral response to vaccination was significantly reduced against all viruses compared to the Wt for the ATIV and only against Sb and Ca2 for the QIV. The strongest reduction was observed in all cases against Sb followed by Ca2. We concluded that ID profile was clearly dominated by Sb followed by Ca2. Additionally, the antibody response evolved with age, increasing the response towards less immunodominant epitopes of HA head. Adjuvants can positively influence ID hierarchy broadening responses towards multiple antigenic sites of HA head.
Collapse
Affiliation(s)
- Laura Sánchez-de Prada
- National Influenza Centre of Valladolid, 47010, Spain.,Hospital Clínico Universitario de Valladolid, 47003, Spain
| | | | | | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
12
|
Chang C, Music N, Cheung M, Rossignol E, Bedi S, Patel H, Safari M, Lee C, Otten GR, Settembre EC, Palladino G, Wen Y. Self-amplifying mRNA bicistronic influenza vaccines raise cross-reactive immune responses in mice and prevent infection in ferrets. Mol Ther Methods Clin Dev 2022; 27:195-205. [PMID: 36320414 PMCID: PMC9589142 DOI: 10.1016/j.omtm.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022]
Abstract
Vaccines are the primary intervention against influenza. Currently licensed inactivated vaccines focus immunity on viral hemagglutinin (HA). Self-amplifying mRNA (sa-mRNA) vaccines offer an opportunity to generate immunity to multiple viral proteins, including additional neuraminidase (NA). This evaluation of a bicistronic approach for sa-mRNA vaccine development compared subgenomic promoter and internal ribosome entry site strategies and found consistent and balanced expression of both HA and NA proteins in transfected cells. In mice, sa-mRNA bicistronic A/H5N1 vaccines raised potent anti-HA and anti-NA neutralizing antibody responses and HA- or NA-specific CD4+ and CD8+ T cell responses. The addition of NA also boosted the cross-neutralizing response to heterologous A/H1N1. Similar immunogenicity results were obtained for bicistronic seasonal A/H3N2 and B/Yamagata vaccines. In ferrets, sa-mRNA bicistronic A/H1N1 vaccine fully protected lung from infection by homologous virus and showed significant reduction of viral load in upper respiratory tract, warranting further evaluation of sa-mRNA bicistronic vaccine in humans.
Collapse
Affiliation(s)
- Cheng Chang
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Nedzad Music
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | - Harsh Patel
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | | | | | - Yingxia Wen
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA,Corresponding author Yingxia Wen, CSL, 50 Hampshire Street, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
A Comparative Analysis of Influenza-Associated Disease Burden with Different Influenza Vaccination Strategies for the Elderly Population in South Korea. Vaccines (Basel) 2022; 10:vaccines10091387. [PMID: 36146465 PMCID: PMC9503807 DOI: 10.3390/vaccines10091387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Influenza affects all age groups, but the risk of hospitalization and death due to influenza is strongly age-related and is at its highest among the elderly aged 65 years and older. The objective of this study is to compare the differences in influenza-associated disease burden under three different influenza vaccination strategies—the standard-dose quadrivalent influenza vaccine (QIV), high-dose QIV (HD-QIV), and MF59®-adjuvanted QIV (aQIV)—for the elderly population aged 65 years and older in South Korea. A one-year decision-tree model was developed to compare influenza disease burdens. The input data for the model were obtained from published literature reviews and surveillance data from the Korea Disease Control and Prevention Agency (KDCA). The analysis indicated that aQIV is more effective than QIV, preventing 35,390 influenza cases, 1602 influenza-associated complications, 709 influenza-associated hospitalizations, and 145 influenza-associated deaths annually. Additionally, aQIV, when compared to HD-QIV, also reduced the influenza-associated burden of disease, preventing 7247 influenza cases, 328 influenza-associated complications, 145 influenza-associated hospitalizations, and 30 influenza-associated deaths annually. Switching the vaccination strategy from QIV to aQIV is predicted to reduce the influenza-associated disease burden for the elderly in South Korea. The public health gains from aQIV and HD-QIV are expected to be comparable. Future studies comparing the effectiveness of the vaccines will further inform future vaccination strategies for the elderly in South Korea.
Collapse
|
14
|
Smart combination of aluminum hydroxide and MF59 to induce strong cellular immune responses. J Control Release 2022; 349:699-711. [PMID: 35907590 DOI: 10.1016/j.jconrel.2022.07.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 11/20/2022]
Abstract
As two of the most widely used adjuvants, aluminum hydroxide and the oil-in-water emulsion MF59 have their intrinsic limitations: classical aluminum gel induces only weak cellular immune responses while MF59 cannot be used as an antigen delivery system due to its poor physical interaction with antigen molecules. Herein, we combined these two adjuvants and constructed a novel nano-vaccine delivery system by inserting aluminum hydroxide into the surface of a modified MF59 nano-emulsion (AlNEs). A model antigen ovalbumin (OVA) and an immune potentiator CpG were adsorbed on the surface of AlNEs (hereinafter AlNEs-OVA-CpG) through a facile mixing step. After subcutaneous injection, AlNEs-OVA-CpG effectively drained to lymph nodes, delivered both cargos into lymph node-resident antigen presenting cells (APCs), and escaped from lysosomes into the cytoplasm, resulting in enhanced antigen cross-presentation. Finally, AlNEs-OVA-CpG induced potent antigen-specific humoral and cellular immune responses, which significantly inhibited tumor growth and prolonged mice survival in a EG7-OVA tumor model. In sum, our results suggested that AlNEs have a great prospect to induce CD8+ T cell responses for subunit antigens.
Collapse
|
15
|
Gärtner BC, Weinke T, Wahle K, Kwetkat A, Beier D, Schmidt KJ, Schwarz TF. Importance and value of adjuvanted influenza vaccine in the care of older adults from a European perspective - A systematic review of recently published literature on real-world data. Vaccine 2022; 40:2999-3008. [PMID: 35459556 DOI: 10.1016/j.vaccine.2022.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/21/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND There is an urgent need for improved influenza vaccines especially for older adults due to the presence of immunosenescence. It is therefore highly relevant to compare enhanced influenza vaccines with traditional influenza vaccines with respect to their effectiveness. OBJECTIVE To compare vaccine efficacy and effectiveness of adjuvanted influenza vaccines (aTIV/aQIV) vs. non-adjuvanted standard-dose (TIV/QIV) and high-dose (TIV-HD/QIV-HD) influenza vaccines regarding influenza-related outcomes in older adults, complementing findings from the European Centre for Disease Prevention and Control (ECDC)'s systematic review of enhanced seasonal influenza vaccines from February 2020. METHODS A systematic literature search was conducted in Embase and MEDLINE to identify randomised controlled trials, observational studies and systematic reviews, published since ECDC's systematic review (between 7 February 2020 and 6 September 2021). Included studies were appraised with either the Cochrane Risk of Bias tool, ROBINS-I or AMSTAR 2. RESULTS Eleven analyses from nine real-world evidence (RWE) studies comprising ∼53 million participants and assessing the relative vaccine effectiveness (rVE) of aTIV vs. TIV, QIV and/or TIV-HD in adults aged ≥65 years over the 2006/07-2008/09 and 2011/12-2019/20 influenza seasons were identified. Nine analyses found that aTIV was significantly more effective than TIV and QIV in reducing influenza-related outcomes by clinical setting and suspected influenza outbreaks (rVE ranging from 7.5% to 25.6% for aTIV vs. TIV and 7.1% to 36.3% for aTIV vs. QIV). Seven analyses found similar effectiveness of aTIV vs. TIV-HD in reducing influenza-related medical encounters, inpatient stays and hospitalisations/emergency room visits. In three analyses, aTIV was significantly more effective than TIV-HD in reducing influenza-related medical encounters and office visits (rVE ranging from 6.6% to 16.6%). Risk of bias of identified studies was moderate to high. CONCLUSIONS Our study suggests that both adjuvanted and high-dose vaccines are effective alternatives for vaccination programmes in older adults and preferable over conventional standard-dose vaccines.
