1
|
Jung M, Kim H, Choi E, Shin MK, Shin SJ. Enhancing vaccine effectiveness in the elderly to counter antibiotic resistance: The potential of adjuvants via pattern recognition receptors. Hum Vaccin Immunother 2024; 20:2317439. [PMID: 39693178 DOI: 10.1080/21645515.2024.2317439] [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: 10/31/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 12/20/2024] Open
Abstract
Vaccines are an effective way to prevent the emergence and spread of antibiotic resistance by preventing diseases and establishing herd immunity. However, the reduced effectiveness of vaccines in the elderly due to immunosenescence is one of the significant contributors to the increasing antibiotic resistance. To counteract this decline and enhance vaccine effectiveness in the elderly, adjuvants play a pivotal role. Adjuvants are designed to augment the effectiveness of vaccines by activating the innate immune system, particularly through pattern recognition receptors on antigen-presenting cells. To improve vaccine effectiveness in the elderly using adjuvants, it is imperative to select the appropriate adjuvants based on an understanding of immunosenescence and the mechanisms of adjuvant functions. This review demonstrates the phenomenon of immunosenescence and explores various types of adjuvants, including their mechanisms and their potential in improving vaccine effectiveness for the elderly, thereby contributing to developing more effective vaccines for this vulnerable demographic.
Collapse
Affiliation(s)
- Myunghwan Jung
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Gyu Choi H, Woong Kwon K, Jae Shin S. Importance of adjuvant selection in tuberculosis vaccine development: Exploring basic mechanisms and clinical implications. Vaccine X 2023; 15:100400. [PMID: 37965276 PMCID: PMC10641539 DOI: 10.1016/j.jvacx.2023.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
The global emergency of unexpected pathogens, exemplified by SARS-CoV-2, has emphasized the importance of vaccines in thwarting infection and curtailing the progression of severe disease. The scourge of tuberculosis (TB), emanating from the Mycobacterium tuberculosis (Mtb) complex, has inflicted a more profound toll in terms of mortality and morbidity than any other infectious agents prior to the SARS-CoV-2 pandemic. Despite the existence of Bacillus Calmette-Guérin (BCG), the only licensed vaccine developed a century ago, its efficacy against TB remains unsatisfactory, particularly in preventing pulmonary Mtb infections in adolescents and adults. However, collaborations between academic and industrial entities have led to a renewed impetus in the development of TB vaccines, with numerous candidates, particularly subunit vaccines with specialized adjuvants, exhibiting promising outcomes in recent clinical studies. Adjuvants are crucial in modulating optimal immunological responses, by endowing immune cells with sufficient antigen and immune signals. As exemplified by the COVID-19 vaccine landscape, the interplay between vaccine efficacy and adverse effects is of paramount importance, particularly for the elderly and individuals with underlying ailments such as diabetes and concurrent infections. In this regard, adjuvants hold the key to optimizing vaccine efficacy and safety. This review accentuates the pivotal roles of adjuvants and their underlying mechanisms in the development of TB vaccines. Furthermore, we expound on the prospects for the development of more efficacious adjuvants and their synergistic combinations for individuals in diverse states, such as aging, HIV co-infection, and diabetes, by examining the immunological alterations that arise with aging and comparing them with those observed in younger cohorts.
Collapse
Affiliation(s)
- Han Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
3
|
Moni SS, Abdelwahab SI, Jabeen A, Elmobark ME, Aqaili D, Ghoal G, Oraibi B, Farasani AM, Jerah AA, Alnajai MMA, Mohammad Alowayni AMH. Advancements in Vaccine Adjuvants: The Journey from Alum to Nano Formulations. Vaccines (Basel) 2023; 11:1704. [PMID: 38006036 PMCID: PMC10674458 DOI: 10.3390/vaccines11111704] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Vaccination is a groundbreaking approach in preventing and controlling infectious diseases. However, the effectiveness of vaccines can be greatly enhanced by the inclusion of adjuvants, which are substances that potentiate and modulate the immune response. This review is based on extensive searches in reputable databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. The goal of this review is to provide a thorough analysis of the advances in the field of adjuvant research, to trace the evolution, and to understand the effects of the various adjuvants. Historically, alum was the pioneer in the field of adjuvants because it was the first to be approved for use in humans. It served as the foundation for subsequent research and innovation in the field. As science progressed, research shifted to identifying and exploiting the potential of newer adjuvants. One important area of interest is nano formulations. These advanced adjuvants have special properties that can be tailored to enhance the immune response to vaccines. The transition from traditional alum-based adjuvants to nano formulations is indicative of the dynamism and potential of vaccine research. Innovations in adjuvant research, particularly the development of nano formulations, are a promising step toward improving vaccine efficacy and safety. These advances have the potential to redefine the boundaries of vaccination and potentially expand the range of diseases that can be addressed with this approach. There is an optimistic view of the future in which improved vaccine formulations will contribute significantly to improving global health outcomes.
Collapse
Affiliation(s)
- Sivakumar S. Moni
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | | | - Aamena Jabeen
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | - Mohamed Eltaib Elmobark
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | - Duaa Aqaili
- Physiology Department, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| | - Gassem Ghoal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| | - Bassem Oraibi
- Medical Research Centre, Jazan University, Jazan 45142, Saudi Arabia (B.O.)
| | | | - Ahmed Ali Jerah
- College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Mahdi Mohammed A. Alnajai
- General Directorate of Health Services and University Hospital, Jazan University, Jazan 45142, Saudi Arabia;
| | | |
Collapse
|
4
|
Zhang Y, Xu JC, Hu ZD, Fan XY. Advances in protein subunit vaccines against tuberculosis. Front Immunol 2023; 14:1238586. [PMID: 37654500 PMCID: PMC10465801 DOI: 10.3389/fimmu.2023.1238586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
Tuberculosis (TB), also known as the "White Plague", is caused by Mycobacterium tuberculosis (Mtb). Before the COVID-19 epidemic, TB had the highest mortality rate of any single infectious disease. Vaccination is considered one of the most effective strategies for controlling TB. Despite the limitations of the Bacille Calmette-Guérin (BCG) vaccine in terms of protection against TB among adults, it is currently the only licensed TB vaccine. Recently, with the evolution of bioinformatics and structural biology techniques to screen and optimize protective antigens of Mtb, the tremendous potential of protein subunit vaccines is being exploited. Multistage subunit vaccines obtained by fusing immunodominant antigens from different stages of TB infection are being used both to prevent and to treat TB. Additionally, the development of novel adjuvants is compensating for weaknesses of immunogenicity, which is conducive to the flourishing of subunit vaccines. With advances in the development of animal models, preclinical vaccine protection assessments are becoming increasingly accurate. This review summarizes progress in the research of protein subunit TB vaccines during the past decades to facilitate the further optimization of protein subunit vaccines that may eradicate TB.
Collapse
Affiliation(s)
- Ying Zhang
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jin-chuan Xu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zhi-dong Hu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| | - Xiao-yong Fan
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Wu N, Chen Q, Zou Y, Miao C, Ma G, Wu J. Chitosan particle-emulsion complex adjuvants: The effect of particle distribution on the immune intensity and response type. Carbohydr Polym 2023; 309:120673. [PMID: 36906359 DOI: 10.1016/j.carbpol.2023.120673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Particle-emulsion complex adjuvants as a new trend in the research of vaccine formulation, can improve the immune strength and balance the immune type. However, the location of the particle in the formulation is a key factor that has not been investigated extensively and its type of immunity. In order to investigate the effect of different combining modes of emulsion and particle on the immune response, three types of particle-emulsion complex adjuvant formulations were designed with the combination of chitosan nanoparticles (CNP) and an o/w emulsion with squalene as the oil phase. The complex adjuvants included the CNP-I group (particle inside the emulsion droplet), CNP-S group (particle on the surface of emulsion droplet) and CNP-O group (particle outside the emulsion droplet), respectively. The formulations with different particle locations behaved with different immunoprotective effects and immune-enhancing mechanisms. Compared with CNP-O, CNP-I and CNP-S significantly improve humoral and cellular immunity. CNP-O was more like two independent systems for immune enhancement. As a result, CNP-S triggered a Th1-type immune bias and CNP-I had more of a Th2-type of the immune response. These data highlight the key influence of the subtle difference of particle location in the droplets for immune response.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qiuting Chen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Yongjuan Zou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
6
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
7
|
Liang H, Lykins WR, Seydoux E, Guderian JA, Phan T, Fox CB, Orr MT. Formulated Phospholipids as Non-Canonical TLR4 Agonists. Pharmaceutics 2022; 14:2557. [PMID: 36559051 PMCID: PMC9788208 DOI: 10.3390/pharmaceutics14122557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic agents known as adjuvants play a critical role in many vaccine formulations. Adjuvants often signal through Toll-like receptor (TLR) pathways, including formulations in licensed vaccines that target TLR4. While TLR4 is predominantly known for responding to lipopolysaccharide (LPS), a component of Gram-negative bacterial membranes, it has been shown to be a receptor for a number of molecular structures, including phospholipids. Therefore, phospholipid-based pharmaceutical formulations might have off-target effects by signaling through TLR4, confounding interpretation of pharmaceutical bioactivity. In this study we examined the individual components of a clinical stage oil-in-water vaccine adjuvant emulsion (referred to as a stable emulsion or SE) and their ability to signal through murine and human TLR4s. We found that the phospholipid 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) activated TLR4 and elicited many of the same immune phenotypes as canonical TLR4 agonists. This pathway was dependent on the saturation, size, and headgroup of the phospholipid. Interestingly, DMPC effects on human cells were evident but overall appeared less impactful than emulsion oil composition. Considering the prevalence of DMPC and other phospholipids used across the pharmaceutical space, these findings may contextualize off-target innate immune responses that could impact preclinical and clinical development.
