1
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Emerging Cationic Nanovaccines. Pharmaceutics 2024; 16:1362. [PMID: 39598488 PMCID: PMC11597065 DOI: 10.3390/pharmaceutics16111362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Cationic vaccines of nanometric sizes can directly perform the delivery of antigen(s) and immunomodulator(s) to dendritic cells in the lymph nodes. The positively charged nanovaccines are taken up by antigen-presenting cells (APCs) of the lymphatic system often originating the cellular immunological defense required to fight intracellular microbial infections and the proliferation of cancers. Cationic molecules imparting the positive charges to nanovaccines exhibit a dose-dependent toxicity which needs to be systematically addressed. Against the coronavirus, mRNA cationic nanovaccines evolved rapidly. Nowadays cationic nanovaccines have been formulated against several infections with the advantage of cationic compounds granting protection of nucleic acids in vivo against biodegradation by nucleases. Up to the threshold concentration of cationic molecules for nanovaccine delivery, cationic nanovaccines perform well eliciting the desired Th 1 improved immune response in the absence of cytotoxicity. A second strategy in the literature involves dilution of cationic components in biocompatible polymeric matrixes. Polymeric nanoparticles incorporating cationic molecules at reduced concentrations for the cationic component often result in an absence of toxic effects. The progress in vaccinology against cancer involves in situ designs for cationic nanovaccines. The lysis of transformed cancer cells releases several tumoral antigens, which in the presence of cationic nanoadjuvants can be systemically presented for the prevention of metastatic cancer. In addition, these local cationic nanovaccines allow immunotherapeutic tumor treatment.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Yunys Pérez-Betancourt
- Department of Microbiology, University of Chicago, Cummings Life Science Center 920 E 58th St., Chicago, IL 60637, USA;
| |
Collapse
|
2
|
Duong VT, Skwarczynski M, Toth I. Towards the development of subunit vaccines against tuberculosis: The key role of adjuvant. Tuberculosis (Edinb) 2023; 139:102307. [PMID: 36706503 DOI: 10.1016/j.tube.2023.102307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
According to the World Health Organization (WHO), tuberculosis (TB) is the leading cause of death triggered by a single infectious agent, worldwide. Bacillus Calmette-Guerin (BCG) is the only currently licensed anti-TB vaccine. However, other strategies, including modification of recombinant BCG vaccine, attenuated Mycobacterium tuberculosis (Mtb) mutant constructs, DNA and protein subunit vaccines, are under extensive investigation. As whole pathogen vaccines can trigger serious adverse reactions, most current strategies are focused on the development of safe anti-TB subunit vaccines; this is especially important given the rising TB infection rate in immunocompromised HIV patients. The whole Mtb genome has been mapped and major antigens have been identified; however, optimal vaccine delivery mode is still to be established. Isolated protein antigens are typically poorly immunogenic so adjuvants are required to induce strong and long-lasting immune responses. This article aims to review the developmental status of anti-TB subunit vaccine adjuvants.
Collapse
Affiliation(s)
- Viet Tram Duong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
3
|
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
|
4
|
Effects of Antimicrobial Peptides Gal-13 on the Growth Performance, Intestinal Microbiota, Digestive Enzyme Activities, Intestinal Morphology, Antioxidative Activities, and Immunity of Broilers. Probiotics Antimicrob Proteins 2022; 15:694-705. [PMID: 35015242 DOI: 10.1007/s12602-021-09905-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 01/06/2023]
Abstract
To evaluate the application effect of antimicrobial peptides Gal-13 (AMP Gal-13) instead of antibiotic feed additives, 90 7-day-old Ross 308 broilers were randomly divided into 3 groups. Group A was fed a basic diet as the control, and Groups B and C were supplemented with AMP Gal-13 (100 mg/kg and 200 mg/kg, respectively). After a 35-day feeding experiment, the weight and average daily gain (ADG) of the broilers in Group B were significantly higher than those of the broilers in Group A. The Enterococcus sp. and Escherichia coli counts in the ileum and cecum in Group A were significantly higher than those in Groups B and C, while the Lactic acid bacteria (LAB) and Bifidobacterium sp. counts were significantly lower. The amylase activity of the jejunum in Group B was significantly higher than that in Group A. The villus length (VL): crypt depth (CD) ratios of the jejunum and ileum in Group B were significantly higher than those in Group A. The glutathione peroxidase (GSH-Px) activities in the liver and serum in Groups B and C were significantly higher than those in Group A, while the malondialdehyde (MDA) activity was significantly lower. The titers of Newcastle disease virus (NDV)-specific antibodies were elevated significantly in Group B at the age of 42 days. Additionally, the weights of the spleen and thymus were significantly increased. The expression levels of Il-2, Il-6, Tgf-β4, Tnf-α, and Mif in the spleen in Groups B and C were significantly downregulated to different degrees; Il-4 expression in Group B was significantly upregulated, while Ifn-γ expression in Group C was significantly upregulated. The results suggested that adding AMP Gal-13 to the diet could improve intestinal digestion, the antioxidant capacity, and immune function, ultimately promoting the growth of broilers.