Collapse
Affiliation(s)
- B C Gärtner
- Institute for Medical Microbiology and Hygiene, Saarland University Hospital and Medical Faculty of Saarland University, Kirrberger Straße, Building 43, 66421 Homburg/Saar, Germany
| | - T Weinke
- Clinic of Gastroenterology and Infectiology, Ernst von Bergmann Klinikum, Charlottenstraße 72, 14467 Potsdam, Germany
| | - K Wahle
- University of Münster, Schlossplatz 2, 48149 Münster, Germany
| | - A Kwetkat
- Department of Geriatrics, Jena University Hospital, Bachstraße 18, 07743 Jena, Germany
| | - D Beier
- Member of Saxon Committee on Vaccinations (SIKO), Elisabeth-Reichelt-Weg 35, 09116 Chemnitz, Germany
| | - K J Schmidt
- Xcenda GmbH, Lange Laube 31, 30159 Hannover, Germany
| | - T F Schwarz
- Institute of Laboratory Medicine and Vaccination Centre, Klinikum Würzburg Mitte, Standort Juliusspital, Salvatorstraße 7, 97074 Würzburg, Germany.
| |
Collapse
|
16
|
Li M, Wang H, Tian L, Pang Z, Yang Q, Huang T, Fan J, Song L, Tong Y, Fan H. COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther 2022; 7:146. [PMID: 35504917 PMCID: PMC9062866 DOI: 10.1038/s41392-022-00996-y] [Citation(s) in RCA: 176] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
With the constantly mutating of SARS-CoV-2 and the emergence of Variants of Concern (VOC), the implementation of vaccination is critically important. Existing SARS-CoV-2 vaccines mainly include inactivated, live attenuated, viral vector, protein subunit, RNA, DNA, and virus-like particle (VLP) vaccines. Viral vector vaccines, protein subunit vaccines, and mRNA vaccines may induce additional cellular or humoral immune regulations, including Th cell responses and germinal center responses, and form relevant memory cells, greatly improving their efficiency. However, some viral vector or mRNA vaccines may be associated with complications like thrombocytopenia and myocarditis, raising concerns about the safety of these COVID-19 vaccines. Here, we systemically assess the safety and efficacy of COVID-19 vaccines, including the possible complications and different effects on pregnant women, the elderly, people with immune diseases and acquired immunodeficiency syndrome (AIDS), transplant recipients, and cancer patients. Based on the current analysis, governments and relevant agencies are recommended to continue to advance the vaccine immunization process. Simultaneously, special attention should be paid to the health status of the vaccines, timely treatment of complications, vaccine development, and ensuring the lives and health of patients. In addition, available measures such as mix-and-match vaccination, developing new vaccines like nanoparticle vaccines, and optimizing immune adjuvant to improve vaccine safety and efficacy could be considered.
Collapse
Affiliation(s)
- Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Han Wang
- Laboratory for Clinical Immunology, Harbin Children's Hospital, Harbin, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Qingkun Yang
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Tianqi Huang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China. .,Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
17
|
Methodological advances in the design of peptide-based vaccines. Drug Discov Today 2022; 27:1367-1380. [DOI: 10.1016/j.drudis.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/02/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
|
18
|
A liposome-displayed hemagglutinin vaccine platform protects mice and ferrets from heterologous influenza virus challenge. Proc Natl Acad Sci U S A 2021; 118:2025759118. [PMID: 34050027 DOI: 10.1073/pnas.2025759118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recombinant influenza virus vaccines based on hemagglutinin (HA) hold the potential to accelerate production timelines and improve efficacy relative to traditional egg-based platforms. Here, we assess a vaccine adjuvant system comprised of immunogenic liposomes that spontaneously convert soluble antigens into a particle format, displayed on the bilayer surface. When trimeric H3 HA was presented on liposomes, antigen delivery to macrophages was improved in vitro, and strong functional antibody responses were induced following intramuscular immunization of mice. Protection was conferred against challenge with a heterologous strain of H3N2 virus, and naive mice were also protected following passive serum transfer. When admixed with the particle-forming liposomes, immunization reduced viral infection severity at vaccine doses as low as 2 ng HA, highlighting dose-sparing potential. In ferrets, immunization induced neutralizing antibodies that reduced the upper respiratory viral load upon challenge with a more modern, heterologous H3N2 viral strain. To demonstrate the flexibility and modular nature of the liposome system, 10 recombinant surface antigens representing distinct influenza virus strains were bound simultaneously to generate a highly multivalent protein particle that with 5 ng individual antigen dosing induced antibodies in mice that specifically recognized the constituent immunogens and conferred protection against heterologous H5N1 influenza virus challenge. Taken together, these results show that stable presentation of recombinant HA on immunogenic liposome surfaces in an arrayed fashion enhances functional immune responses and warrants further attention for the development of broadly protective influenza virus vaccines.
Collapse
|
19
|
Behzadi M, Vakili B, Ebrahiminezhad A, Nezafat N. Iron nanoparticles as novel vaccine adjuvants. Eur J Pharm Sci 2021; 159:105718. [PMID: 33465476 DOI: 10.1016/j.ejps.2021.105718] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
The poor immunogenicity of peptide vaccines compared to conventional ones re usually improved by applying different adjuvants. As chemical or biological substances, adjuvants are added to vaccines to enhance and prolong the immune response. According to considerable investigations over the recent years in the context of finding new adjuvants, a handful of vaccine adjuvants have been licensed for human use. Recently, engineered nanoparticles (NPs) have been introduced as novel alternatives to traditional vaccine adjuvant. Metallic nanoparticles (MeNPs) are among the most promising NPs used for vaccine adjuvant as well as the delivery system that can improve immune responses against pathogens. Iron NPs, as an important class of MeNPs, have gained increasing attention as novel vaccine adjuvants. These particles have shown acceptable results in preclinical studies. Hence, understanding the physicochemical properties of iron NPs, including size, surface properties, charge and route of administration, is of substantial importance. The aim of this review is to provide an overview of the immunomodulatory effects of iron NPs as novel adjuvants. Furthermore, physicochemical properties of these NPs were also discussed.
Collapse
Affiliation(s)
- Maryam Behzadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Vakili
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Ebrahiminezhad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
20
|
Nagashima KA, Mousa JJ. Next-Generation Influenza HA Immunogens and Adjuvants in Pursuit of a Broadly Protective Vaccine. Viruses 2021; 13:v13040546. [PMID: 33805245 PMCID: PMC8064354 DOI: 10.3390/v13040546] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/20/2022] Open
Abstract
Influenza virus, a highly mutable respiratory pathogen, causes significant disease nearly every year. Current vaccines are designed to protect against circulating influenza strains of a given season. However, mismatches between vaccine strains and circulating strains, as well as inferior vaccine effectiveness in immunodeficient populations, represent major obstacles. In an effort to expand the breadth of protection elicited by influenza vaccination, one of the major surface glycoproteins, hemagglutinin (HA), has been modified to develop immunogens that display conserved regions from multiple viruses or elicit a highly polyclonal antibody response to broaden protection. These approaches, which target either the head or the stalk domain of HA, or both domains, have shown promise in recent preclinical and clinical studies. Furthermore, the role of adjuvants in bolstering the robustness of the humoral response has been studied, and their effects on the vaccine-elicited antibody repertoire are currently being investigated. This review will discuss the progress made in the universal influenza vaccine field with respect to influenza A viruses from the perspectives of both antigen and adjuvant, with a focus on the elicitation of broadly neutralizing antibodies.
Collapse
Affiliation(s)
- Kaito A. Nagashima
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Correspondence:
| |
Collapse
|
21
|
Pollet J, Chen WH, Strych U. Recombinant protein vaccines, a proven approach against coronavirus pandemics. Adv Drug Deliv Rev 2021; 170:71-82. [PMID: 33421475 PMCID: PMC7788321 DOI: 10.1016/j.addr.2021.01.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/15/2020] [Accepted: 01/01/2021] [Indexed: 02/06/2023]
Abstract
With the COVID-19 pandemic now ongoing for close to a year, people all over the world are still waiting for a vaccine to become available. The initial focus of accelerated global research and development efforts to bring a vaccine to market as soon as possible was on novel platform technologies that promised speed but had limited history in the clinic. In contrast, recombinant protein vaccines, with numerous examples in the clinic for many years, missed out on the early wave of investments from government and industry. Emerging data are now surfacing suggesting that recombinant protein vaccines indeed might offer an advantage or complement to the nucleic acid or viral vector vaccines that will likely reach the clinic faster. Here, we summarize the current public information on the nature and on the development status of recombinant subunit antigens and adjuvants targeting SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Jeroen Pollet
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States of America; Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, 1102 Bates Street, Houston, TX, United States of America.
| | - Wen-Hsiang Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States of America; Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, 1102 Bates Street, Houston, TX, United States of America
| | - Ulrich Strych
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States of America; Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, 1102 Bates Street, Houston, TX, United States of America
| |
Collapse
|
22
|
The impact of immuno-aging on SARS-CoV-2 vaccine development. GeroScience 2021; 43:31-51. [PMID: 33569701 PMCID: PMC7875765 DOI: 10.1007/s11357-021-00323-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
The SARS-CoV-2 pandemic has almost 56 million confirmed cases resulting in over 1.3 million deaths as of November 2020. This infection has proved more deadly to older adults (those >65 years of age) and those with immunocompromising conditions. The worldwide population aged 65 years and older is increasing, and the total number of aged individuals will outnumber those younger than 65 years by the year 2050. Aging is associated with a decline in immune function and chronic activation of inflammation that contributes to enhanced viral susceptibility and reduced responses to vaccination. Here we briefly review the pathogenicity of the virus, epidemiology and clinical response, and the underlying mechanisms of human aging in improving vaccination. We review current methods to improve vaccination in the older adults using novel vaccine platforms and adjuvant systems. We conclude by summarizing the existing clinical trials for a SARS-CoV-2 vaccine and discussing how to address the unique challenges for vaccine development presented with an aging immune system.