Collapse
Affiliation(s)
- Hong Liang
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - William R. Lykins
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Emilie Seydoux
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Jeffrey A. Guderian
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Tony Phan
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Christopher B. Fox
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| | - Mark T. Orr
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Liu ZM, Yang MH, Yu K, Lian ZX, Deng SL. Toll-like receptor (TLRs) agonists and antagonists for COVID-19 treatments. Front Pharmacol 2022; 13:989664. [PMID: 36188605 PMCID: PMC9518217 DOI: 10.3389/fphar.2022.989664] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) rapidly infects humans and animals which make coronavirus disease 2019 (COVID-19) a grievous epidemic worldwide which broke out in 2020. According to data analysis of the other coronavirus family, for instance severe acute respiratory syndrome SARS coronavirus (SARS-CoV), can provide experience for the mutation of SARS-CoV-2 and the prevention and treatment of COVID-19. Toll-like receptors (TLRs) as a pattern recognition receptor (PRRs), have an indispensable function in identifying the invader even activate the innate immune system. It is possible for organism to activate different TLR pathways which leads to secretion of proinflammatory cytokines such as Interleukin 1 (IL-1), Interleukin 6 (IL-6), Tumor necrosis factor α (TNFα) and type Ⅰ interferon. As a component of non-specific immunity, TLRs pathway may participate in the SARS-CoV-2 pathogenic processes, due to previous works have proved that TLRs are involved in the invasion and infection of SARS-CoV and MERS to varying degrees. Different TLR, such as TLR2, TLR4, TLR7, TLR8 and TLR9 probably have a double-sided in COVID-19 infection. Therefore, it is of great significance for a correctly acknowledging how TLR take part in the SARS-CoV-2 pathogenic processes, which will be the development of treatment and prevention strategies.
Collapse
Affiliation(s)
- Zhi-Mei Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ming-Hui Yang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, China
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| |
Collapse
|
9
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
10
|
Rostamizadeh L, Molavi O, Rashid M, Ramazani F, Baradaran B, Lavasanaifar A, Lai R. Recent advances in cancer immunotherapy: Modulation of tumor microenvironment by Toll-like receptor ligands. BIOIMPACTS : BI 2022; 12:261-290. [PMID: 35677663 PMCID: PMC9124882 DOI: 10.34172/bi.2022.23896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
![]()
Immunotherapy is considered a promising approach for cancer treatment. An important strategy for cancer immunotherapy is the use of cancer vaccines, which have been widely used for cancer treatment. Despite the great potential of cancer vaccines for cancer treatment, their therapeutic effects in clinical settings have been limited. The main reason behind the lack of significant therapeutic outcomes for cancer vaccines is believed to be the immunosuppressive tumor microenvironment (TME). The TME counteracts the therapeutic effects of immunotherapy and provides a favorable environment for tumor growth and progression. Therefore, overcoming the immunosuppressive TME can potentially augment the therapeutic effects of cancer immunotherapy in general and therapeutic cancer vaccines in particular. Among the strategies developed for overcoming immunosuppression in TME, the use of toll-like receptor (TLR) agonists has been suggested as a promising approach to reverse immunosuppression. In this paper, we will review the application of the four most widely studied TLR agonists including agonists of TLR3, 4, 7, and 9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Rostamizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramazani
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanaifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Canada
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
11
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
12
|
O'Hagan DT, van der Most R, Lodaya RN, Coccia M, Lofano G. "World in motion" - emulsion adjuvants rising to meet the pandemic challenges. NPJ Vaccines 2021; 6:158. [PMID: 34934069 PMCID: PMC8692316 DOI: 10.1038/s41541-021-00418-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
Emulsion adjuvants such as MF59 and AS03 have been used for more than two decades as key components of licensed vaccines, with over 100 million doses administered to diverse populations in more than 30 countries. Substantial clinical experience of effectiveness and a well-established safety profile, along with the ease of manufacturing have established emulsion adjuvants as one of the leading platforms for the development of pandemic vaccines. Emulsion adjuvants allow for antigen dose sparing, more rapid immune responses, and enhanced quality and quantity of adaptive immune responses. The mechanisms of enhancement of immune responses are well defined and typically characterized by the creation of an "immunocompetent environment" at the site of injection, followed by the induction of strong and long-lasting germinal center responses in the draining lymph nodes. As a result, emulsion adjuvants induce distinct immunological responses, with a mixed Th1/Th2 T cell response, long-lived plasma cells, an expanded repertoire of memory B cells, and high titers of cross-neutralizing polyfunctional antibodies against viral variants. Because of these various properties, emulsion adjuvants were included in pandemic influenza vaccines deployed during the 2009 H1N1 influenza pandemic, are still included in seasonal influenza vaccines, and are currently at the forefront of the development of vaccines against emerging SARS-CoV-2 pandemic variants. Here, we comprehensively review emulsion adjuvants, discuss their mechanism of action, and highlight their profile as a benchmark for the development of additional vaccine adjuvants and as a valuable tool to allow further investigations of the general principles of human immunity.
Collapse
|
13
|
Kurup D, Fisher CR, Scher G, Yankowski C, Testa A, Keshwara R, Abreu-Mota T, Lambert R, Ferguson M, Rinaldi W, Ruiz L, Wirblich C, Schnell MJ. Tetravalent Rabies-Vectored Filovirus and Lassa Fever Vaccine Induces Long-term Immunity in Nonhuman Primates. J Infect Dis 2021; 224:995-1004. [PMID: 33421072 PMCID: PMC8448432 DOI: 10.1093/infdis/jiab014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/08/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The objective of this study is to evaluate the immunogenicity of adjuvanted monovalent rabies virus (RABV)-based vaccine candidates against Ebola virus (FILORAB1), Sudan virus (FILORAB2), Marburg virus (FILORAB3), Lassa virus (LASSARAB1), and combined trivalent vaccine candidate (FILORAB1-3) and tetravalent vaccine candidate (FILORAB1-3 and LASSARAB) in nonhuman primates. METHODS Twenty-four Macaca fascicularis were randomly assigned into 6 groups of 4 animals. Each group was vaccinated with either a single adjuvanted vaccine, the trivalent vaccine, or the tetravalent vaccine at days 0 and 28. We followed the humoral immune responses for 1 year by antigen-specific enzyme-linked immunosorbent assays and RABV neutralization assays. RESULTS High titers of filovirus and/or Lassa virus glycoprotein-specific immunoglobulin G were induced in the vaccinated animals. There were no significant differences between immune responses in animals vaccinated with single vaccines vs trivalent or tetravalent vaccines. In addition, all vaccine groups elicited strong rabies neutralizing antibody titers. The antigen-specific immune responses were detectable for 1 year in all groups. CONCLUSIONS In summary, this study shows the longevity of the immune responses up to 365 days for a pentavalent vaccine-against Ebola virus, Sudan virus, Marburg virus, Lassa virus, and RABV-using a safe and effective vaccine platform.