Collapse
|
5
|
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a leading cause of mortality and morbidity due to a single infectious agent. Aerosol infection with Mtb can result in a range of responses from elimination, active, incipient, subclinical, and latent Mtb infections (LTBI), depending on the host's immune response and the dose and nature of infecting bacilli. Currently, BCG is the only vaccine approved to prevent TB. Although BCG confers protection against severe forms of childhood TB, its use in adults and those with comorbid conditions, such as HIV infection, is questionable. Novel vaccines, including recombinant BCG (rBCG), were developed to improve BCG's efficacy and use as an alternative to BCG in a vulnerable population. The first-generation rBCG vaccines had different Mtb antigens and were tested as a prime, prime-boost, or immunotherapeutic intervention. The novel vaccines target one or more of the following requirements, namely prevention of infection (POI), prevention of disease (POD), prevention of recurrence (POR), and therapeutic vaccines to treat a TB disease. Several vaccine candidates currently in development are classified into four primary categories: live attenuated whole-cell vaccine, inactivated whole-cell vaccine, adjuvanted protein subunit vaccine, and viral-vectored vaccine. Each vaccine's immunogenicity, safety, and efficacy are tested in preclinical animal models and further validated through various phases of clinical trials. This chapter summarizes the various TB vaccine candidates under different clinical trial stages and promises better protection against TB.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Department of Bacteriology, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamilnadu, India
| | - Selvakumar Subbian
- The Public Health Research Institute Center at New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
6
|
Abstract
Guinea pigs have been used as a model for Mycobacterium tuberculosis infection for many years and have been more recently adopted as a model for testing new tuberculosis (TB) vaccines. From the time of Robert Koch, who used guinea pigs to test theories about the newly discovered pathogen, the guinea pig has modeled active human infections, as it is susceptible to infection with low numbers of organisms. This article describes the modern use of the guinea pig to examine the pathology of the disease and the protocols used to examine specific outcomes associated with aerosol infection with virulent M. tuberculosis. The guinea pig is used extensively to investigate the ability of new TB vaccines to reduce TB disease, and two models have been employed. The first is the long-term disease model, in which vaccinated guinea pigs are monitored for disease after infection, and the second is the short-term assessment of mycobacterial burden model, which can determine the ability of a vaccine to reduce organism burden. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Preparation of virulent Mycobacterium tuberculosis seed stocks for animal infections Basic Protocol 2: Preparation of virulent Mycobacterium tuberculosis working stocks for animal infections Basic Protocol 3: Preparation of M. tuberculosis for aerosol infection of guinea pigs Basic Protocol 4: Injection of guinea pigs Basic Protocol 5: Blood collection from live guinea pigs Basic Protocol 6: Guinea pig euthanasia.
Collapse
Affiliation(s)
| | - Linda Izzo
- Colorado State University, Fort Collins, Colorado
| | | | - Angelo A Izzo
- Colorado State University, Fort Collins, Colorado
- Centenary Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Cho T, Khatchadourian C, Nguyen H, Dara Y, Jung S, Venketaraman V. A review of the BCG vaccine and other approaches toward tuberculosis eradication. Hum Vaccin Immunother 2021; 17:2454-2470. [PMID: 33769193 PMCID: PMC8475575 DOI: 10.1080/21645515.2021.1885280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 02/02/2023] Open
Abstract
Despite aggressive eradication efforts, Tuberculosis (TB) remains a global health burden, one that disproportionally affects poorer, less developed nations. The only vaccine approved for TB, the Bacillus of Calmette and Guérin (BCG) vaccine remains controversial because it's stated efficacy has been cited as anywhere from 0 to 80%. Nevertheless, there have been exciting discoveries about the mechanism of action of the BCG vaccine that suggests it has a role in immunization schedules today. We review recent data suggesting the vaccine imparts protection against both tuberculosis and non-tuberculosis pathogens via a newly discovered immune system called trained immunity. BCG's efficacy also appears to be tied to its affect on granulocytes at the epigenetic and hematopoietic stem cell levels, which we discuss in this article at length. We also write about how the different strains of the BCG vaccine elicit different immune responses, suggesting that certain BCG strains are more immunogenic than others. Finally, our review delves into how the current vaccine is being reformulated to be more efficacious, and track the development of the next generation vaccines against TB.
Collapse
Affiliation(s)
- Thomas Cho
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | | | - Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Yash Dara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Shuna Jung
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
8
|
Troy A, Esparza-Gonzalez SC, Bartek A, Creissen E, Izzo L, Izzo AA. Pulmonary mucosal immunity mediated through CpG provides adequate protection against pulmonary Mycobacterium tuberculosis infection in the mouse model. A role for type I interferon. Tuberculosis (Edinb) 2020; 123:101949. [PMID: 32741537 DOI: 10.1016/j.tube.2020.101949] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/28/2022]
Abstract
Toll-Like Receptor (TLR) 9 stimulation is required for induction of potent immune responses against pathogen invasion. The use of unmethylated CpG as adjuvants in vaccines provides an excellent means of stimulating adaptive immunity. Our data demonstrate that CpG-C provided prolonged immune responses in the mouse model of tuberculosis when formulated with liposomes and the Mycobacterium tuberculosis antigen ESAT-6. A reduction in the mycobacterial burden was best achieved when administered as an intranasal vaccine and was dependent on type I interferon (IFN). There was a significant difference between CpG-C inoculated wild type and IFN-αR1-/- mice, indicating that type I IFN plays a role in the immune response following CpG-C inoculation. Further analysis showed that early NK cell presence was not an absolute requirement, although elevated IFN-γ levels were detected in the lungs of mice within 48 h. The reduction in mycobacterial burden was MyD88-independent as CpG-C inoculated MyD88-/- mice showed comparable mycobacterial burdens to wild type mice with no detriment due to the lack of MyD88. Together our data show that pulmonary stimulation of TLR9 bearing antigen presenting cells resulted in the induction of protective immunity against M. tuberculosis infection that was dependent on type I IFN signaling.