Collapse
|
23
|
Tokuhara D, Hikita N. Cord Blood-Based Approach to Assess Candidate Vaccine Adjuvants Designed for Neonates and Infants. Vaccines (Basel) 2021; 9:vaccines9020095. [PMID: 33514054 PMCID: PMC7911524 DOI: 10.3390/vaccines9020095] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Neonates and infants are particularly susceptible to infections, for which outcomes tend to be severe. Vaccination is a key strategy for preventing infectious diseases, but the protective immunity achieved through vaccination typically is weaker in infants than in healthy adults. One possible explanation for the poor acquisition of vaccine-induced immunity in infants is that their innate immune response, represented by toll-like receptors, is immature. The current system for developing pediatric vaccines relies on the confirmation of their safety and effectiveness in studies involving the use of mature animals or adult humans. However, creating vaccines for neonates and infants requires an understanding of their uniquely immature innate immunity. Here we review current knowledge regarding the innate immune system of neonates and infants and challenges in developing vaccine adjuvants for those children through analyses of cord blood.
Collapse
|
24
|
Exploring the Potential of T-Cells for a Universal Influenza Vaccine. Vaccines (Basel) 2020; 8:vaccines8040598. [PMID: 33050614 PMCID: PMC7711579 DOI: 10.3390/vaccines8040598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 01/18/2023] Open
|
25
|
Boudreau CM, Yu WH, Suscovich TJ, Talbot HK, Edwards KM, Alter G. Selective induction of antibody effector functional responses using MF59-adjuvanted vaccination. J Clin Invest 2020; 130:662-672. [PMID: 31845904 DOI: 10.1172/jci129520] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/15/2019] [Indexed: 12/14/2022] Open
Abstract
Seasonal and pandemic influenza infection remains a major public health concern worldwide. Driving robust humoral immunity has been a challenge given preexisting, often cross-reactive, immunity and in particular, poorly immunogenic avian antigens. To overcome immune barriers, the adjuvant MF59 has been used in seasonal influenza vaccines to increase antibody titers and improve neutralizing activity, translating to a moderate increase in protection in vulnerable populations. However, its effects on stimulating antibody effector functions, including NK cell activation, monocyte phagocytosis, and complement activity, all of which have been implicated in protection against influenza, have yet to be defined. Using systems serology, we assessed changes in antibody functional profiles in individuals who received H5N1 avian influenza vaccine administered with MF59, with alum, or delivered unadjuvanted. MF59 elicited antibody responses that stimulated robust neutrophil phagocytosis and complement activity. Conversely, vaccination with MF59 recruited NK cells poorly and drove moderate monocyte phagocytic activity, both likely compromised because of the induction of antibodies that did not bind FCGR3A. Collectively, defining the humoral antibody functions induced by distinct adjuvants may provide a path to designing next-generation vaccines that can selectively leverage the humoral immune functions, beyond binding and neutralization, resulting in better protection from infection.
Collapse
Affiliation(s)
- Carolyn M Boudreau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA.,PhD program in Virology, Division of Medical Sciences, Harvard University, Boston, Massachusetts, USA
| | - Wen-Han Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Todd J Suscovich
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - H Keipp Talbot
- Department of Medicine.,Department of Health Policy, and
| | - Kathryn M Edwards
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
26
|
Gupta T, Gupta SK. Potential adjuvants for the development of a SARS-CoV-2 vaccine based on experimental results from similar coronaviruses. Int Immunopharmacol 2020; 86:106717. [PMID: 32585611 PMCID: PMC7301105 DOI: 10.1016/j.intimp.2020.106717] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
The extensive efforts around the globe are being made to develop a suitable vaccine against COVID-19 (Coronavirus Disease-19) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus-2). An effective vaccine should be able to induce high titers of neutralizing antibodies to prevent the virus from attaching to the host cell receptors. However, to elicit the protective levels of antibodies, a vaccine may require multiple doses or assistance from other immunostimulatory molecules. Further, the vaccine should be able to induce protective levels of antibodies rapidly with the least amount of antigen used. This decreases the cost of a vaccine and makes it affordable. As the pandemic has hit most countries across the globe, there will be an overwhelming demand for the vaccine in a quick time. Incorporating a suitable adjuvant in a SARS-CoV-2 vaccine may address these requirements. This review paper will discuss the experimental results of the adjuvanted vaccine studies with similar coronaviruses (CoVs) which might be useful to select an appropriate adjuvant for a vaccine against rapidly emergingSARS-CoV-2. We also discuss the current progress in the development of adjuvanted vaccines against the disease.
Collapse
Affiliation(s)
- Tania Gupta
- Dr GC Negi College of Veterinary and Animal Sciences, Palampur 176062, Himachal Pradesh, India.
| | - Shishir K Gupta
- CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|
27
|
Zou Y, Wu N, Miao C, Yue H, Wu J, Ma G. A novel multiple emulsion enhanced immunity via its biomimetic delivery approach. J Mater Chem B 2020; 8:7365-7374. [DOI: 10.1039/d0tb01318h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A special emulsion with biomimetic structural dynamic properties was fabricated, inducing efficient vaccine–cell interaction and robust immunity.
Collapse
Affiliation(s)
- Yongjuan Zou
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Nan Wu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| |
Collapse
|
28
|
Panatto D, Haag M, Lai PL, Tomczyk S, Amicizia D, Lino MM. Enhanced Passive Safety Surveillance (EPSS) confirms an optimal safety profile of the use of MF59 ® -adjuvanted influenza vaccine in older adults: Results from three consecutive seasons. Influenza Other Respir Viruses 2020; 14:61-66. [PMID: 31617965 PMCID: PMC6928029 DOI: 10.1111/irv.12685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/06/2019] [Accepted: 09/18/2019] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND In Europe, the enhanced safety surveillance (ESS) of seasonal influenza vaccines is mandatory, in order to detect any potential increase in reactogenicity when the vaccine composition is updated. The MF59® -adjuvanted influenza vaccine (Fluad™) is the first and the only licensed adjuvanted seasonal influenza vaccine in Europe. OBJECTIVE Our objective was to summarize the safety data of Fluad™ over three consecutive seasons. METHODS A passive approach to ESS (EPSS) was adopted, in which reporting of spontaneous adverse events (AEs) by vaccinees and vaccine exposure was estimated, in order to generate a near real-time reporting rate. EPSS was conducted in Italy during the 2015, 2016, and 2017 influenza seasons in the primary care setting. All AEs reported within 7 days following immunization were analyzed by season, type and seriousness. Fisher's exact test was used to compare frequencies between seasons. RESULTS Total exposure accounted for approximately 1,000 doses of Fluad™ for each season. A total of 0.5% (2015), 0.7% (2016), and 0.5% (2017) individual case safety reports (ICSRs) were received, corresponding to a total of 9 (2015), 18 (2016), and 12 (2017) spontaneous AEs. The frequencies of AEs of interest were below those expected on the basis of the known safety profile of the vaccine. Most AEs were mild-to-moderate in severity. No between-season difference was found. CONCLUSIONS Our analyses confirmed that the safety data observed were consistent with the known safety profile of Fluad™, which has been amply established over the last 20 years. No significant changes in the safety profile were observed.