Collapse
Affiliation(s)
- Drishya Kurup
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christine R Fisher
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Gabrielle Scher
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Catherine Yankowski
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - AnnaMarie Testa
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Rohan Keshwara
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Tiago Abreu-Mota
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Rachael Lambert
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | - Christoph Wirblich
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Jefferson Vaccine Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Zeigler DF, Gage E, Clegg CH. Epitope-targeting platform for broadly protective influenza vaccines. PLoS One 2021; 16:e0252170. [PMID: 34043704 PMCID: PMC8158873 DOI: 10.1371/journal.pone.0252170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/10/2021] [Indexed: 11/18/2022] Open
Abstract
Seasonal influenza vaccines are often ineffective because they elicit strain-specific antibody responses to mutation-prone sites on the hemagglutinin (HA) head. Vaccines that provide long-lasting immunity to conserved epitopes are needed. Recently, we reported a nanoparticle-based vaccine platform produced by solid-phase peptide synthesis (SPPS) for targeting linear and helical protein-based epitopes. Here, we illustrate its potential for building broadly protective influenza vaccines. Targeting known epitopes in the HA stem, neuraminidase (NA) active site, and M2 ectodomain (M2e) conferred 50-75% survival against 5LD50 influenza B and H1N1 challenge; combining stem and M2e antigens increased survival to 90%. Additionally, protein sequence and structural information were employed in tandem to identify alternative epitopes that stimulate greater protection; we report three novel HA and NA sites that are highly conserved in type B viruses. One new target in the HA stem stimulated 100% survival, highlighting the value of this simple epitope discovery strategy. A candidate influenza B vaccine targeting two adjacent HA stem sites led to >104-fold reduction in pulmonary viral load. These studies describe a compelling platform for building vaccines that target conserved influenza epitopes.
Collapse
Affiliation(s)
- David F. Zeigler
- TRIA Bioscience Corp., Seattle, Washington, United States of America
| | - Emily Gage
- TRIA Bioscience Corp., Seattle, Washington, United States of America
| | | |
Collapse
|
15
|
Baldwin SL, Reese VA, Larsen SE, Beebe E, Guderian J, Orr MT, Fox CB, Reed SG, Coler RN. Prophylactic efficacy against Mycobacterium tuberculosis using ID93 and lipid-based adjuvant formulations in the mouse model. PLoS One 2021; 16:e0247990. [PMID: 33705411 PMCID: PMC7951850 DOI: 10.1371/journal.pone.0247990] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 11/19/2022] Open
Abstract
An estimated 10 million people developed tuberculosis (TB) disease in 2019 which underscores the need for a vaccine that prevents disease and reduces transmission. The aim of our current studies is to characterize and test a prophylactic tuberculosis vaccine comprised of ID93, a polyprotein fusion antigen, and a liposomal formulation [including a synthetic TLR4 agonist (glucopyranosyl lipid adjuvant, GLA) and QS-21] in a preclinical mouse model of TB disease. Comparisons of the ID93+GLA-LSQ vaccines are also made to the highly characterized ID93+GLA-SE oil-in-water emulsion adjuvant, which are also included these studies. The recent success of vaccine candidate M72 combined with adjuvant AS01E (GlaxoSmithKline Biologicals) in reducing progression to active disease is promising and has renewed excitement for experimental vaccines currently in the TB vaccine pipeline. The AS01E adjuvant contains monophosphoryl lipid A (MPL) and QS-21 (a saponin) in a liposomal formulation. While AS01E has demonstrated potent adjuvant activity as a component of both approved and experimental vaccines, developing alternatives to this adjuvant system will become important to fill the high demand envisioned for future vaccine needs. Furthermore, replacement sources of potent adjuvants will help to supply the demand of a TB vaccine [almost one-quarter of the world's population are estimated to have latent Mycobacterium tuberculosis (Mtb) according to the WHO 2019 global TB report], addressing (a) cost of goods, (b) supply of goods, and (c) improved efficacy of subunit vaccines against Mtb. We show that both ID93+GLA-SE (containing an emulsion adjuvant) and ID93+GLA-LSQ (containing a liposomal adjuvant) induce ID93-specific TH1 cellular immunity including CD4+CD44+ T cells expressing IFNγ, TNF, and IL-2 (using flow cytometry and intracellular cytokine staining) and vaccine-specific IgG2 antibody responses (using an ELISA). In addition, both ID93+GLA-SE and ID93+GLA-LSQ effectively decrease the bacterial load within the lungs of mice infected with Mtb. Formulations based on this liposomal adjuvant formulation may provide an alternative to AS01 adjuvant systems.
Collapse
Affiliation(s)
- Susan L. Baldwin
- Seattle Children’s Research Institute, Seattle, WA, United States of America
- * E-mail:
| | - Valerie A. Reese
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sasha E. Larsen
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Elyse Beebe
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Jeff Guderian
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Mark T. Orr
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Christopher B. Fox
- Infectious Disease Research Institute, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Rhea N. Coler
- Seattle Children’s Research Institute, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
16
|
Bruxelle JF, Kirilenko T, Trattnig N, Yang Y, Cattin M, Kosma P, Pantophlet R. A glycoside analog of mammalian oligomannose formulated with a TLR4-stimulating adjuvant elicits HIV-1 cross-reactive antibodies. Sci Rep 2021; 11:4637. [PMID: 33633304 PMCID: PMC7907241 DOI: 10.1038/s41598-021-84116-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/12/2021] [Indexed: 01/31/2023] Open
Abstract
The occurrence of oligomannose-specific broadly neutralizing antibodies (bnAbs) has spurred efforts to develop immunogens that can elicit similar antibodies. Here, we report on the antigenicity and immunogenicity of a CRM197-conjugate of a previously reported oligomannose mimetic. Oligomannose-specific bnAbs that are less dependent on interactions with the HIV envelope protein sequence showed strong binding to the glycoconjugates, with affinities approximating those reported for their cognate epitope. The glycoconjugate is also recognized by inferred germline precursors of oligomannose-specific bnAbs, albeit with the expected low avidity, supporting its potential as an immunogen. Immunization of human-antibody transgenic mice revealed that only a TLR4-stimulating adjuvant formulation resulted in antibodies able to bind a panel of recombinant HIV trimers. These antibodies bound at relatively modest levels, possibly explaining their inability to neutralize HIV infectivity. Nevertheless, these findings contribute further to understanding conditions for eliciting HIV-cross-reactive oligomannose-specific antibodies and inform on next steps for improving on the elicited response.
Collapse
Affiliation(s)
- Jean-François Bruxelle
- grid.61971.380000 0004 1936 7494Faculty of Health Sciences, Simon Fraser University, Burnaby, BC Canada
| | - Tess Kirilenko
- grid.61971.380000 0004 1936 7494Faculty of Health Sciences, Simon Fraser University, Burnaby, BC Canada ,grid.479077.aPresent Address: AbCellera Biologics Inc., Vancouver, BC Canada
| | - Nino Trattnig
- grid.5173.00000 0001 2298 5320Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria ,grid.5477.10000000120346234Present Address: Department of Chemical Biology and Drug Discovery, Utrecht University, Utrecht, The Netherlands
| | - Yiqiu Yang
- grid.61971.380000 0004 1936 7494Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC Canada
| | - Matteo Cattin
- grid.5173.00000 0001 2298 5320Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Paul Kosma
- grid.5173.00000 0001 2298 5320Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Ralph Pantophlet
- grid.61971.380000 0004 1936 7494Faculty of Health Sciences, Simon Fraser University, Burnaby, BC Canada ,grid.61971.380000 0004 1936 7494Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC Canada
| |
Collapse
|
17
|
Franco AR, Peri F. Developing New Anti-Tuberculosis Vaccines: Focus on Adjuvants. Cells 2021; 10:cells10010078. [PMID: 33466444 PMCID: PMC7824815 DOI: 10.3390/cells10010078] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) that sits in the top 10 leading causes of death in the world today and is the current leading cause of death among infectious diseases. Although there is a licensed vaccine against TB, the Mycobacterium bovis bacilli Calmette–Guérin (BCG) vaccine, it has several limitations, namely its high variability of efficacy in the population and low protection against pulmonary tuberculosis. New vaccines for TB are needed. The World Health Organization (WHO) considers the development and implementation of new TB vaccines to be a priority. Subunit vaccines are promising candidates since they can overcome safety concerns and optimize antigen targeting. Nevertheless, these vaccines need adjuvants in their formulation in order to increase immunogenicity, decrease the needed antigen dose, ensure a targeted delivery and optimize the antigens delivery and interaction with the immune cells. This review aims to focus on adjuvants being used in new formulations of TB vaccines, namely candidates already in clinical trials and others in preclinical development. Although no correlates of protection are defined, most research lines in the field of TB vaccination focus on T-helper 1 (Th1) type of response, namely polyfunctional CD4+ cells expressing simultaneously IFN-γ, TNF-α, and IL-2 cytokines, and also Th17 responses. Accordingly, most of the adjuvants reviewed here are able to promote such responses. In the future, it might be advantageous to consider a wider array of immune parameters to better understand the role of adjuvants in TB immunity and establish correlates of protection.