Collapse
Affiliation(s)
- Amber Troy
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Sandra C Esparza-Gonzalez
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Alicia Bartek
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Elizabeth Creissen
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Linda Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Angelo A Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA.
| |
Collapse
|
9
|
Wahid AA, Doekhie A, Sartbaeva A, van den Elsen JMH. Ensilication Improves the Thermal Stability of the Tuberculosis Antigen Ag85b and an Sbi-Ag85b Vaccine Conjugate. Sci Rep 2019; 9:11409. [PMID: 31391509 PMCID: PMC6685958 DOI: 10.1038/s41598-019-47657-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/16/2019] [Indexed: 02/03/2023] Open
Abstract
There is an urgent need for the development of vaccine thermostabilisation methodologies as the maintenance of a continuous and reliable cold chain remains a major hurdle to the global distribution of safe and effective vaccines. Ensilication, a method that encases proteins in a resistant silica cage has been shown to physically prevent the thermal denaturation of a number of model proteins. In this study we investigate the utility of this promising approach in improving the thermal stability of antigens and vaccine conjugates highly relevant to the development of candidate tuberculosis vaccines, including antigen 85b conjugated with the Staphylococcus aureus-protein based adjuvant Sbi. Here we analyse the sensitivity of these constructs to thermal denaturation and demonstrate for the first time the benefits of ensilication in conferring these vaccine-relevant proteins with protection against temperature-induced loss of structure and function without the need for refrigeration. Our results reveal the potential of ensilication in facilitating the storage and transport of vaccines at ambient temperatures in the future and therefore in delivering life-saving vaccines globally, and in particular to remote areas of developing countries where disease rates are often highest.
Collapse
Affiliation(s)
- A A Wahid
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - A Doekhie
- Department of Chemistry, University of Bath, Bath, UK
| | - A Sartbaeva
- Department of Chemistry, University of Bath, Bath, UK.
| | | |
Collapse
|
10
|
Soundarya JSV, Ranganathan UD, Tripathy SP. Current trends in tuberculosis vaccine. Med J Armed Forces India 2019; 75:18-24. [PMID: 30705473 DOI: 10.1016/j.mjafi.2018.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 12/21/2022] Open
Abstract
Despite the global efforts made to control tuberculosis (TB) and the large number of available new anti-TB drugs, TB still affects one-third of the world population. The conventional vaccine bacille Calmette-Guérin (BCG) shows varying efficacy in different populations, and there are safety issues in immunocompromised patients. Hence, there is an urgent requirement for a new and better TB vaccine candidate than BCG. There are several alternate vaccines available for TB such as DNA, subunit, adjuvant, and live-attenuated vaccines. Use of auxotrophic vaccine is an emerging technology. Newer vaccine technologies include vaccine delivery methods such as adenovirus- and cytomegalovirus (CMV)-based vector delivery, chimeric monoclonal antibody, single-chain fragment variable, RNA-lipoplexes, and nanoparticle-based technology. Based on its application, TB vaccines are classified as conventional, prophylactic, booster, therapeutic, and reinfection preventive vaccines. Currently, there are 12 vaccine candidates in clinical trials. In this review, we have briefly discussed about each of these vaccines in different phases of clinical trials. These vaccines should be analyzed further for developing a safe and more efficacious vaccine for TB.
Collapse
Affiliation(s)
- J S V Soundarya
- PhD Research Scholar, Department of Immunology, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Uma Devi Ranganathan
- Scientist 'D', Department of Immunology, National Institute for Research in Tuberculosis, Chennai 600031, India
| | - Srikanth P Tripathy
- Scientist 'G' & Director-in-charge, National Institute for Research in Tuberculosis, Chennai 600031, India
| |
Collapse
|
11
|
Adjuvant Potential of Poly-α-l-Glutamine from the Cell Wall of Mycobacterium tuberculosis. Infect Immun 2018; 86:IAI.00537-18. [PMID: 30104212 DOI: 10.1128/iai.00537-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022] Open
Abstract
Novel adjuvants are in demand for improving the efficacy of human vaccines. The immunomodulatory properties of Mycobacterium tuberculosis cell wall components have been highlighted in the formulation of complete Freund's adjuvant (CFA). We have explored the adjuvant potential of poly-α-l-glutamine (PLG), a lesser-known constituent of the pathogenic mycobacterial cell wall. Immune parameters indicated that the adjuvant potency of PLG was statistically comparable to that of CFA and better than that of alum in the context of H1 antigen (Ag85B and ESAT-6 fusion). At 1 mg/dose, PLG augmented the immune response of Ag85B, BP26, and protective antigen (PA) by increasing serum antibodies and cytokines in the culture supernatant of antigen-stimulated splenocytes. PLG modulated the humoral response of vaccine candidate ESAT-6, eliciting significantly higher levels of total IgG and isotypes (IgG1, IgG2a, and IgG2b). Additionally, the splenocytes from PLG-adjuvanted mice displayed a robust increase in the Th1-specific gamma interferon, tumor necrosis factor alpha, interleukin-2 (IL-2), Th2-specific IL-6 and IL-10, and Th17-specific IL-17A cytokines upon antigenic stimulation. PLG improved the protective efficacy of ESAT-6 by reducing bacillary load in the lung and spleen as well as granuloma formation, and it helped in maintaining vital health parameters of mice challenged with M. tuberculosis The median survival time of PLG-adjuvanted mice was 205 days, compared to 146 days for dimethyl-dioctadecyl ammonium bromide-monophosphoryl lipid A (DDA-MPL)-vaccinated groups and 224 days for Mycobacterium bovis BCG-vaccinated groups. PLG enhanced the efficiency of the ESAT-6 vaccine to the level of BCG and better than that of DDA-MPL (P < 0.05), with no ill effect in C57BL/6J mice. Our results propose that PLG is a promising adjuvant candidate for advanced experimentation.