Collapse
Affiliation(s)
- Donatella Panatto
- Department of Health SciencesUniversity of GenoaGenoaItaly
- Interuniversity Research Center on Influenza and other Transmissible Infections (CIRI‐IT)GenoaItaly
| | - Mendel Haag
- Clinical DevelopmentSeqirus Netherlands B.V.Amsterdamthe Netherlands
| | - Piero Luigi Lai
- Department of Health SciencesUniversity of GenoaGenoaItaly
- Interuniversity Research Center on Influenza and other Transmissible Infections (CIRI‐IT)GenoaItaly
| | | | - Daniela Amicizia
- Department of Health SciencesUniversity of GenoaGenoaItaly
- Interuniversity Research Center on Influenza and other Transmissible Infections (CIRI‐IT)GenoaItaly
| | | |
Collapse
|
29
|
Priming with MF59 adjuvanted versus nonadjuvanted seasonal influenza vaccines in children - A systematic review and a meta-analysis. Vaccine 2019; 38:608-619. [PMID: 31735505 DOI: 10.1016/j.vaccine.2019.10.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/11/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Identifying optimal priming strategies for children <2 years could substantially improve the public health benefits of influenza vaccines. Adjuvanted seasonal influenza vaccines were designed to promote a better immune response among young vaccine-naïve children. METHODS We systematically reviewed randomized trials to assess hemagglutination inhibition (HAI) antibody response to MF59-adjuvanted inactivated influenza vaccine (aIIV) versus nonadjuvanted IIV among children. We estimated pooled ratios of post-vaccination HAI geometric mean titer (GMT) for aIIV versus IIV and confidence intervals (CIs) using the pooled variances derived from reported CIs. RESULTS Mean age was 28 months (range, 6-72 months). Children received vaccines with either 7.5 μg (6-35 months) or 15 μg (≥36 months) hemagglutinin of each strain depending on age. Seven of eight trials administered trivalent vaccines and one used quadrivalent vaccine. Pooled post-vaccination GMT ratios against the three influenza vaccine strains were 2.5-3.5 fold higher after 2-dose-aIIV versus 2-dose-IIV among children 6-72 months, and point estimates were higher among children 6-35 months compared with older children. When comparing 1-dose-aIIV to 2-dose-IIV doses, pooled GMT ratios were not significantly different against A/H1N1 (1.0; 95% CI: 0.5-1.8; p = 0.90) and A/H3N2 viruses (1.0; 95% CI: 0.7-1.5; p = 0.81) and were significantly lower against B viruses (0.6; 95% CI: 0.4-0.8; p < 0.001) for both age groups. Notably, GMT ratios for vaccine-mismatched heterologous viruses after 2-dose-aIIV compared with 2-dose-IIV were higher against A/H1N1 (2.0; 95% CI: 1.1-3.4), A/H3N2 (2.9; 95% CI: 1.9-4.2), and B-lineage viruses (2.1; 95% CI: 1.8-2.6). CONCLUSIONS Two doses of adjuvanted IIV consistently induced better humoral immune responses against Type A and B influenza viruses compared with nonadjuvanted IIVs in young children, particularly among those 6-35 months. One adjuvanted IIV dose had a similar response to two nonadjuvanted IIV doses against Type A influenza viruses. Longer-term benefits from imprinting and cell-mediated immunity, including trials of clinical efficacy, are gaps that warrant investigation.
Collapse
|
30
|
Evaluation of aqueous extracts of Cistanche deserticola as a polysaccharide adjuvant for seasonal influenza vaccine in young adult mice. Immunol Lett 2019; 213:1-8. [DOI: 10.1016/j.imlet.2019.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/01/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022]
|
31
|
Tegenge MA, Von Tungeln LS, Anderson SA, Mitkus RJ, Vanlandingham MM, Forshee RA, Beland FA. Comparative pharmacokinetic and biodistribution study of two distinct squalene-containing oil-in-water emulsion adjuvants in H5N1 influenza vaccines. Regul Toxicol Pharmacol 2019; 108:104436. [PMID: 31381939 DOI: 10.1016/j.yrtph.2019.104436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/15/2019] [Accepted: 08/01/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND In recent years, there has been great interest from academia, industry and government scientists for an increased understanding of the mode of action of vaccine adjuvants to characterize the safety and efficacy of vaccines. In this context, pharmacokinetic (PK) and biodistribution studies are useful for quantifying the concentration of vaccine adjuvants in mechanistically or toxicologically relevant target tissues. METHODS In this study, we conducted a comparative analysis of the PK and biodistribution profile of radiolabeled squalene for up to 336 h (14 days) after intramuscular injection of mice with adjuvanted H5N1 influenza vaccines. The evaluated adjuvants included an experimental-grade squalene-in-water (SQ/W) emulsion (AddaVax®) and an adjuvant system (AS03®) that contained squalene and α-tocopherol in the oil phase of the emulsion. RESULTS The half-life of the initial exponential decay from quadriceps muscle was 1.5 h for AS03 versus 12.9 h for AddaVax. At early time points (1-6 h), there was about a 10-fold higher concentration of labeled squalene in draining lymph nodes following AS03 injection compared to AddaVax. The area-under-concentration curve up to 336 h (AUC0-336hr) and peak concentration of squalene in spleen (immune organ) was about 1.7-fold higher following injection of AS03 than AddaVax. The peak systemic tissue concentration of squalene from the two adjuvants, with or without antigen, remained below 1% of injected dose for toxicologically relevant target tissues, such as spinal cord, brain, and kidney. The pharmacokinetics of AS03 was unaffected by the presence of H5N1 antigen. CONCLUSIONS This study demonstrates a rapid decline of AS03 from the quadriceps muscles of mice as compared to conventional SQ/W emulsion adjuvant, with an increased transfer to mechanistically relevant tissues such as local lymph nodes. Systemic tissue exposure to potential toxicological target tissues was very low.
Collapse
Affiliation(s)
- Million A Tegenge
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, 20993, USA.
| | - Linda S Von Tungeln
- Division of Biochemical Toxicology, National Center for Toxicological Research, FDA, Jefferson, AR, 72079, USA
| | - Steven A Anderson
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, 20993, USA
| | - Robert J Mitkus
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, 20993, USA; Current Address: BASF Corporation, 26 Davis Drive, Durham, NC, 27709, USA
| | - Michelle M Vanlandingham
- Division of Biochemical Toxicology, National Center for Toxicological Research, FDA, Jefferson, AR, 72079, USA
| | - Richard A Forshee
- Office of Biostatistics & Epidemiology, Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, 20993, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, FDA, Jefferson, AR, 72079, USA
| |
Collapse
|
32
|
Trimaille T, Lacroix C, Verrier B. Self-assembled amphiphilic copolymers as dual delivery system for immunotherapy. Eur J Pharm Biopharm 2019; 142:232-239. [PMID: 31229673 DOI: 10.1016/j.ejpb.2019.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/03/2019] [Accepted: 06/19/2019] [Indexed: 01/07/2023]
Abstract
Subunit vaccines using recombinant antigens appear as the privileged vaccination technology for safety reasons but still require the development of carriers/adjuvants ensuring optimal immunogenicity and efficacy. Micelles from self-assembled amphiphilic copolymers have recently emerged as highly relevant and promising candidates owing to their ease of preparation, low size (entering in lymphatic capillaries for reaching lymph nodes), size/surface tunability and chemical versatility enabling introduction of stimuli (e.g. pH) responsive features and biofunctionalization with dedicated molecules. In particular, research efforts have increasingly focused on dendritic cells (DCs) targeting and activation by co-delivering (with antigen) ligands of pattern recognition receptors (PRRs, e.g. toll-like receptors). Such strategy has appeared as one of the most effective for eliciting CD 8+ T-cell response, which is crucial in the eradication of tumors and numerous infectious diseases. In this short review, we highlight the recent advances in such micelle-based carriers in subunit vaccination and how their precise engineering can be a strong asset for guiding and controlling immune responses.
Collapse
Affiliation(s)
- Thomas Trimaille
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire, Marseille, France.
| | - Céline Lacroix
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| | - Bernard Verrier
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| |
Collapse
|
33
|
Loperto I, Simonetti A, Nardone A, Triassi M. Use of adjuvanted trivalent influenza vaccine in older-age adults: a systematic review of economic evidence. Hum Vaccin Immunother 2019; 15:1035-1047. [PMID: 30735465 DOI: 10.1080/21645515.2019.1578597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Seasonal influenza is a very common disease. Yearly vaccination of at-risk population groups is a well-recognized cost-effective/cost-saving preventive measure. It is, however, unclear which available alternative has the most favorable economic profile. Some available options are: trivalent (TIV) and quadrivalent (QIV) inactivated vaccines, adjuvanted TIV (aTIV). Because of immunosenescence, aTIV has been specifically developed for elderly. The present study aimed at assessing the available evidence of aTIV use in elderly from the economic perspective. A systematic literature review targeting aTIV economic evaluations in adults aged ≥65 years was performed using Medline via Ovid, Embase, DARE and NHS/EED. Of a total of 3,654 papers screened, 18 studies (13 full papers, 5 conference abstracts) were included. It emerged that compared with both non-vaccination or non-adjuvanted vaccines, aTIV was cost-effective or cost-saving. The vaccinations strategies incorporating aTIV based on age and/or risk profile are associated with the most favorable economic outcomes.