Collapse
|
18
|
Clemmensen HS, Knudsen NPH, Billeskov R, Rosenkrands I, Jungersen G, Aagaard C, Andersen P, Mortensen R. Rescuing ESAT-6 Specific CD4 T Cells From Terminal Differentiation Is Critical for Long-Term Control of Murine Mtb Infection. Front Immunol 2020; 11:585359. [PMID: 33240275 PMCID: PMC7677256 DOI: 10.3389/fimmu.2020.585359] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 12/25/2022] Open
Abstract
In most cases, Mycobacterium tuberculosis (Mtb) causes life-long chronic infections, which poses unique challenges for the immune system. Most of the current tuberculosis (TB) subunit vaccines incorporate immunodominant antigens and at this point, it is poorly understood how the CD4 T cell subsets recognizing these antigens are affected during long-term infection. Very little is known about the requirements for sustainable vaccine protection against TB. To explore this, we screened 62 human-recognized Mtb antigens during chronic murine Mtb infection and identified the four most immunodominant antigens in this setting (MPT70, Rv3020c, and Rv3019c and ESAT-6). Combined into a subunit vaccine, this fusion protein induced robust protection both in a standard short-term model and in a long-term infection model where immunity from BCG waned. Importantly, replacement of ESAT-6 with another ESAT-6-family antigen, Rv1198, led to similar short-term protection but a complete loss of bacterial control during chronic infection. This observation was further underscored, as the ESAT-6 containing vaccine mediated sustainable protection in a model of post-exposure vaccination, where the ESAT-6-replacement vaccine did not. An individual comparison of the CD4 T cell responses during Mtb infection revealed that ESAT-6-specific T cells were more terminally differentiated than the other immunodominant antigens and immunization with the ESAT-6 containing vaccine led to substantially greater reduction in the overall T cell differentiation status. Our data therefore associates long-term bacterial control with the ability of a vaccine to rescue infection-driven CD4T cell differentiation and future TB antigen discovery programs should focus on identifying antigens with the highest accompanying T cell differentiation, like ESAT-6. This also highlights the importance of long-term readouts in both preclinical and clinical studies with TB vaccines.
Collapse
Affiliation(s)
- Helena Strand Clemmensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Claus Aagaard
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
19
|
Zawawi A, Else KJ. Soil-Transmitted Helminth Vaccines: Are We Getting Closer? Front Immunol 2020; 11:576748. [PMID: 33133094 PMCID: PMC7565266 DOI: 10.3389/fimmu.2020.576748] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023] Open
Abstract
Parasitic helminths infect over one-fourth of the human population resulting in significant morbidity, and in some cases, death in endemic countries. Despite mass drug administration (MDA) to school-aged children and other control measures, helminth infections are spreading into new areas. Thus, there is a strong rationale for developing anthelminthic vaccines as cost-effective, long-term immunological control strategies, which, unlike MDA, are not haunted by the threat of emerging drug-resistant helminths nor limited by reinfection risk. Advances in vaccinology, immunology, and immunomics include the development of new tools that improve the safety, immunogenicity, and efficacy of vaccines; and some of these tools have been used in the development of helminth vaccines. The development of anthelminthic vaccines is fraught with difficulty. Multiple lifecycle stages exist each presenting stage-specific antigens. Further, helminth parasites are notorious for their ability to dampen down and regulate host immunity. One of the first significant challenges in developing any vaccine is identifying suitable candidate protective antigens. This review explores our current knowledge in lead antigen identification and reports on recent pre-clinical and clinical trials in the context of the soil-transmitted helminths Trichuris, the hookworms and Ascaris. Ultimately, a multivalent anthelminthic vaccine could become an essential tool for achieving the medium-to long-term goal of controlling, or even eliminating helminth infections.
Collapse
Affiliation(s)
- Ayat Zawawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Ayat Zawawi
| | - Kathryn J. Else
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine, and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom,Kathryn J. Else
| |
Collapse
|
20
|
Ehsan M, Hu RS, Liang QL, Hou JL, Song X, Yan R, Zhu XQ, Li X. Advances in the Development of Anti- Haemonchus contortus Vaccines: Challenges, Opportunities, and Perspectives. Vaccines (Basel) 2020; 8:vaccines8030555. [PMID: 32971770 PMCID: PMC7565421 DOI: 10.3390/vaccines8030555] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal nematode parasite Haemonchus contortus (H. contortus) is a resident of tropical and subtropical regions worldwide that imposes significant production losses, economic losses, and animal health issues in the small ruminant industry, particularly sheep and goats. Considerable efforts have been made to understand how immunity is elicited against H. contortus infection. Various potential vaccine antigens have been tested by different methods and strategies applied in animal models, and significant progress has been made in the development of vaccines against H. contortus. This review highlighted and shared the knowledge about the current understanding of host immune responses to H. contortus and ongoing challenges in the development of a protective, effective, and long-lasting vaccine against H. contortus infection. We have also pinpointed some achievements and failures in the development and testing of vaccines, which will establish a road map for future research directions to explore new effective vaccine candidates for controlling and preventing H. contortus infection.
Collapse
Affiliation(s)
- Muhammad Ehsan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (M.E.); (R.-S.H.); (Q.-L.L.); (J.-L.H.)
| | - Rui-Si Hu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (M.E.); (R.-S.H.); (Q.-L.L.); (J.-L.H.)
| | - Qin-Li Liang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (M.E.); (R.-S.H.); (Q.-L.L.); (J.-L.H.)
| | - Jun-Ling Hou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (M.E.); (R.-S.H.); (Q.-L.L.); (J.-L.H.)
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.S.); (R.Y.); (X.L.)
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.S.); (R.Y.); (X.L.)
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (M.E.); (R.-S.H.); (Q.-L.L.); (J.-L.H.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China
- Correspondence: or ; Tel.: +86-354-628-8993
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.S.); (R.Y.); (X.L.)
| |
Collapse
|
21
|
Rudicell RS, Garinot M, Kanekiyo M, Kamp HD, Swanson K, Chou TH, Dai S, Bedel O, Simard D, Gillespie RA, Yang K, Reardon M, Avila LZ, Besev M, Dhal PK, Dharanipragada R, Zheng L, Duan X, Dinapoli J, Vogel TU, Kleanthous H, Mascola JR, Graham BS, Haensler J, Wei CJ, Nabel GJ. Comparison of adjuvants to optimize influenza neutralizing antibody responses. Vaccine 2019; 37:6208-6220. [PMID: 31493950 DOI: 10.1016/j.vaccine.2019.08.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/26/2019] [Accepted: 08/17/2019] [Indexed: 12/14/2022]
Abstract
Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.
Collapse
Affiliation(s)
| | | | - Masaru Kanekiyo
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Rebecca A Gillespie
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
22
|
Keshwara R, Hagen KR, Abreu-Mota T, Papaneri AB, Liu D, Wirblich C, Johnson RF, Schnell MJ. A Recombinant Rabies Virus Expressing the Marburg Virus Glycoprotein Is Dependent upon Antibody-Mediated Cellular Cytotoxicity for Protection against Marburg Virus Disease in a Murine Model. J Virol 2019; 93:e01865-18. [PMID: 30567978 PMCID: PMC6401435 DOI: 10.1128/jvi.01865-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022] Open
Abstract
Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.
Collapse
Affiliation(s)
- Rohan Keshwara
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Katie R Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Maryland, USA
| | - Tiago Abreu-Mota
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Life and Health Sciences Research Institute (ICVS) School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Amy B Papaneri
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Liu
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Maryland, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Tandon A, Pathak M, Harioudh MK, Ahmad S, Sayeed M, Afshan T, Siddiqi MI, Mitra K, Bhattacharya SM, Ghosh JK. A TLR4-derived non-cytotoxic, self-assembling peptide functions as a vaccine adjuvant in mice. J Biol Chem 2018; 293:19874-19885. [PMID: 30385503 DOI: 10.1074/jbc.ra118.002768] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/01/2018] [Indexed: 12/18/2022] Open
Abstract
Vaccination is devised/formulated to stimulate specific and prolonged immune responses for long-term protection against infection or disease. A vaccine component, namely adjuvant, enhances antigen recognition by the host immune system and thereby stimulates its cellular and adaptive responses. Especially synthetic Toll-like receptor (TLR) agonists having self-assembling properties are considered as good candidates for adjuvant development. Here, a human TLR4-derived 20-residue peptide (TR-433), present in the dimerization interface of the TLR4-myeloid differentiation protein-2 (MD2) complex, displayed self-assembly and adopted a nanostructure. Both in vitro studies and in vivo experiments in mice indicated that TR-433 is nontoxic. TR-433 induced pro-inflammatory responses in THP-1 monocytes and HEK293T cells that were transiently transfected with TLR4/CD14/MD2 and also in BALB/c mice. In light of the self-assembly and pro-inflammatory properties of TR-433, we immunized with a mixture of TR-433 and either ovalbumin or filarial antigen trehalose-6-phosphate phosphatase (TPP). A significant amount of IgG titers was produced, suggesting adjuvanting capability of TR-433 that was comparable with that of Freund's complete adjuvant (FCA) and appreciably higher than that of alum. We found that TR-433 preferentially activates type 1 helper T cell (Th1) response rather than type 2 helper T cell (Th2) response. To our knowledge, this is the first report on the identification of a short TLR4-derived peptide that possesses both self-assembling and pro-inflammatory properties and has significant efficacy as an adjuvant, capable of activating cellular responses in mice. These results indicate that TR-433 possesses significant potential for development as a new adjuvant in therapeutic application.