Collapse
|
12
|
Rhodes SJ, Guedj J, Fletcher HA, Lindenstrøm T, Scriba TJ, Evans TG, Knight GM, White RG. Using vaccine Immunostimulation/Immunodynamic modelling methods to inform vaccine dose decision-making. NPJ Vaccines 2018; 3:36. [PMID: 30245860 PMCID: PMC6141590 DOI: 10.1038/s41541-018-0075-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 06/30/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
Unlike drug dose optimisation, mathematical modelling has not been applied to vaccine dose finding. We applied a novel Immunostimulation/Immunodynamic mathematical modelling framework to translate multi-dose TB vaccine immune responses from mice, to predict most immunogenic dose in humans. Data were previously collected on IFN-γ secreting CD4+ T cells over time for novel TB vaccines H56 and H1 adjuvanted with IC31 in mice (1 dose groups (0.1-1.5 and 15 μg H56 + IC31), 45 mice) and humans (1 dose (50 μg H56/H1 + IC31), 18 humans). A two-compartment mathematical model, describing the dynamics of the post-vaccination IFN-γ T cell response, was fitted to mouse and human data, separately, using nonlinear mixed effects methods. We used these fitted models and a vaccine dose allometric scaling assumption, to predict the most immunogenic human dose. Based on the changes in model parameters by mouse H56 + IC31 dose and by varying the H56 dose allometric scaling factor between mouse and humans, we established that, at a late time point (224 days) doses of 0.8-8 μg H56 + IC31 in humans may be the most immunogenic. A 0.8-8 μg of H-series TB vaccines in humans, may be as, or more, immunogenic, as larger doses. The Immunostimulation/Immunodynamic mathematical modelling framework is a novel, and potentially revolutionary tool, to predict most immunogenic vaccine doses, and accelerate vaccine development.
Collapse
Affiliation(s)
- Sophie J. Rhodes
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Jeremie Guedj
- IAME, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France
| | - Helen A. Fletcher
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Gwenan M. Knight
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| | - Richard G. White
- TB Modelling Group, CMMID, TB Centre, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
13
|
Khoshnood S, Heidary M, Haeili M, Drancourt M, Darban-Sarokhalil D, Nasiri MJ, Lohrasbi V. Novel vaccine candidates against Mycobacterium tuberculosis. Int J Biol Macromol 2018; 120:180-188. [PMID: 30098365 DOI: 10.1016/j.ijbiomac.2018.08.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) is now among the top ten causes of mortality worldwide being resulted in 1.7 million deaths including 0.4 million among people with HIV in 2016. The Bacille Calmette-Guerin (BCG) is the only available TB vaccine which fails to provide consistent protection against pulmonary TB in adults and adolescents despite being efficacious at protecting infants and young children from the most severe, often deadly forms of TB disease. To achieve the goal of global TB elimination by 2050 we will need new interventions including more improved vaccines that are effective in adult individuals who have not been infected with Mycobacterium tuberculosis as well as latently infected or immunocompromised subjects. In recent decades, multiple new vaccine candidates including whole cell vaccines, adjuvanted proteins, and vectored subunit vaccines have entered into the clinical trials. These new TB vaccines are hoped to provide encouraging safety and immunogenicity under various conditions including prevention of TB disease in adolescents and adults, as BCG replacement/boosters, or as therapeutic vaccines to reduce the duration of TB therapy. In this review, we will discuss the status of novel TB vaccine candidates currently under development in preclinical or clinical phases.
Collapse
Affiliation(s)
- Saeed Khoshnood
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Heidary
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehri Haeili
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Michel Drancourt
- Aix-Marseille Univ., IRD, MEPHI, Institut Hospital-Universitaire (IHU) Méditerranée Infection, Marseille, France
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Lohrasbi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Lang R, Schick J. Review: Impact of Helminth Infection on Antimycobacterial Immunity-A Focus on the Macrophage. Front Immunol 2017; 8:1864. [PMID: 29312343 PMCID: PMC5743664 DOI: 10.3389/fimmu.2017.01864] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/08/2017] [Indexed: 12/16/2022] Open
Abstract
Successful immune control of Mycobacterium tuberculosis (MTB) requires robust CD4+ T cell responses, with IFNγs as the key cytokine promoting killing of intracellular mycobacteria by macrophages. By contrast, helminth infections typically direct the immune system toward a type 2 response, characterized by high levels of the cytokines IL-4 and IL-10, which can antagonize IFNγ production and its biological effects. In many countries with high burden of tuberculosis, helminth infections are endemic and have been associated with increased risk to develop tuberculosis or to inhibit vaccination-induced immunity. Mechanistically, regulation of the antimycobacterial immune response by helminths has been mostly been attributed to the T cell compartment. Here, we review the current status of the literature on the impact of helminths on vaccine-induced and natural immunity to MTB with a focus on the alterations enforced on the capacity of macrophages to function as sensors of mycobacteria and effector cells to control their replication.