Collapse
Affiliation(s)
- Ilaria Loperto
- a Department of Public Health , University of Naples "Federico II" , Naples , Italy
| | - Andrea Simonetti
- a Department of Public Health , University of Naples "Federico II" , Naples , Italy
| | - Antonio Nardone
- a Department of Public Health , University of Naples "Federico II" , Naples , Italy
| | - Maria Triassi
- a Department of Public Health , University of Naples "Federico II" , Naples , Italy
| |
Collapse
|
34
|
Boudreau CM, Alter G. Extra-Neutralizing FcR-Mediated Antibody Functions for a Universal Influenza Vaccine. Front Immunol 2019; 10:440. [PMID: 30949165 PMCID: PMC6436086 DOI: 10.3389/fimmu.2019.00440] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/19/2019] [Indexed: 12/22/2022] Open
Abstract
While neutralizing antibody titers measured by hemagglutination inhibition have been proposed as a correlate of protection following influenza vaccination, neutralization alone is a modest predictor of protection against seasonal influenza. Instead, emerging data point to a critical role for additional extra-neutralizing functions of antibodies in protection from infection. Specifically, beyond binding and neutralization, antibodies mediate a variety of additional immune functions via their ability to recruit and deploy innate immune effector function. Along these lines, antibody-dependent cellular cytotoxicity, antibody-mediated macrophage phagocytosis and activation, antibody-driven neutrophil activation, antibody-dependent complement deposition, and non-classical Fc-receptor antibody trafficking have all been implicated in protection from influenza infection. However, the precise mechanism(s) by which the immune system actively tunes antibody functionality to drive protective immunity has been poorly characterized. Here we review the data related to Fc-effector functional protection from influenza and discuss prospects to leverage this humoral immune activity for the development of a universal influenza vaccine.
Collapse
Affiliation(s)
- Carolyn M Boudreau
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States.,Harvard Ph.D. Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| |
Collapse
|
35
|
Hoare R, Jung SJ, Ngo TPH, Bartie K, Bailey J, Thompson KD, Adams A. Efficacy and safety of a non-mineral oil adjuvanted injectable vaccine for the protection of Atlantic salmon (Salmo salar L.) against Flavobacterium psychrophilum. FISH & SHELLFISH IMMUNOLOGY 2019; 85:44-51. [PMID: 29017943 DOI: 10.1016/j.fsi.2017.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 06/07/2023]
Abstract
Flavobacterium psychrophilum is the causative agent of Rainbow Trout Fry Syndrome which has had a major impact on global salmonid aquaculture. Recent outbreaks in Atlantic salmon in Scotland and Chile have added to the need for a vaccine to protect both salmon and trout. At present no licensed vaccines are available in Europe, leaving antibiotics as the only course of action to contain disease outbreaks. Outbreaks generally occur in fry at temperatures between 10 and 15 °C. Recently outbreaks in larger fish have given added impetus to the development of a vaccine which can provide long term protection from this highly heterogeneous pathogen. Most fish injectable vaccines are formulated with oil emulsion adjuvants to induce strong and long lasting immunity, but which are known to cause side effects. Alternative adjuvants are currently sought to minimise these adverse effects. The current study was performed to assess the efficacy of a polyvalent, whole cell vaccine containing formalin-inactivated F. psychrophilum to induce protective immunity in Atlantic salmon. The vaccine was formulated with an adjuvant containing squalene and aluminium hydroxide, and was compared to a vaccine formulated with a traditional oil adjuvant, Montanide ISA 760VG, and a non-adjuvanted vaccine. Duplicate groups of salmon (23.5 ± 6.8 g) were vaccinated with each of the vaccine formulations or phosphate buffered saline by intraperitoneal injection. Fish were challenged by intramuscular injection with F. psychrophilum six weeks post-vaccination to test the efficacy of the vaccines. Cumulative mortality reached 70% in the control salmon, while the groups of salmon that received vaccine had significantly lower mortality than the controls (p = 0.0001), with no significant difference in survival between vaccinated groups. The squalene/alum adjuvant was safe, more readily metabolised by the fish and induced less histopathological changes than the traditional oil adjuvant.
Collapse
Affiliation(s)
- R Hoare
- Institute of Aquaculture, University of Stirling, Stirling, UK.
| | - S-J Jung
- Chonnam National University, Yeosu, Republic of Korea
| | - T P H Ngo
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - K Bartie
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - J Bailey
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - K D Thompson
- Moredun Research Institute, Pentland Science Park, Penicuik, UK
| | - A Adams
- Institute of Aquaculture, University of Stirling, Stirling, UK
| |
Collapse
|
36
|
Liu Z, Ru J, Sun S, Teng Z, Dong H, Song P, Yang Y, Guo H. Uniform dendrimer-like mesoporous silica nanoparticles as a nano-adjuvant for foot-and-mouth disease virus-like particle vaccine. J Mater Chem B 2019. [DOI: 10.1039/c8tb03315c] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dendrimer-like mesoporous silica nanoparticles (MSNs) with large center-radial mesopores have been prepared for macromolecular protein loading and delivery.
Collapse
Affiliation(s)
- Zhijun Liu
- School of Petrochemical Engineering
- Lanzhou University of Technology
- Lanzhou 730050
- P. R. China
- State Key Laboratory of Veterinary Etiological Biology
| | - Jiaxi Ru
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| | - Zhidong Teng
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| | - Hu Dong
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| | - Pin Song
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| | - Yunshang Yang
- School of Petrochemical Engineering
- Lanzhou University of Technology
- Lanzhou 730050
- P. R. China
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology
- Lanzhou Veterinary Research Institute
- Chinese Academy of Agricultural Sciences
- Lanzhou
- P. R. China
| |
Collapse
|
37
|
Schaffner W, van Buynder P, McNeil S, Osterhaus ADME. Seasonal influenza immunisation: Strategies for older adults. Int J Clin Pract 2018; 72:e13249. [PMID: 30216647 DOI: 10.1111/ijcp.13249] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
Adults over the age of 60-65 years suffer disproportionally from seasonal influenza, experiencing high rates of complications, exacerbation of underlying medical comorbidities, and excess mortality. Thus, older adults are an important priority for influenza immunisation campaigns. Unfortunately, older adults generally display lower immune responses to standard influenza vaccines because of immunosenescence, with resulting suboptimal vaccine effectiveness. Thus, the development of improved vaccines that heighten immune responses and improve effectiveness is an important medical need. To this end, enhanced influenza vaccines specifically targeting this age group have been developed, which seek to overcome the inherent limitations in the immune responses of older adults. Both the licensed high-dose trivalent influenza vaccine (hdTIV) containing fourfold higher antigen contents than standard vaccine, and the MF59® -adjuvanted trivalent influenza vaccine (aTIV) have been proven to be safe and well-tolerated while enhancing the immune response. Healthcare providers for populations of older adults should be advised to routinely use these enhanced influenza vaccines in seasonal immunisation campaigns to provide improved immunity against influenza and its consequences in this particularly susceptible age group.
Collapse
Affiliation(s)
| | - Paul van Buynder
- School of Medicine, Griffith University, Gold Coast, Qld, Australia
| | - Shelly McNeil
- Canadian Center for Vaccinology, IWK Health Center and Nova Scotia Health Authority, Dalhousie University, Halifax, NS, Canada
| | - Albert D M E Osterhaus
- Research Centre for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hanover, LS, Germany
| |
Collapse
|
38
|
Song S, Liu Z, Zhou J, Cai W, Yang H, Ma L, Gao J, Li W, Liao G. An adjuvant compound that enhances immunogenicity at fractional doses of the Sabin-inactivated poliovirus vaccine (sIPV) with a long duration of protection in a rat model. J Med Virol 2018; 91:14-21. [PMID: 30168587 DOI: 10.1002/jmv.25298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND At the same dosage, the new generation of Sabin-inactivated poliovirus vaccine (sIPV) is less immunogenic than the traditional oral polio vaccine (OPV) dosage in China. The useful adjuvant might be a necessary strategy to strengthen the immune protective effects. METHODS In this study, we produced an adjuvant compound (named KML05) that could promote immunogenicity and fractional doses of sIPV with a long duration of protection in a rat model. The compound adjuvant had both advantages and a function of MF59 and carbopol971P. RESULTS The effect seroconversion of experimental animals immunized with KML05 could be extended to one-eighth of the dose. According to the result of the geometric mean titers (GMTs), KML05 adjuvant could save eight times the amount of sIPV D-antigen usage, but aluminum hydroxide adjuvant could save twice at the same titers. Additionally, it was found that there was a significant difference in the GMT titer of poliovirus type 2 between animals immunized by KML05 and alum adjuvant (P < 0.05). At 12th-month postvaccination, the neutralization titers stimulated by IPV-KML05 were maintained over a longer time period in immunized animals. CONCLUSION Our research team developed KML05 adjuvant, which combined carbopol971P with MF59, increased antibody responses to sIPV for a longer duration of protection in a rat model.