Collapse
Affiliation(s)
| | | | | | | | - Mohd Sayeed
- From the Molecular and Structural Biology Division
| | | | - M I Siddiqi
- From the Molecular and Structural Biology Division
| | - Kalyan Mitra
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road Lucknow-226 031, India
| | | | | |
Collapse
|
24
|
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
|
25
|
Zhang W, Molehin AJ, Rojo JU, Sudduth J, Ganapathy PK, Kim E, Siddiqui AJ, Freeborn J, Sennoune SR, May J, Lazarus S, Nguyen C, Redman WK, Ahmad G, Torben W, Karmakar S, Le L, Kottapalli KR, Kottapalli P, Wolf RF, Papin JF, Carey D, Gray SA, Bergthold JD, Damian RT, Mayer BT, Marks F, Reed SG, Carter D, Siddiqui AA. Sm-p80-based schistosomiasis vaccine: double-blind preclinical trial in baboons demonstrates comprehensive prophylactic and parasite transmission-blocking efficacy. Ann N Y Acad Sci 2018; 1425:38-51. [PMID: 30133707 PMCID: PMC6110104 DOI: 10.1111/nyas.13942] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023]
Abstract
Schistosomiasis is of public health importance to an estimated one billion people in 79 countries. A vaccine is urgently needed. Here, we report the results of four independent, double-blind studies of an Sm-p80-based vaccine in baboons. The vaccine exhibited potent prophylactic efficacy against transmission of Schistosoma mansoni infection and was associated with significantly less egg-induced pathology, compared with unvaccinated control animals. Specifically, the vaccine resulted in a 93.45% reduction of pathology-producing female worms and significantly resolved the major clinical manifestations of hepatic/intestinal schistosomiasis by reducing the tissue egg-load by 89.95%. A 35-fold decrease in fecal egg excretion in vaccinated animals, combined with an 81.51% reduction in hatching of eggs into the snail-infective stage (miracidia), demonstrates the parasite transmission-blocking potential of the vaccine. Substantially higher Sm-p80 expression in female worms and Sm-p80-specific antibodies in vaccinated baboons appear to play an important role in vaccine-mediated protection. Preliminary analyses of RNA sequencing revealed distinct molecular signatures of vaccine-induced effects in baboon immune effector cells. This study provides comprehensive evidence for the effectiveness of an Sm-p80-based vaccine for schistosomiasis.
Collapse
Affiliation(s)
- Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Adebayo J. Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Juan U. Rojo
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH
| | - Justin Sudduth
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Pramodh K. Ganapathy
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Eunjee Kim
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Arif J. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Jasmin Freeborn
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Souad R. Sennoune
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Jordan May
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Samra Lazarus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Catherine Nguyen
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Whitni K. Redman
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Gul Ahmad
- Department of Natural Sciences, Peru State College, Peru, NE
| | | | - Souvik Karmakar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Loc Le
- Biomedical Research Institute, Rockville, MD
| | | | | | - Roman F. Wolf
- Oklahoma City VA Health Care System, Oklahoma City, OK
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - David Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | | | - Raymond T. Damian
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Bryan T. Mayer
- Vaccine Immunology Statistical Center, Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Florian Marks
- International Vaccine Institute SNU Research Park, Seoul, South Korea
- The Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Darrick Carter
- PAI Life Sciences, Seattle, Washington, WA
- Infectious Disease Research Institute, Seattle, WA
| | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| |
Collapse
|
26
|
Seydoux E, Liang H, Dubois Cauwelaert N, Archer M, Rintala ND, Kramer R, Carter D, Fox CB, Orr MT. Effective Combination Adjuvants Engage Both TLR and Inflammasome Pathways To Promote Potent Adaptive Immune Responses. THE JOURNAL OF IMMUNOLOGY 2018; 201:98-112. [PMID: 29769270 DOI: 10.4049/jimmunol.1701604] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/24/2018] [Indexed: 11/19/2022]
Abstract
The involvement of innate receptors that recognize pathogen- and danger-associated molecular patterns is critical to programming an effective adaptive immune response to vaccination. The synthetic TLR4 agonist glucopyranosyl lipid adjuvant (GLA) synergizes with the squalene oil-in-water emulsion (SE) formulation to induce strong adaptive responses. Although TLR4 signaling through MyD88 and TIR domain-containing adapter inducing IFN-β are essential for GLA-SE activity, the mechanisms underlying the synergistic activity of GLA and SE are not fully understood. In this article, we demonstrate that the inflammasome activation and the subsequent release of IL-1β are central effectors of the action of GLA-SE, as infiltration of innate cells into the draining lymph nodes and production of IFN-γ are reduced in ASC-/- animals. Importantly, the early proliferation of Ag-specific CD4+ T cells was completely ablated after immunization in ASC-/- animals. Moreover, numbers of Ag-specific CD4+ T and B cells as well as production of IFN-γ, TNF-α, and IL-2 and Ab titers were considerably reduced in ASC-/-, NLRP3-/-, and IL-1R-/- mice compared with wild-type mice and were completely ablated in TLR4-/- animals. Also, extracellular ATP, a known trigger of the inflammasome, augments Ag-specific CD4+ T cell responses, as hydrolyzing it with apyrase diminished adaptive responses induced by GLA-SE. These data thus demonstrate that GLA-SE adjuvanticity acts through TLR4 signaling and NLRP3 inflammasome activation to promote robust Th1 and B cell responses to vaccine Ags. The findings suggest that engagement of both TLR and inflammasome activators may be a general paradigm for induction of robust CD4 T cell immunity with combination adjuvants such as GLA-SE.
Collapse
Affiliation(s)
- Emilie Seydoux
- Infectious Disease Research Institute, Seattle, WA 98102; and
| | - Hong Liang
- Infectious Disease Research Institute, Seattle, WA 98102; and
| | | | - Michelle Archer
- Infectious Disease Research Institute, Seattle, WA 98102; and
| | | | - Ryan Kramer
- Infectious Disease Research Institute, Seattle, WA 98102; and
| | - Darrick Carter
- Infectious Disease Research Institute, Seattle, WA 98102; and.,Department of Global Health, University of Washington, Seattle, WA 98195
| | - Christopher B Fox
- Infectious Disease Research Institute, Seattle, WA 98102; and.,Department of Global Health, University of Washington, Seattle, WA 98195
| | - Mark T Orr
- Infectious Disease Research Institute, Seattle, WA 98102; and .,Department of Global Health, University of Washington, Seattle, WA 98195
| |
Collapse
|
27
|
Muñoz-Wolf N, Lavelle EC. A Guide to IL-1 family cytokines in adjuvanticity. FEBS J 2018; 285:2377-2401. [PMID: 29656546 DOI: 10.1111/febs.14467] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/21/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022]
Abstract
Growing awareness of the multiplicity of roles for the IL-1 family in immune regulation has prompted research exploring these cytokines in the context of vaccine-induced immunity. While tightly regulated, cytokines of the IL-1 family are normally released in response to cellular stress and in combination with other danger-/damage-associated molecular patterns (DAMPs), triggering potent local and systemic immune responses. In the context of infection or autoimmunity, engagement of IL-1 family receptors links robust innate responses to adaptive immunity. Clinical and experimental evidence has revealed that many vaccine adjuvants induce the release of one or multiple IL-1 family cytokines. The coordinated release of IL-1 family members in response to adjuvant-induced damage or cell death may be a determining factor in the transition from local inflammation to the induction of an adaptive response. Here, we analyse the effects of IL-1 family cytokines on innate and adaptive immunity with a particular emphasis on activation of antigen-presenting cells and induction of T cell-mediated immunity, and we address in detail the contribution of these cytokines to the modes of action of vaccine adjuvants including those currently approved for human use.