Collapse
Affiliation(s)
- Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Judith Schick
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
15
|
Sharma SK, Katoch K, Sarin R, Balambal R, Kumar Jain N, Patel N, Murthy KJR, Singla N, Saha PK, Khanna A, Singh U, Kumar S, Sengupta A, Banavaliker JN, Chauhan DS, Sachan S, Wasim M, Tripathi S, Dutt N, Jain N, Joshi N, Penmesta SRR, Gaddam S, Gupta S, Khamar B, Dey B, Mitra DK, Arora SK, Bhaskar S, Rani R. Efficacy and Safety of Mycobacterium indicus pranii as an adjunct therapy in Category II pulmonary tuberculosis in a randomized trial. Sci Rep 2017; 7:3354. [PMID: 28611374 PMCID: PMC5469738 DOI: 10.1038/s41598-017-03514-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/28/2017] [Indexed: 11/09/2022] Open
Abstract
Prolonged treatment of tuberculosis (TB) often leads to poor compliance, default and relapse, converting primary TB patients into category II TB (Cat IITB) cases, many of whom may convert to multi-drug resistant TB (MDR-TB). We have evaluated the immunotherapeutic potential of Mycobacterium indicus pranii (MIP) as an adjunct to Anti-Tubercular Treatment (ATT) in Cat II pulmonary TB (PTB) patients in a prospective, randomized, double blind, placebo controlled, multicentric clinical trial. 890 sputum smear positive Cat II PTB patients were randomized to receive either six intra-dermal injections (2 + 4) of heat-killed MIP at a dose of 5 × 108 bacilli or placebo once in 2 weeks for 2 months. Sputum smear and culture examinations were performed at different time points. MIP was safe with no adverse effects. While sputum smear conversion did not show any statistically significant difference, significantly higher number of patients (67.1%) in the MIP group achieved sputum culture conversion at fourth week compared to the placebo (57%) group (p = 0.0002), suggesting a role of MIP in clearance of the bacilli. Since live bacteria are the major contributors for sustained incidence of TB, the potential of MIP in clearance of the bacilli has far reaching implications in controlling the spread of the disease.
Collapse
Affiliation(s)
| | - Kiran Katoch
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Rohit Sarin
- National Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - Raman Balambal
- National Institute of Research in Tuberculosis (ICMR), Chennai, India
| | - Nirmal Kumar Jain
- SMS Medical College (Hospital for Chest Diseases and TB), Jaipur, Rajasthan, India
| | - Naresh Patel
- NHL Municipal Medical College, Ahmadabad, Gujarat, India
| | | | - Neeta Singla
- National Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - P K Saha
- All India Institute of Medical Sciences, New Delhi, India
| | - Ashwani Khanna
- All India Institute of Medical Sciences, New Delhi, India
| | - Urvashi Singh
- All India Institute of Medical Sciences, New Delhi, India
| | - Sanjiv Kumar
- All India Institute of Medical Sciences, New Delhi, India
| | - A Sengupta
- All India Institute of Medical Sciences, New Delhi, India.,Chest Clinic and Hospital, New Delhi, India
| | - J N Banavaliker
- All India Institute of Medical Sciences, New Delhi, India.,RBTB Hospital, New Delhi, India
| | - D S Chauhan
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Shailendra Sachan
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | - Mohammad Wasim
- National JALMA Institute of Leprosy and Other Mycobacterial Diseases (ICMR), Agra, India
| | | | - Nilesh Dutt
- NHL Municipal Medical College, Ahmadabad, Gujarat, India
| | - Nitin Jain
- SMS Medical College (Hospital for Chest Diseases and TB), Jaipur, Rajasthan, India
| | - Nalin Joshi
- SMS Medical College (Hospital for Chest Diseases and TB), Jaipur, Rajasthan, India
| | | | - Sumanlatha Gaddam
- Mahavir Hospital and Research Centre, Hyderabad, Andhra Pradesh, India
| | - Sanjay Gupta
- Catalyst Clinical Services Pvt. Ltd., New Delhi, India
| | | | - Bindu Dey
- Department of Biotechnology, New Delhi, India
| | | | - Sunil K Arora
- Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | | | - Rajni Rani
- National Institute of Immunology, New Delhi, India. .,Systems Biology laboratory, CSIR-Institute of Genomics & Integrative Biology, New Delhi, India.