Collapse
Affiliation(s)
- Shaohui Song
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Ze Liu
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Jian Zhou
- The Outreach Department of Certification with World Health Organization, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Wei Cai
- The Fourth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Huijuan Yang
- The Sixth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Lei Ma
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Jingxia Gao
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Weidong Li
- The Regulatory Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Guoyang Liao
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| |
Collapse
|
39
|
Abstract
Adjuvants are included in vaccine formulations to enhance the immunogenicity and efficacy of vaccines. MF59® is an oil-in-water emulsion adjuvant and licensed for use in pandemic and seasonal influenza vaccines in many countries. MF59 is safe and well tolerated in humans. MF59-adjuvanted vaccination spares vaccine dose and enhances hemagglutination inhibiting antibodies against homologous and heterologous influenza virus strains. The mechanisms of MF59 involve rapid induction of chemokines, inflammatory cytokines, recruiting multiple immune cells, uric acid and benign apoptosis of certain innate immune cells. The adjuvant effects of MF59 on generating vaccine-specific isotype-switched IgG antibodies, effector CD8 T cells, and protective immunity were retained even in a CD4-deficient condition by inducing effective immune-competent microenvironment with various innate and antigen presenting cells in a mouse model. CD4-independent adjuvant effects of MF59 might contribute to improving the vaccine efficacy in children, the elderly, and immunocompromised patients as well as in healthy adults. Further studies will be needed to broaden the use of MF59 in various vaccine antigens and populations as well as lead to better understanding of the action mechanisms of MF59 adjuvant.
Collapse
Affiliation(s)
- Eun-Ju Ko
- a Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA.,b Vaccine Branch, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , MD , USA
| | - Sang-Moo Kang
- a Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA
| |
Collapse
|
40
|
Schaffner W, Chen WH, Hopkins RH, Neuzil K. Effective Immunization of Older Adults Against Seasonal Influenza. Am J Med 2018; 131:865-873. [PMID: 29544989 DOI: 10.1016/j.amjmed.2018.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 01/23/2018] [Accepted: 02/20/2018] [Indexed: 12/19/2022]
Abstract
The 2017-2018 influenza season reminds us that it is important for health care professionals to be prepared for the annual onslaught of this contagious respiratory disease associated with potentially serious complications. Vaccination is by far the best method to prevent and control influenza, reducing illness, hospitalizations, and mortality. The highest rates of influenza-associated morbidity and mortality are observed in older adults. The immune function of older adults decreases with increasing age, a phenomenon termed immunosenescence. Immunosenescence not only confers increased susceptibility to influenza disease, but also renders vaccination less effective. To address the need for improved vaccines that provide enhanced protection to this high-risk group, 2 formulations-a high-dose vaccine and an adjuvanted vaccine-have been approved in recent years specifically for people aged 65 years and over. Here we discuss: the challenges of influenza immunization in those 65 years and older; the recent advancements in vaccines targeted at this age group; and the latest influenza vaccine recommendations for the 2017-2018 influenza season in the United States.
Collapse
Affiliation(s)
- William Schaffner
- Department of Health Policy and Division of Infectious Diseases, School of Medicine, Vanderbilt University, Nashville, Tenn.
| | - Wilbur H Chen
- Center for Vaccine Development, School of Medicine, University of Maryland, Baltimore
| | - Robert H Hopkins
- Division of General Internal Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock
| | - Kathleen Neuzil
- Center for Vaccine Development, School of Medicine, University of Maryland, Baltimore
| |
Collapse
|
41
|
Shukla NM, Arimoto KI, Yao S, Fan JB, Zhang Y, Sato-Kaneko F, Lao FS, Hosoya T, Messer K, Pu M, Cottam HB, Carson DA, Hayashi T, Zhang DE, Corr M. Identification of Compounds That Prolong Type I Interferon Signaling as Potential Vaccine Adjuvants. SLAS DISCOVERY 2018; 23:960-973. [PMID: 29751735 DOI: 10.1177/2472555218774308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vaccines are reliant on adjuvants to enhance the immune stimulus, and type I interferons (IFNs) have been shown to be beneficial in augmenting this response. We were interested in identifying compounds that would sustain activation of an endogenous type I IFN response as a co-adjuvant. We began with generation of a human monocytic THP-1 cell line with an IFN-stimulated response element (ISRE)-β-lactamase reporter construct for high-throughput screening. Pilot studies were performed to optimize the parameters and conditions for this cell-based Förster resonance energy transfer (FRET) reporter assay for sustaining an IFN-α-induced ISRE activation signal. These conditions were confirmed in an initial pilot screen, followed by the main screen for evaluating prolongation of an IFN-α-induced ISRE activation signal at 16 h. Hit compounds were identified using a structure enrichment strategy based on chemoinformatic clustering and a naïve "Top X" approach. A select list of confirmed hits was then evaluated for toxicity and the ability to sustain IFN activity by gene and protein expression. Finally, for proof of concept, a panel of compounds was used to immunize mice as co-adjuvant with a model antigen and an IFN-inducing Toll-like receptor 4 agonist, lipopolysaccharide, as an adjuvant. Selected compounds significantly augmented antigen-specific immunoglobulin responses.
Collapse
Affiliation(s)
- Nikunj M Shukla
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kei-Ichiro Arimoto
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Shiyin Yao
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jun-Bao Fan
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Yue Zhang
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Fumi Sato-Kaneko
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Fitzgerald S Lao
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Tadashi Hosoya
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Karen Messer
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,2 Department of Family Medicine and Public Health, Division of Biostatistics and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Minya Pu
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,2 Department of Family Medicine and Public Health, Division of Biostatistics and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Howard B Cottam
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Dennis A Carson
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Dong-Er Zhang
- 1 Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA.,3 Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- 4 Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
42
|
Wang J, Hilchey SP, DeDiego M, Perry S, Hyrien O, Nogales A, Garigen J, Amanat F, Huertas N, Krammer F, Martinez-Sobrido L, Topham DJ, Treanor JJ, Sangster MY, Zand MS. Broad cross-reactive IgG responses elicited by adjuvanted vaccination with recombinant influenza hemagglutinin (rHA) in ferrets and mice. PLoS One 2018; 13:e0193680. [PMID: 29641537 PMCID: PMC5894995 DOI: 10.1371/journal.pone.0193680] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/15/2018] [Indexed: 12/17/2022] Open
Abstract
Annual immunization against influenza virus is a large international public health effort. Accumulating evidence suggests that antibody mediated cross-reactive immunity against influenza hemagglutinin (HA) strongly correlates with long-lasting cross-protection against influenza virus strains that differ from the primary infection or vaccination strain. However, the optimal strategies for achieving highly cross-reactive antibodies to the influenza virus HA have not yet to be defined. In the current study, using Luminex-based mPlex-Flu assay, developed by our laboratory, to quantitatively measure influenza specific IgG antibody mediated cross-reactivity, we found that prime-boost-boost vaccination of ferrets with rHA proteins admixed with adjuvant elicited higher magnitude and broader cross-reactive antibody responses than that induced by actual influenza viral infection, and this cross-reactive response likely correlated with increased anti-stalk reactive antibodies. We observed a similar phenomenon in mice receiving three sequential vaccinations with rHA proteins from either A/California/07/2009 (H1N1) or A/Hong Kong/1/1968 (H3N2) viruses admixed with Addavax, an MF59-like adjuvant. Using this same mouse vaccination model, we determined that Addavax plays a more significant role in the initial priming event than in subsequent boosts. We also characterized the generation of cross-reactive antibody secreting cells (ASCs) and memory B cells (MBCs) when comparing vaccination to viral infection. We have also found that adjuvant plays a critical role in the generation of long-lived ASCs and MBCs cross-reactive to influenza viruses as a result of vaccination with rHA of influenza virus, and the observed increase in stalk-reactive antibodies likely contributes to this IgG mediated broad cross-reactivity.