Collapse
Affiliation(s)
- Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Ireland
| |
Collapse
|
28
|
O'Konek JJ, Landers JJ, Janczak KW, Goel RR, Mondrusov AM, Wong PT, Baker JR. Nanoemulsion adjuvant-driven redirection of T H2 immunity inhibits allergic reactions in murine models of peanut allergy. J Allergy Clin Immunol 2018; 141:2121-2131. [PMID: 29655584 DOI: 10.1016/j.jaci.2018.01.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 01/17/2018] [Accepted: 01/27/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immunotherapy for food allergies involves progressive increased exposures to food that result in desensitization to food allergens in some subjects but not tolerance to the food. Therefore new approaches to suppress allergic immunity to food are necessary. Previously, we demonstrated that intranasal immunization with a nanoemulsion (NE) adjuvant induces robust mucosal antibody and TH17-polarized immunity, as well as systemic TH1-biased cellular immunity with suppression of pre-existing TH2-biased immunity. OBJECTIVE We hypothesized that immunization with food in conjunction with the nanoemulsion adjuvant could lead to modulation of allergic reactions in food allergy by altering pre-existing allergic immunity and enhancing mucosal immunity. METHODS Mice were sensitized to peanut with aluminum hydroxide or cholera toxin. The animals were then administered 3 monthly intranasal immunizations with peanut in the nanoemulsion adjuvant or saline. Mice were then challenged with peanut to examine allergen reactivity. RESULTS The NE intranasal immunizations resulted in marked decreases in TH2 cytokine, IgG1, and IgE levels, whereas TH1 and mucosal TH17 immune responses were increased. After allergen challenge, these mice showed significant reductions in allergic hypersensitivity. Additionally, the NE immunizations significantly increased antigen-specific IL-10 production and regulatory T-cell counts, and the protection induced by NE was dependent in part on IL-10. Control animals immunized with intranasal peanut in saline had no modulation of their allergic response. CONCLUSIONS NE adjuvant-mediated induction of mucosal TH17 and systemic TH1-biased immunity can suppress TH2-mediated allergy through multiple mechanisms and protect against anaphylaxis. These results suggest the potential therapeutic utility of this approach in the setting of food allergy.
Collapse
Affiliation(s)
- Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| | - Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | | | - Rishi R Goel
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Anna M Mondrusov
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Pamela T Wong
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
29
|
Stutzer C, Richards SA, Ferreira M, Baron S, Maritz-Olivier C. Metazoan Parasite Vaccines: Present Status and Future Prospects. Front Cell Infect Microbiol 2018; 8:67. [PMID: 29594064 PMCID: PMC5859119 DOI: 10.3389/fcimb.2018.00067] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Eukaryotic parasites and pathogens continue to cause some of the most detrimental and difficult to treat diseases (or disease states) in both humans and animals, while also continuously expanding into non-endemic countries. Combined with the ever growing number of reports on drug-resistance and the lack of effective treatment programs for many metazoan diseases, the impact that these organisms will have on quality of life remain a global challenge. Vaccination as an effective prophylactic treatment has been demonstrated for well over 200 years for bacterial and viral diseases. From the earliest variolation procedures to the cutting edge technologies employed today, many protective preparations have been successfully developed for use in both medical and veterinary applications. In spite of the successes of these applications in the discovery of subunit vaccines against prokaryotic pathogens, not many targets have been successfully developed into vaccines directed against metazoan parasites. With the current increase in -omics technologies and metadata for eukaryotic parasites, target discovery for vaccine development can be expedited. However, a good understanding of the host/vector/pathogen interface is needed to understand the underlying biological, biochemical and immunological components that will confer a protective response in the host animal. Therefore, systems biology is rapidly coming of age in the pursuit of effective parasite vaccines. Despite the difficulties, a number of approaches have been developed and applied to parasitic helminths and arthropods. This review will focus on key aspects of vaccine development that require attention in the battle against these metazoan parasites, as well as successes in the field of vaccine development for helminthiases and ectoparasites. Lastly, we propose future direction of applying successes in pursuit of next generation vaccines.
Collapse
Affiliation(s)
- Christian Stutzer
- Tick Vaccine Group, Department of Genetics, University of Pretoria, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
30
|
Gao Y, Wijewardhana C, Mann JFS. Virus-Like Particle, Liposome, and Polymeric Particle-Based Vaccines against HIV-1. Front Immunol 2018. [PMID: 29541072 PMCID: PMC5835502 DOI: 10.3389/fimmu.2018.00345] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is acknowledged that vaccines remain the best hope for eliminating the HIV-1 epidemic. However, the failure to produce effective vaccine immunogens and the inability of conventional delivery strategies to elicit the desired immune responses remains a central theme and has ultimately led to a significant roadblock in HIV vaccine development. Consequently, significant efforts have been applied to generate novel vaccine antigens and delivery agents, which mimic viral structures for optimal immune induction. Here, we review the latest developments that have occurred in the nanoparticle vaccine field, with special emphasis on strategies that are being utilized to attain highly immunogenic, systemic, and mucosal anti-HIV humoral and cellular immune responses. This includes the design of novel immunogens, the central role of antigen-presenting cells, delivery routes, and biodistribution of nanoparticles to lymph nodes. In particular, we will focus on virus-like-particle formulations and their preclinical uses within the HIV prophylactic vaccine setting.
Collapse
Affiliation(s)
- Yong Gao
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Chanuka Wijewardhana
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Jamie F S Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
31
|
Anderson J, Olafsdottir TA, Kratochvil S, McKay PF, Östensson M, Persson J, Shattock RJ, Harandi AM. Molecular Signatures of a TLR4 Agonist-Adjuvanted HIV-1 Vaccine Candidate in Humans. Front Immunol 2018. [PMID: 29535712 PMCID: PMC5834766 DOI: 10.3389/fimmu.2018.00301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Systems biology approaches have recently provided new insights into the mechanisms of action of human vaccines and adjuvants. Here, we investigated early transcriptional signatures induced in whole blood of healthy subjects following vaccination with a recombinant HIV-1 envelope glycoprotein subunit CN54gp140 adjuvanted with the TLR4 agonist glucopyranosyl lipid adjuvant-aqueous formulation (GLA-AF) and correlated signatures to CN54gp140-specific serum antibody responses. Fourteen healthy volunteers aged 18–45 years were immunized intramuscularly three times at 1-month intervals and whole blood samples were collected at baseline, 6 h, and 1, 3, and 7 days post first immunization. Subtle changes in the transcriptomic profiles were observed following immunization, ranging from over 300 differentially expressed genes (DEGs) at day 1 to nearly 100 DEGs at day 7 following immunization. Functional pathway analysis revealed blood transcription modules (BTMs) related to general cell cycle activation, and innate immune cell activation at early time points, as well as BTMs related to T cells and B cell activation at the later time points post-immunization. Diverse CN54gp140-specific serum antibody responses of the subjects enabled their categorization into high or low responders, at early (<1 month) and late (up to 6 months) time points post vaccination. BTM analyses revealed repression of modules enriched in NK cells, and the mitochondrial electron chain, in individuals with high or sustained antigen-specific antibody responses. However, low responders showed an enhancement of BTMs associated with enrichment in myeloid cells and monocytes as well as integrin cell surface interactions. Flow cytometry analysis of peripheral blood mononuclear cells obtained from the subjects revealed an enhanced frequency of CD56dim NK cells in the majority of vaccines 14 days after vaccination as compared with the baseline. These results emphasize the utility of a systems biology approach to enhance our understanding on the mechanisms of action of TLR4 adjuvanted human vaccines.
Collapse
Affiliation(s)
- Jenna Anderson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thorunn A Olafsdottir
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sven Kratochvil
- Department of Medicine, Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, London, United Kingdom
| | - Paul F McKay
- Department of Medicine, Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, London, United Kingdom
| | - Malin Östensson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Robin J Shattock
- Department of Medicine, Mucosal Infection and Immunity Group, Section of Virology, Imperial College London, London, United Kingdom
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
32
|
Tang J, Sun M, Shi G, Xu Y, Han Y, Li X, Dong W, Zhan L, Qin C. Toll-Like Receptor 8 Agonist Strengthens the Protective Efficacy of ESAT-6 Immunization to Mycobacterium tuberculosis Infection. Front Immunol 2018; 8:1972. [PMID: 29416532 PMCID: PMC5787779 DOI: 10.3389/fimmu.2017.01972] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/20/2017] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence suggests important functions for human Toll-like receptor 8 in vivo in tuberculosis and autoimmune diseases. However, these studies are limited by the lack of specific agonists and by the fact that the homology of TLR8 in human and mice is not sufficient to rely on mouse models. In this study, we examined the role of human TLR8 in the disease progression of experimental Mycobacterium tuberculosis (Mtb) infection, as well as the benefits provided by a TLR8 agonist against Mtb challenge in a human TLR8 transgenic mouse. We found that the expression of human TLR8 in C57BL/6 mice permits higher bacilli load in tissues. A vaccine formulated with ESAT-6, aluminum hydroxide, and TLR8 agonist provided protection against Mtb challenge, with a high percentage of CD44hiCD62Lhi TCM. Using ovalbumin as a model antigen, we demonstrated that the activation of TLR8 enhanced the innate and adaptive immune response, and provided a sustained TCM formation and Th1 type humoral response, which were mainly mediated by type I IFN signaling. Further research is required to optimize the vaccine formulation and seek optimal combinations of different TLR agonists, such as TLR4, for better adjuvanticity in this animal model.