| |
Collapse
|
16
|
Tkachuk AP, Gushchin VA, Potapov VD, Demidenko AV, Lunin VG, Gintsburg AL. Multi-subunit BCG booster vaccine GamTBvac: Assessment of immunogenicity and protective efficacy in murine and guinea pig TB models. PLoS One 2017; 12:e0176784. [PMID: 28453555 PMCID: PMC5409163 DOI: 10.1371/journal.pone.0176784] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/17/2017] [Indexed: 11/18/2022] Open
Abstract
New innovative vaccines are highly needed to combat the global threat posed by tuberculosis. Efficient components-antigens and adjuvants-are crucial for development of modern recombinant TB vaccines. This study describes a new vaccine (GamTBvac) consisting of two mycobacterial antigen fusions (Ag85A and ESAT6-CFP10)-with dextran-binding domain immobilized on dextran and mixed with an adjuvant consisting of DEAE-dextran core, and with CpG oligodeoxynucleotides (TLR9 agonists). GamTBvac and its components were assessed for immunogenicity and protective efficacy in GamTBvac-prime/boost and BCG-prime/ GamTBvac-boost in murine and guinea pig TB models. Results show that in both infectious models, GamTBvac has a strong immunogenicity and significant protective effect against Mycobacterium tuberculosis strain H37Rv under aerosol and intravenous challenges. GamTBvac showed a particularly strong protective effect as a BCG booster vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intravenous
- Aerosols
- Animals
- Antibodies, Bacterial/blood
- BCG Vaccine/immunology
- Cell Proliferation/physiology
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Female
- Guinea Pigs
- Immunization
- Immunization, Secondary
- Immunogenicity, Vaccine
- Lung/immunology
- Lymph Nodes/immunology
- Male
- Mice, Inbred C57BL
- Mycobacterium tuberculosis/immunology
- Spleen/immunology
- T-Lymphocytes/immunology
- Tuberculosis/immunology
- Tuberculosis/prevention & control
- Tuberculosis Vaccines/immunology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- A. P. Tkachuk
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. A. Gushchin
- Translational Biomedicine Laboratory, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - V. D. Potapov
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Russia
| | - A. V. Demidenko
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - V. G. Lunin
- Laboratory of bioactive nanostructures, N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - A. L. Gintsburg
- N.F. Gamaleya Federal Research Centre for Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
17
|
Norrby M, Vesikari T, Lindqvist L, Maeurer M, Ahmed R, Mahdavifar S, Bennett S, McClain JB, Shepherd BM, Li D, Hokey DA, Kromann I, Hoff ST, Andersen P, de Visser AW, Joosten SA, Ottenhoff THM, Andersson J, Brighenti S. Safety and immunogenicity of the novel H4:IC31 tuberculosis vaccine candidate in BCG-vaccinated adults: Two phase I dose escalation trials. Vaccine 2017; 35:1652-1661. [PMID: 28216183 DOI: 10.1016/j.vaccine.2017.01.055] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 12/28/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Novel vaccine strategies are required to provide protective immunity in tuberculosis (TB) and prevent development of active disease. We investigated the safety and immunogenicity of a novel TB vaccine candidate, H4:IC31 (AERAS-404) that is composed of a fusion protein of M. tuberculosis antigens Ag85B and TB10.4 combined with an IC31® adjuvant. METHODS BCG-vaccinated healthy subjects were immunized with various antigen (5, 15, 50, 150μg) and adjuvant (0, 100, 500nmol) doses of the H4:IC31 vaccine (n=106) or placebo (n=18) in two randomized, double-blind, placebo-controlled phase I studies conducted in a low TB endemic setting in Sweden and Finland. The subjects were followed for adverse events and CD4+ T cell responses. RESULTS H4:IC31 vaccination was well tolerated with a safety profile consisting of mostly mild to moderate self-limited injection site pain, myalgia, arthralgia, fever and post-vaccination inflammatory reaction at the screening tuberculin skin test injection site. The H4:IC31 vaccine elicited antigen-specific CD4+ T cell proliferation and cytokine production that persisted 18weeks after the last vaccination. CD4+ T cell expansion, IFN-γ production and multifunctional CD4+ Th1 responses were most prominent after two doses of H4:IC31 containing 5, 15, or 50μg of H4 in combination with the 500nmol IC31 adjuvant dose. CONCLUSIONS The novel TB vaccine candidate, H4:IC31, demonstrated an acceptable safety profile and was immunogenic, capable of triggering multifunctional CD4+ T cell responses in previously BCG-vaccinated healthy individuals. These dose-escalation trials provided evidence that the optimal antigen-adjuvant dose combinations are 5, 15, or 50μg of H4 and 500nmol of IC31. TRIAL REGISTRATION ClinicalTrials.gov, NCT02066428 and NCT02074956.