Collapse
Affiliation(s)
- Jiong Wang
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Shannon P. Hilchey
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Marta DeDiego
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Sheldon Perry
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ollivier Hyrien
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aitor Nogales
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jessica Garigen
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Fatima Amanat
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Nelson Huertas
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Luis Martinez-Sobrido
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - David J. Topham
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - John J. Treanor
- Division of Infectious Disease, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Mark Y. Sangster
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Martin S. Zand
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
- Rochester Center for Health Informatics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
43
|
Efforts to Improve the Seasonal Influenza Vaccine. Vaccines (Basel) 2018; 6:vaccines6020019. [PMID: 29601497 PMCID: PMC6027170 DOI: 10.3390/vaccines6020019] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 01/22/2023] Open
Abstract
Influenza viruses infect approximately 20% of the global population annually, resulting in hundreds of thousands of deaths. While there are Food and Drug Administration (FDA) approved antiviral drugs for combating the disease, vaccination remains the best strategy for preventing infection. Due to the rapid mutation rate of influenza viruses, vaccine formulations need to be updated every year to provide adequate protection. In recent years, a great amount of effort has been focused on the development of a universal vaccine capable of eliciting broadly protective immunity. While universal influenza vaccines clearly have the best potential to provide long-lasting protection against influenza viruses, the timeline for their development, as well as the true universality of protection they afford, remains uncertain. In an attempt to reduce influenza disease burden while universal vaccines are developed and tested, many groups are working on a variety of strategies to improve the efficacy of the standard seasonal vaccine. This review will highlight the different techniques and technologies that have been, or are being, developed to improve the seasonal vaccination efforts against influenza viruses.
Collapse
|
44
|
Clemens EB, van de Sandt C, Wong SS, Wakim LM, Valkenburg SA. Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine. Vaccines (Basel) 2018; 6:vaccines6020018. [PMID: 29587436 PMCID: PMC6027237 DOI: 10.3390/vaccines6020018] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Next-generation vaccines that utilize T cells could potentially overcome the limitations of current influenza vaccines that rely on antibodies to provide narrow subtype-specific protection and are prone to antigenic mismatch with circulating strains. Evidence from animal models shows that T cells can provide heterosubtypic protection and are crucial for immune control of influenza virus infections. This has provided hope for the design of a universal vaccine able to prime against diverse influenza virus strains and subtypes. However, multiple hurdles exist for the realisation of a universal T cell vaccine. Overall primary concerns are: extrapolating human clinical studies, seeding durable effective T cell resident memory (Trm), population human leucocyte antigen (HLA) coverage, and the potential for T cell-mediated immune escape. Further comprehensive human clinical data is needed during natural infection to validate the protective role T cells play during infection in the absence of antibodies. Furthermore, fundamental questions still exist regarding the site, longevity and duration, quantity, and phenotype of T cells needed for optimal protection. Standardised experimental methods, and eventually simplified commercial assays, to assess peripheral influenza-specific T cell responses are needed for larger-scale clinical studies of T cells as a correlate of protection against influenza infection. The design and implementation of a T cell-inducing vaccine will require a consensus on the level of protection acceptable in the community, which may not provide sterilizing immunity but could protect the individual from severe disease, reduce the length of infection, and potentially reduce transmission in the community. Therefore, increasing the standard of care potentially offered by T cell vaccines should be considered in the context of pandemic preparedness and zoonotic infections, and in combination with improved antibody vaccine targeting methods. Current pandemic vaccine preparedness measures and ongoing clinical trials under-utilise T cell-inducing vaccines, reflecting the myriad questions that remain about how, when, where, and which T cells are needed to fight influenza virus infection. This review aims to bring together basic fundamentals of T cell biology with human clinical data, which need to be considered for the implementation of a universal vaccine against influenza that harnesses the power of T cells.
Collapse
Affiliation(s)
- E Bridie Clemens
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Carolien van de Sandt
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sook San Wong
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sophie A Valkenburg
- HKU Pasteur Research Pole, School of Public Health, University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
45
|
Zhao C, Kim Y, Zeng Y, Li M, Wang X, Hu C, Gorman C, Dai SY, Ding SY, Yuan JS. Co-Compartmentation of Terpene Biosynthesis and Storage via Synthetic Droplet. ACS Synth Biol 2018; 7:774-781. [PMID: 29439563 DOI: 10.1021/acssynbio.7b00368] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Traditional bioproduct engineering focuses on pathway optimization, yet is often complicated by product inhibition, downstream consumption, and the toxicity of certain products. Here, we present the co-compartmentation of biosynthesis and storage via a synthetic droplet as an effective new strategy to improve the bioproduct yield, with squalene as a model compound. A hydrophobic protein was designed and introduced into the tobacco chloroplast to generate a synthetic droplet for terpene storage. Simultaneously, squalene biosynthesis enzymes were introduced to chloroplasts together with the droplet-forming protein to co-compartmentalize the biosynthesis and storage of squalene. The strategy has enabled a record yield of squalene at 2.6 mg/g fresh weight without compromising plant growth. Confocal fluorescent microscopy imaging, stimulated Raman scattering microscopy, and droplet composition analysis confirmed the formation of synthetic storage droplet in chloroplast. The co-compartmentation of synthetic storage droplet with a targeted metabolic pathway engineering represents a new strategy for enhancing bioproduct yield.
Collapse
Affiliation(s)
- Cheng Zhao
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - YongKyoung Kim
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - Yining Zeng
- Biosciences Center , National Renewable Energy Laboratory , Golden , Colorado 80401 , United States
| | - Man Li
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - Xin Wang
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - Cheng Hu
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - Connor Gorman
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| | - Susie Y Dai
- State Hygienic Lab , University of Iowa , Coralville , Iowa 52241 , United States
| | - Shi-You Ding
- Department of Plant Biology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Joshua S Yuan
- Texas A&M Agrilife Synthetic and Systems Biology Innovation Hub , Texas A&M University , College Station , Texas 77843 , United States
- Department of Plant Pathology and Microbiology , Texas A&M University , College Station , Texas 77843 , United States
- Institute for Plant Genomics and Biotechnology , Texas A&M University , College Station , Texas 77843 , United States
| |
Collapse
|
46
|
Yang LY, Zhou H, Yang Y, Tong YN, Peng LS, Zhu BH, Diao WB, Zeng H, Sun HW, Zou QM. Protective effects of a nanoemulsion adjuvant vaccine (2C-Staph/NE) administered intranasally against invasive Staphylococcus aureus pneumonia. RSC Adv 2018; 8:9996-10008. [PMID: 35540845 PMCID: PMC9078739 DOI: 10.1039/c7ra13630g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 02/13/2018] [Indexed: 12/03/2022] Open
Abstract
No licensed Staphylococcus aureus (S. aureus) vaccine is currently available. To develop an effective S. aureus vaccine, we selected the recombinant proteins staphylococcal enterotoxin B (rSEB) and manganese transport protein C (rMntC) as vaccine candidates and formulated a 2C-Staph vaccine. Based on the optimised formation of nanoemulsion (NE) technology, we constructed a novel NE adjuvant vaccine, 2C-Staph/NE. The 2C-Staph/NE particles showed a suitable diameter (24.9 ± 0.14 nm), a good protein structure of integrity and specificity, and high thermodynamic stability. 2C-Staph formulated with an NE adjuvant induced higher survival rates than a 2C-Staph/MF59 vaccine in sepsis and pneumonia models. Moreover, intramuscular vaccination with 2C-Staph/NE yielded protection efficacy in a sepsis model, and the intranasal vaccination route induced a potent protective effect in a pneumonia model. Intranasal vaccination with 2C-Staph/NE induced a strong mucosal response with high levels of IgA and IL-17A in bronchoalveolar lavage fluid (BALF), and the IgG levels in the BALF were comparable to those induced by the intramuscular vaccination route. Furthermore, the serum and BALF induced by intranasal administration showed potent opsonophagocytic activity against S. aureus. And, the IL-17A played a protective role in the pneumonia model demonstrated by a cytokine neutralization test. Taken together, our results showed that intranasal administration of 2C-Staph formulated with an NE adjuvant yielded ideal protection in a murine S. aureus pneumonia model.