Collapse
Affiliation(s)
- Jun Tang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing, China
| | - Mengmeng Sun
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Guiying Shi
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Yanfeng Xu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Yunlin Han
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Xiang Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Wei Dong
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China
| | - Lingjun Zhan
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing, China
| | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China.,Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, China.,Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing, China
| |
Collapse
|
33
|
Speth MT, Repnik U, Müller E, Spanier J, Kalinke U, Corthay A, Griffiths G. Poly(I:C)-Encapsulating Nanoparticles Enhance Innate Immune Responses to the Tuberculosis Vaccine Bacille Calmette-Guérin (BCG) via Synergistic Activation of Innate Immune Receptors. Mol Pharm 2017; 14:4098-4112. [PMID: 28974092 DOI: 10.1021/acs.molpharmaceut.7b00795] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The attenuated live vaccine strain bacille Calmette-Guérin (BCG) is currently the only available vaccine against tuberculosis (TB), but is largely ineffective against adult pulmonary TB, the most common disease form. This is in part due to BCG's ability to interfere with the host innate immune response, a feature that might be targeted to enhance the potency of this vaccine. Here, we investigated the ability of chitosan-based nanoparticles (pIC-NPs) containing polyinosinic-polycytidylic acid (poly(I:C)), an inducer of innate immunity via Toll-like receptor 3 (TLR3), to enhance the immunogenicity of BCG in mouse bone marrow derived macrophages (BMDM) in vitro. Incorporation of poly(I:C) into NPs protected it against degradation by ribonucleases and increased its uptake by mouse BMDM. Whereas soluble poly(I:C) was ineffective, pIC-NPs strongly enhanced the proinflammatory immune response of BCG-infected macrophages in a synergistic fashion, as evident by increased production of cytokines and induction of nitric oxide synthesis. Using macrophages from mice deficient in key signaling molecules involved in the pathogen recognition response, we identified combined activation of MyD88- and TRIF-dependent TLR signaling pathways to be essential for the synergistic effect between BCG and NP. Moreover, synergy was strongly dependent on the order of the two stimuli, with TLR activation by BCG functioning as the priming event for the subsequent pIC-NP stimulus, which acted through an auto-/paracrine type I interferon (IFN) feedback loop. Our results provide a foundation for a promising new approach to enhance BCG-vaccine immunogenicity by costimulation with NPs. They also contribute to a molecular understanding of the observed synergistic interaction between the pIC-NPs and BCG vaccine.
Collapse
Affiliation(s)
- Martin T Speth
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| | - Elisabeth Müller
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway.,Tumor Immunology lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo , N-0424 Oslo, Norway
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School , D-30625 Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Center for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School , D-30625 Hannover, Germany
| | - Alexandre Corthay
- Tumor Immunology lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo , N-0424 Oslo, Norway
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo , N-0371 Oslo, Norway
| |
Collapse
|
34
|
Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early IFNγ response promoting vaccine immunogenicity. NPJ Vaccines 2017; 2:25. [PMID: 29263880 PMCID: PMC5627273 DOI: 10.1038/s41541-017-0027-3] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 12/24/2022] Open
Abstract
Combining immunostimulants in adjuvants can improve the quality of the immune response to vaccines. Here, we report a unique mechanism of molecular and cellular synergy between a TLR4 ligand, 3-O-desacyl-4’-monophosphoryl lipid A (MPL), and a saponin, QS-21, the constituents of the Adjuvant System AS01. AS01 is part of the malaria and herpes zoster vaccine candidates that have demonstrated efficacy in phase III studies. Hours after injection of AS01-adjuvanted vaccine, resident cells, such as NK cells and CD8+ T cells, release IFNγ in the lymph node draining the injection site. This effect results from MPL and QS-21 synergy and is controlled by macrophages, IL-12 and IL-18. Depletion strategies showed that this early IFNγ production was essential for the activation of dendritic cells and the development of Th1 immunity by AS01-adjuvanted vaccine. A similar activation was observed in the lymph node of AS01-injected macaques as well as in the blood of individuals receiving the malaria RTS,S vaccine. This mechanism, previously described for infections, illustrates how adjuvants trigger naturally occurring pathways to improve the efficacy of vaccines. A mechanism is revealed by which vaccine components co-operate to stimulate the immune system and improve vaccine efficacy. Some vaccines are formulated with adjuvants—compounds that induce a greater immune response to the vaccine and help to elicit greater protection against future infections. Arnaud Didierlaurent and his team of researchers at GSK Vaccines, Belgium, demonstrate that the two immunostimulants in the adjuvant AS01, used in several recently developed vaccines, works in tandem to trigger the activation of important immune system moderators. The synergistic effect of the immunostimulants modulate specific immune cells at the site of the vaccination to better prepare the body against future infection. Studies such as this allow us to better understand how vaccines work and lay the foundation for more informed research into future vaccine development.
Collapse
|
35
|
Abstract
Adjuvants have been deliberately added to vaccines since the 1920's when alum was discovered to boost antibody responses, leading to better protection. The first adjuvants were discovered by accident and were used in the safer but less immunogenic subunit vaccines, supposedly by providing an antigen depot to extend antigen presentation. Since that time, much has been discovered about how these adjuvants impact cells at the tissue site to activate innate immune responses, mobilize dendritic cells and drive adaptive immunity. New approaches to vaccine construction for infectious diseases that have so far not been well addressed by conventional vaccines often attempt to induce antibodies, polyfunctional CD4+ T cells and CD8+ CTLs. The discovery of pattern recognition receptors and ligands that drive desired T cell responses has led to development of novel adjuvant strategies using immunomodulatory agents to direct appropriate immune responses.
Collapse
Affiliation(s)
- Amy S McKee
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Philippa Marrack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, 1400, Jackson St., Denver, CO 80206, USA
| |
Collapse
|
36
|
O'Hagan DT, Friedland LR, Hanon E, Didierlaurent AM. Towards an evidence based approach for the development of adjuvanted vaccines. Curr Opin Immunol 2017; 47:93-102. [PMID: 28755542 DOI: 10.1016/j.coi.2017.07.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/12/2017] [Indexed: 01/29/2023]
Abstract
In the last two decades, several vaccines formulated with a new generation of adjuvants have been licensed or approved to target diseases such as influenza, hepatitis B, cervical cancer, and malaria. These new generation adjuvants appear to work by delivering a localized activation signal to the innate immune system, which in turn promotes antigen-specific adaptive immunity. Advances in understanding of the innate immune system together with high-throughput discovery of synthetic immune potentiators are now expanding the portfolio of new generation adjuvants available for evaluation. Meanwhile, omics and systems biology are providing molecular benchmarks or signatures to assess vaccine safety and effectiveness. This accumulating knowledge and experience raises the prospect that the future selection of the right antigen/adjuvant combination can be more evidence based and can speed up the clinical development program for new adjuvanted vaccines.