Collapse
Affiliation(s)
- Maria Norrby
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Timo Vesikari
- Vaccine Research Center, University of Tampere, Tampere, Finland
| | - Lars Lindqvist
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Markus Maeurer
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | - Raija Ahmed
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | - Shahnaz Mahdavifar
- TIM, Department of Laboratory Medicine and CAST, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | - Adriëtte W de Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Andersson
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Center for Infectious Medicine (CIM), Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
H1:IC31 vaccination is safe and induces long-lived TNF-α +IL-2 +CD4 T cell responses in M. tuberculosis infected and uninfected adolescents: A randomized trial. Vaccine 2016; 35:132-141. [PMID: 27866772 DOI: 10.1016/j.vaccine.2016.11.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/20/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Control of the tuberculosis epidemic requires a novel vaccine that is effective in preventing tuberculosis in adolescents, a key target population for vaccination against TB. METHODS Healthy adolescents, stratified by M. tuberculosis-infection status, were enrolled into this observer-blinded phase II clinical trial of the protein-subunit vaccine candidate, H1:IC31, comprising a fusion protein (H1) of Ag85B and ESAT-6, formulated with the IC31 adjuvant. Local and systemic adverse events and induced T cell responses were measured after one or two administrations of either 15μg or 50μg of the H1 protein. RESULTS Two hundred and forty participants were recruited and followed up for 224days. No notable safety events were observed regardless of H1 dose or vaccination schedule. H1:IC31 vaccination induced antigen-specific CD4 T cells, co-expressing IFN-γ, TNF-α and/or IL-2. H1:IC31 vaccination of M.tb-uninfected individuals preferentially drove the emergence of Ag85B and ESAT-6 specific TNF-α+IL-2+CD4 T cells, while H1:IC31 vaccination of M.tb-infected individuals resulted in the expansion of Ag85B-specific but not ESAT-6-specific TNF-α+IL-2+CD4 T cells. CONCLUSIONS H1:IC31 was safe and immunogenic in uninfected and M.tb-infected adolescents. Two administrations of the 15μg H1:IC31 dose induced the greatest magnitude immune response, and was considered optimal (South African National Clinical Trials Register, DoH-27-0612-3947; Pan African Clinical Trial Registry, PACTR201403000464306).
Collapse
|
19
|
Sayes F, Pawlik A, Frigui W, Gröschel MI, Crommelynck S, Fayolle C, Cia F, Bancroft GJ, Bottai D, Leclerc C, Brosch R, Majlessi L. CD4+ T Cells Recognizing PE/PPE Antigens Directly or via Cross Reactivity Are Protective against Pulmonary Mycobacterium tuberculosis Infection. PLoS Pathog 2016; 12:e1005770. [PMID: 27467705 PMCID: PMC4965174 DOI: 10.1371/journal.ppat.1005770] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/27/2016] [Indexed: 01/01/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), possesses at least three type VII secretion systems, ESX-1, -3 and -5 that are actively involved in pathogenesis and host-pathogen interaction. We recently showed that an attenuated Mtb vaccine candidate (Mtb Δppe25-pe19), which lacks the characteristic ESX-5-associated pe/ppe genes, but harbors all other components of the ESX-5 system, induces CD4+ T-cell immune responses against non-esx-5-associated PE/PPE protein homologs. These T cells strongly cross-recognize the missing esx-5-associated PE/PPE proteins. Here, we characterized the fine composition of the functional cross-reactive Th1 effector subsets specific to the shared PE/PPE epitopes in mice immunized with the Mtb Δppe25-pe19 vaccine candidate. We provide evidence that the Mtb Δppe25-pe19 strain, despite its significant attenuation, is comparable to the WT Mtb strain with regard to: (i) its antigenic repertoire related to the different ESX systems, (ii) the induced Th1 effector subset composition, (iii) the differentiation status of the Th1 cells induced, and (iv) its particular features at stimulating the innate immune response. Indeed, we found significant contribution of PE/PPE-specific Th1 effector cells in the protective immunity against pulmonary Mtb infection. These results offer detailed insights into the immune mechanisms underlying the remarkable protective efficacy of the live attenuated Mtb Δppe25-pe19 vaccine candidate, as well as the specific potential of PE/PPE proteins as protective immunogens. Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, is one of the most widely spread human pathogens, responsible for more than 9.6 million of new tuberculosis cases and 1.5 million deaths, annually. The resurgence of pulmonary tuberculosis in immuno-compromised patients, including HIV-co-infected populations, and increasing spread of drug-resistant Mtb strains are worrying. Given the estimated 2 billion cases of latent Mtb infections and the only partial efficacy of the unique, currently available tuberculosis-vaccine Mycobacterium bovis BCG (Bacille Calmette-Guerin) it is necessary to develop improved vaccines. Here, we demonstrate that the host cellular immunity, mediated by CD4+ T lymphocytes, specific to the “PE/PPE” families of mycobacterial antigens, contribute to the protection against Mtb-induced disease. We revealed the fine composition of the PE/PPE-specific T cells by characterizing their effector functions and differentiation status. We previously described a live attenuated mycobacterial strain as a vaccine candidate that is able to induce such CD4+ T cells and which displays particular properties at stimulating the cells of the innate immune system. These responses play a central role in the initiation of the host defense and in the protection against tuberculosis. Our results pave the way for further development of candidates in preclinical models of anti-tuberculosis vaccination.