Collapse
Affiliation(s)
- Liu-Yang Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Heng Zhou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Yun Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Ya-Nan Tong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Liu-Sheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Bao-Hang Zhu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Wei-Bo Diao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Hong-Wu Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| | - Quan-Ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University Chongqing 400038 PR China
| |
Collapse
|
47
|
Synthetic Toll-Like Receptor 4 (TLR4) and TLR7 Ligands Work Additively via MyD88 To Induce Protective Antiviral Immunity in Mice. J Virol 2017; 91:JVI.01050-17. [PMID: 28724768 DOI: 10.1128/jvi.01050-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/17/2017] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated that the combination of synthetic small-molecule Toll-like receptor 4 (TLR4) and TLR7 ligands is a potent adjuvant for recombinant influenza virus hemagglutinin, inducing rapid and sustained immunity that is protective against influenza viruses in homologous, heterologous, and heterosubtypic murine challenge models. Combining the TLR4 and TLR7 ligands balances Th1 and Th2-type immune responses for long-lived cellular and neutralizing humoral immunity against the viral hemagglutinin. Here, we demonstrate that the protective response induced in mice by this combined adjuvant is dependent upon TLR4 and TLR7 signaling via myeloid differentiation primary response gene 88 (MyD88), indicating that the adjuvants function in vivo via their known receptors, with negligible off-target effects, to induce protective immunity. The combined adjuvant acts via MyD88 in both bone marrow-derived and non-bone marrow-derived radioresistant cells to induce hemagglutinin-specific antibodies and protect mice against influenza virus challenge. The protective efficacy generated by immunization with this adjuvant and recombinant hemagglutinin antigen is transferable with serum from immunized mice to recipient mice in a homologous, but not a heterologous, H1N1 viral challenge model. Depletion of CD4+ cells after an established humoral response in immunized mice does not impair protection from a homologous challenge; however, it does significantly impair recovery from a heterologous challenge virus, highlighting an important role for vaccine-induced CD4+ cells in cross-protective vaccine efficacy. The combination of the two TLR agonists allows for significant dose reductions of each component to achieve a level of protection equivalent to that afforded by either single agent at its full dose.IMPORTANCE Development of novel adjuvants is needed to enhance immunogenicity to provide better protection from seasonal influenza virus infection and improve pandemic preparedness. We show here that several dose combinations of synthetic TLR4 and TLR7 ligands are potent adjuvants for recombinant influenza virus hemagglutinin antigen induction of humoral and cellular immunity against viral challenges. The components of the combined adjuvant work additively to enable both antigen and adjuvant dose sparing while retaining efficacy. Understanding an adjuvant's mechanism of action is a critical component for preclinical safety evaluation, and we demonstrate here that a combined TLR4 and TLR7 adjuvant signals via the appropriate receptors and the MyD88 adaptor protein. This novel adjuvant combination contributes to a more broadly protective vaccine while demonstrating an attractive safety profile.
Collapse
|
48
|
Chan M, Ahmadi A, Yao S, Sato-Kaneko F, Messer K, Pu M, Nguyen B, Hayashi T, Corr M, Carson DA, Cottam HB, Shukla NM. Identification of Biologically Active Pyrimido[5,4-b]indoles That Prolong NF-κB Activation without Intrinsic Activity. ACS COMBINATORIAL SCIENCE 2017; 19:533-543. [PMID: 28657707 DOI: 10.1021/acscombsci.7b00080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Most vaccine adjuvants directly stimulate and activate antigen presenting cells but do not sustain immunostimulation of these cells. A high throughput screening (HTS) strategy was designed to identify compounds that would sustain NF-κB activation by a stimulus from the Toll-like receptor (TLR)4 ligand, lipopolysaccharide (LPS). Several pilot studies optimized the parameters and conditions for a cell based NF-κB reporter assay in human monocytic THP-1 cells. The final assay evaluated prolongation of LPS induced NF-κB activation at 12 h. The dynamic range of the assay was confirmed in a pilot screen of 14 631 compounds and subsequently in a main extensive screen with 166 304 compounds. Hit compounds were identified using an enrichment strategy based on unsupervised chemoinformatic clustering, and also by a naı̈ve "Top X" approach. A total of 2011 compounds were then rescreened for levels of coactivation with LPS at 5 h and 12 h, which provided kinetic profiles. Of the 407 confirmed hits, compounds that showed correlation of the kinetic profiles with the structural similarities led to identification of four chemotypes: pyrimido[5,4-b]indoles, 4H-chromene-3-carbonitriles, benzo[d][1,3]dioxol-2-ylureas, and tetrahydrothieno[2,3-c]pyridines, which were segregated by 5 h and 12 h kinetic characteristics. Unlike the TLR4 agonistic pyrimidoindoles identified in previous studies, the revealed pyrimidoindoles in the present work did not intrinsically stimulate TLR4 nor induce NF-κB but rather prolonged NF-κB signaling induced by LPS. A 42-member combinatorial library was synthesized which led to identification of potent N3-alkyl substituted pyrimidoindoles that were not only active in vitro but also enhanced antibody responses in vivo when used as a coadjuvant. The novel HTS strategy led to identification of compounds that are intrinsically quiescent but functionally prolong stimulation by a TLR4 ligand and thereby potentiate vaccine efficacy.
Collapse
Affiliation(s)
- Michael Chan
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Alast Ahmadi
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Shiyin Yao
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Fumi Sato-Kaneko
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Karen Messer
- Division
of Biostatistics, University of California San Diego, La Jolla, California 92093, United States
| | - Minya Pu
- Division
of Biostatistics, University of California San Diego, La Jolla, California 92093, United States
| | - Brandon Nguyen
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Tomoko Hayashi
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Maripat Corr
- Department
of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Dennis A. Carson
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Howard B. Cottam
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Nikunj M. Shukla
- Moores
UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
49
|
Shim SM, Song EJ, Song D, Lee TY, Kim DJ, Nam JH, Gwin Jeong D, Lee CK, Kim SH, Kim JK. Nontoxic outer membrane vesicles efficiently increase the efficacy of an influenza vaccine in mice and ferrets. Vaccine 2017; 35:3741-3748. [PMID: 28576571 DOI: 10.1016/j.vaccine.2017.05.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 02/06/2023]
Abstract
In this study, we developed a further-modified outer membrane vesicle (fmOMV) from the ΔmsbB/ΔpagP mutant of Escherichia coli transformed with the plasmid, pLpxF, in order to use it as an adjuvant for pandemic H1N1 (pH1N1) influenza vaccine. We evaluated the efficacy of the pH1N1 influenza vaccine containing the fmOMV in animal models as compared to the commercial adjuvants, alum or AddaVaxTM. The fmOMV-adjuvanted pH1N1 influenza vaccine induced a significant increase in the humoral immunity; however, this effect was less than that of the AddaVaxTM. The fmOMV-adjuvanted vaccine displayed pronounced an enhanced protective efficacy with increased T cell immune response and reduced the viral load in the lungs of the infected mice after challenging them with a lethal dose of the homologous virus. Moreover, it resulted in a significantly higher cross-protection against heterologous virus challenge than that of the pH1N1 vaccine with alum or with no adjuvants. In ferrets, the fmOMV-adjuvanted vaccine elicited a superior antibody response based on the HI titer and efficiently protected the animals from the lethal viral challenges. Taken together, the nontoxic fmOMV could be a promising adjuvant for inducing robust T cell priming into the pH1N1 vaccine and might be broadly applicable to the development of preventive measures against influenza virus infection.
Collapse
Affiliation(s)
- Sang-Mu Shim
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Eun-Jung Song
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Daesub Song
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Tae-Young Lee
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Doo-Jin Kim
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Hyun Nam
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Dae Gwin Jeong
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Chong-Kil Lee
- Department of Pharmacy, College of Pharmacy, Chungbuk National University, 52 Naesudong-ro, Heungdeok-gu, Cheongju, Chungcheongbuk-do 28644, Republic of Korea
| | - Sang-Hyun Kim
- College of Veterinary Medicine, Gyeongsang National University, 501 Jinjudae-ro, Jinju, Gyeongsangnam-do 52828, Republic of Korea.
| | - Jeong-Ki Kim
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea.
| |
Collapse
|
50
|
Inactivated influenza virus vaccines: the future of TIV and QIV. Curr Opin Virol 2017; 23:102-106. [PMID: 28505524 DOI: 10.1016/j.coviro.2017.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/24/2017] [Indexed: 11/20/2022]
Abstract
Influenza viruses continue to be a major public health concern, despite the availability of vaccines. Currently licensed influenza vaccines aim at the induction of antibodies that target hemagglutinin, the major antigenic determinant on the surface of influenza virions that is responsible for attachment of the virus to the host cell that is to be infected. Currently licensed influenza vaccines come as inactivated or live attenuated influenza vaccines and are trivalent or quadrivalent as they contain antigens of two influenza A and one or two influenza B strains that circulate in the human population, respectively. In this review we briefly compare trivalent and quadrivalent inactivated influenza vaccines (TIV and QIV) with live attenuated influenza vaccines (LAIV). The use of the latter vaccine type in children age 2-8 has been disrecommended recently by the American Centers for Disease Control and Prevention due to inferior vaccine effectiveness in this age group in recent seasons. This recommendation will favor the use of TIV and QIV over LAIV in the near future. However, there is much evidence from studies in humans that illustrate the benefit of LAIV and we discuss some of the mechanisms that contribute to broader protection against influenza viruses of different subtypes induced by natural infection and LAIV. The future challenge will be to apply these insights to allow induction of broader and long-lasting protection provided by TIV and QIV vaccines, for example, by the use of adjuvants or combining LAIV with TIV and QIV. Other immune factors than serum hemagglutination inhibiting antibodies have shown to correlate with protection provided by TIV and QIV, which illustrates the need for other correlates of protection than hemagglutination inhibition by serum antibodies and justifies more focus on influenza antigens in the TIV and QIV other than hemagglutinin.
Collapse
|