Collapse
Affiliation(s)
- Derek T O'Hagan
- GSK Vaccines, 14200 Shady Grove Road, Rockville, MD, USA. derek.t.o'
| | | | - Emmanuel Hanon
- GSK Vaccines, Rue de l'Institut 89, 1330 Rixensart, Belgium
| | | |
Collapse
|
37
|
Pérez-Toledo M, Valero-Pacheco N, Pastelin-Palacios R, Gil-Cruz C, Perez-Shibayama C, Moreno-Eutimio MA, Becker I, Pérez-Tapia SM, Arriaga-Pizano L, Cunningham AF, Isibasi A, Bonifaz LC, López-Macías C. Salmonella Typhi Porins OmpC and OmpF Are Potent Adjuvants for T-Dependent and T-Independent Antigens. Front Immunol 2017; 8:230. [PMID: 28337196 PMCID: PMC5344031 DOI: 10.3389/fimmu.2017.00230] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/17/2017] [Indexed: 02/05/2023] Open
Abstract
Several microbial components, such as bacterial DNA and flagellin, have been used as experimental vaccine adjuvants because of their inherent capacity to efficiently activate innate immune responses. Likewise, our previous work has shown that the major Salmonella Typhi (S. Typhi) outer membrane proteins OmpC and OmpF (porins) are highly immunogenic protective antigens that efficiently stimulate innate and adaptive immune responses in the absence of exogenous adjuvants. Moreover, S. Typhi porins induce the expression of costimulatory molecules on antigen-presenting cells through toll-like receptor canonical signaling pathways. However, the potential of major S. Typhi porins to be used as vaccine adjuvants remains unknown. Here, we evaluated the adjuvant properties of S. Typhi porins against a range of experimental and clinically relevant antigens. Co-immunization of S. Typhi porins with ovalbumin (OVA), an otherwise poorly immunogenic antigen, enhanced anti-OVA IgG titers, antibody class switching, and affinity maturation. This adjuvant effect was dependent on CD4+ T-cell cooperation and was associated with an increase in IFN-γ, IL-17A, and IL-2 production by OVA-specific CD4+ T cells. Furthermore, co-immunization of S. Typhi porins with an inactivated H1N1 2009 pandemic influenza virus experimental vaccine elicited higher hemagglutinating anti-influenza IgG titers, antibody class switching, and affinity maturation. Unexpectedly, co-administration of S. Typhi porins with purified, unconjugated Vi capsular polysaccharide vaccine (Vi CPS)—a T-independent antigen—induced higher IgG antibody titers and class switching. Together, our results suggest that S. Typhi porins OmpC and OmpF are versatile vaccine adjuvants, which could be used to enhance T-cell immune responses toward a Th1/Th17 profile, while improving antibody responses to otherwise poorly immunogenic T-dependent and T-independent antigens.
Collapse
Affiliation(s)
- Marisol Pérez-Toledo
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Nuriban Valero-Pacheco
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen , St. Gallen , Switzerland
| | | | - Mario A Moreno-Eutimio
- Immunity and Inflammation Research Unit, Hospital Juárez de México, Ministry of Health , Mexico City , Mexico
| | - Ingeborg Becker
- Facultad de Medicina, Departamento de Medicina Experimental, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Sonia Mayra Pérez-Tapia
- Unit of R&D in Bioprocesses (UDIBI), Department of Immunology, National School of Biological Sciences, National Polytechnic Institute , Mexico City , Mexico
| | - Lourdes Arriaga-Pizano
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Armando Isibasi
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Laura C Bonifaz
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Constantino López-Macías
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Rao M, Cadieux N, Fitzpatrick M, Reed S, Arsenian S, Valentini D, Parida S, Dodoo E, Zumla A, Maeurer M. Mycobacterium tuberculosis proteins involved in cell wall lipid biosynthesis improve BCG vaccine efficacy in a murine TB model. Int J Infect Dis 2017; 56:274-282. [PMID: 28161464 DOI: 10.1016/j.ijid.2017.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVES Advances in tuberculosis (TB) vaccine development are urgently required to enhance global disease management. We evaluated the potential of Mycobacterium tuberculosis (M. tb)-derived protein antigens Rv0447c, Rv2957 and Rv2958c to boost BCG vaccine efficacy in the presence or absence of glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) adjuvant. METHODS Mice received the BCG vaccine, followed by Rv0447c, Rv2957 and Rv2958c protein boosting with or without GLA-SE adjuvant 3 and 6 weeks later. Immune responses were examined at given time points. 9 weeks post vaccination, mice were aerosol-challenged with M. tb, and sacrificed at 6 and 12 weeks to assess bacterial burden. RESULTS Vaccination of mice with BCG and M. tb proteins in the presence of GLA-SE adjuvant triggered strong IFN-γ and IL-2 production by splenocytes; more TNF-α was produced without GLA-SE addition. Antibody responses to all three antigens did not differ, with or without GLA-SE adjuvant. Protein boosting without GLA-SE adjuvant resulted in vaccinated animals having better control of pulmonary M. tb load at 6 and 12 weeks post aerosol infection, while animals receiving the protein boost with GLA-SE adjuvant exhibited more bacteria in the lungs. CONCLUSIONS Our data provides evidence for developing Rv2958c, Rv2957 and Rv0447c in a heterologous prime-boost vaccination strategy with BCG.
Collapse
Affiliation(s)
- Martin Rao
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | | | | | - Steven Reed
- Infectious Disease Research Institute (IDRI), Seattle, USA
| | - Sergei Arsenian
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Davide Valentini
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Shreemanta Parida
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Ernest Dodoo
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London and the NIHR Biomedical Research centre at UCL Hospitals NHS Foundation Trust London, UK
| | - Markus Maeurer
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
39
|
Desbien AL, Dubois Cauwelaert N, Reed SJ, Bailor HR, Liang H, Carter D, Duthie MS, Fox CB, Reed SG, Orr MT. IL-18 and Subcapsular Lymph Node Macrophages are Essential for Enhanced B Cell Responses with TLR4 Agonist Adjuvants. THE JOURNAL OF IMMUNOLOGY 2016; 197:4351-4359. [PMID: 27794001 DOI: 10.4049/jimmunol.1600993] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/24/2016] [Indexed: 11/19/2022]
Abstract
Designing modern vaccine adjuvants depends on understanding the cellular and molecular events that connect innate and adaptive immune responses. The synthetic TLR4 agonist glycopyranosyl lipid adjuvant (GLA) formulated in a squalene-in-water emulsion (GLA-SE) augments both cellular and humoral immune responses to vaccine Ags. This adjuvant is currently included in several vaccines undergoing clinical evaluation including those for tuberculosis, leishmaniasis, and influenza. Delineation of the mechanisms of adjuvant activity will enable more informative evaluation of clinical trials. Early after injection, GLA-SE induces substantially more Ag-specific B cells, higher serum Ab titers, and greater numbers of T follicular helper (TFH) and Th1 cells than alum, the SE alone, or GLA without SE. GLA-SE augments Ag-specific B cell differentiation into germinal center and memory precursor B cells as well as preplasmablasts that rapidly secrete Abs. CD169+ SIGNR1+ subcapsular medullary macrophages are the primary cells to take up GLA-SE after immunization and are critical for the innate immune responses, including rapid IL-18 production, induced by GLA-SE. Depletion of subcapsular macrophages (SCMф) or abrogation of IL-18 signaling dramatically impairs the Ag-specific B cell and Ab responses augmented by GLA-SE. Depletion of SCMф also drastically reduces the Th1 but not the TFH response. Thus the GLA-SE adjuvant operates through interaction with IL-18-producing SCMф for the rapid induction of B cell expansion and differentiation, Ab secretion, and Th1 responses, whereas augmentation of TFH numbers by GLA-SE is independent of SCMф.
Collapse
Affiliation(s)
| | | | - Steven J Reed
- Infectious Disease Research Institute, Seattle, WA 98102
| | | | - Hong Liang
- Infectious Disease Research Institute, Seattle, WA 98102
| | - Darrick Carter
- Infectious Disease Research Institute, Seattle, WA 98102.,Department of Global Health, University of Washington, Seattle, WA 98195; and.,PAI Life Sciences, Seattle, WA 98102
| | - Malcolm S Duthie
- Infectious Disease Research Institute, Seattle, WA 98102.,Department of Global Health, University of Washington, Seattle, WA 98195; and
| | - Christopher B Fox
- Infectious Disease Research Institute, Seattle, WA 98102.,Department of Global Health, University of Washington, Seattle, WA 98195; and
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102.,Department of Global Health, University of Washington, Seattle, WA 98195; and
| | - Mark T Orr
- Infectious Disease Research Institute, Seattle, WA 98102; .,Department of Global Health, University of Washington, Seattle, WA 98195; and
| |
Collapse
|
40
|
Sabur A, Asad M, Ali N. Lipid based delivery and immuno-stimulatory systems: Master tools to combat leishmaniasis. Cell Immunol 2016; 309:55-60. [PMID: 27470274 DOI: 10.1016/j.cellimm.2016.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/06/2016] [Accepted: 07/20/2016] [Indexed: 11/25/2022]
Abstract
Disease management of leishmaniasis is appalling due to lack of a human vaccine and the toxicity and resistance concerns with limited therapeutic drugs. The challenges in development of a safe vaccine for generation and maintenance of robust antileishmanial protective immunity through a human administrable route of immunization can be addressed through immunomodulation and targeted delivery. The versatility of lipid based particulate system for deliberate delivery of diverse range of molecules including immunomodulators, antigens and drugs have essentially found pivotal role in design of proficient vaccination and therapeutic strategies against leishmaniasis. The prospects of lipid based preventive and curative formulations for leishmaniasis have been highlighted in this review.
Collapse
Affiliation(s)
- Abdus Sabur
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India.
| | - Mohammad Asad
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India.
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India.
| |
Collapse
|