Collapse
Affiliation(s)
- Fadel Sayes
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Alexandre Pawlik
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Matthias I. Gröschel
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Samuel Crommelynck
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Catherine Fayolle
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Paris, France
- INSERM U1041, Paris, France
| | - Felipe Cia
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Daria Bottai
- University of Pisa, Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Pisa, Italy
| | - Claude Leclerc
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Paris, France
- INSERM U1041, Paris, France
| | - Roland Brosch
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
| | - Laleh Majlessi
- Institut Pasteur, Unité de Pathogénomique Mycobactérienne Intégrée, Paris, France
- * E-mail:
| |
Collapse
|
20
|
Agger EM. Novel adjuvant formulations for delivery of anti-tuberculosis vaccine candidates. Adv Drug Deliv Rev 2016; 102:73-82. [PMID: 26596558 DOI: 10.1016/j.addr.2015.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 01/18/2023]
Abstract
There is an urgent need for a new and improved vaccine against tuberculosis for controlling this disease that continues to pose a global health threat. The current research strategy is to replace the present BCG vaccine or boost BCG-immunity with subunit vaccines such as viral vectored- or protein-based vaccines. The use of recombinant proteins holds a number of production advantages including ease of scalability, but requires an adjuvant inducing cell-mediated immune responses. A number of promising novel adjuvant formulations have recently been designed and show evidence of induction of cellular immune responses in humans. A common trait of effective TB adjuvants including those already in current clinical testing is a two-component approach combining a delivery system with an appropriate immunomodulator. This review summarizes the status of current TB adjuvant research with a focus on the division of labor between delivery systems and immunomodulators.
Collapse
Affiliation(s)
- Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
21
|
Kaufmann E, Spohr C, Battenfeld S, De Paepe D, Holzhauser T, Balks E, Homolka S, Reiling N, Gilleron M, Bastian M. BCG Vaccination Induces Robust CD4+ T Cell Responses to Mycobacterium tuberculosis Complex–Specific Lipopeptides in Guinea Pigs. THE JOURNAL OF IMMUNOLOGY 2016; 196:2723-32. [DOI: 10.4049/jimmunol.1502307] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/12/2016] [Indexed: 01/11/2023]
|
22
|
Abstract
Through thousands of years of reciprocal coevolution, Mycobacterium tuberculosis has become one of humanity's most successful pathogens, acquiring the ability to establish latent or progressive infection and persist even in the presence of a fully functioning immune system. The ability of M. tuberculosis to avoid immune-mediated clearance is likely to reflect a highly evolved and coordinated program of immune evasion strategies that interfere with both innate and adaptive immunity. These include the manipulation of their phagosomal environment within host macrophages, the selective avoidance or engagement of pattern recognition receptors, modulation of host cytokine production, and the manipulation of antigen presentation to prevent or alter the quality of T-cell responses. In this article we review an extensive array of published studies that have begun to unravel the sophisticated program of specific mechanisms that enable M. tuberculosis and other pathogenic mycobacteria to persist and replicate in the face of considerable immunological pressure from their hosts. Unraveling the mechanisms by which M. tuberculosis evades or modulates host immune function is likely to be of major importance for the development of more effective new vaccines and targeted immunotherapy against tuberculosis.
Collapse
|
23
|
Knudsen NPH, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, Fox CB, Meinke A, D'Oro U, Casini D, Bonci A, Billeskov R, De Gregorio E, Rappuoli R, Harandi AM, Andersen P, Agger EM. Different human vaccine adjuvants promote distinct antigen-independent immunological signatures tailored to different pathogens. Sci Rep 2016; 6:19570. [PMID: 26791076 PMCID: PMC4726129 DOI: 10.1038/srep19570] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
The majority of vaccine candidates in clinical development are highly purified proteins and peptides relying on adjuvants to enhance and/or direct immune responses. Despite the acknowledged need for novel adjuvants, there are still very few adjuvants in licensed human vaccines. A vast number of adjuvants have been tested pre-clinically using different experimental conditions, rendering it impossible to directly compare their activity. We performed a head-to-head comparison of five different adjuvants Alum, MF59®, GLA-SE, IC31® and CAF01 in mice and combined these with antigens from M. tuberculosis, influenza, and chlamydia to test immune-profiles and efficacy in infection models using standardized protocols. Regardless of antigen, each adjuvant had a unique immunological signature suggesting that the adjuvants have potential for different disease targets. Alum increased antibody titers; MF59® induced strong antibody and IL-5 responses; GLA-SE induced antibodies and Th1; CAF01 showed a mixed Th1/Th17 profile and IC31® induced strong Th1 responses. MF59® and GLA-SE were strong inducers of influenza HI titers while CAF01, GLA-SE and IC31® enhanced protection to TB and chlamydia. Importantly, this is the first extensive attempt to categorize clinical-grade adjuvants based on their immune profiles and protective efficacy to inform a rational development of next generation vaccines for human use.
Collapse
Affiliation(s)
- Niels Peter H Knudsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Cecilia Buonsanti
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Yuan Zhang
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA, USA
| | | | | | - Ugo D'Oro
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Daniele Casini
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Alessandra Bonci
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ennio De Gregorio
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Ali M Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
24
|
Differential influence of nutrient-starved Mycobacterium tuberculosis on adaptive immunity results in progressive tuberculosis disease and pathology. Infect Immun 2015; 83:4731-9. [PMID: 26416911 DOI: 10.1128/iai.01055-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/17/2015] [Indexed: 12/25/2022] Open
Abstract
When infected with Mycobacterium tuberculosis, most individuals will remain clinically healthy but latently infected. Latent infection has been proposed to partially involve M. tuberculosis in a nonreplicating stage, which therefore represents an M. tuberculosis phenotype that the immune system most likely will encounter during latency. It is therefore relevant to examine how this particular nonreplicating form of M. tuberculosis interacts with the host immune system. To study this, we first induced a state of nonreplication through prolonged nutrient starvation of M. tuberculosis in vitro. This resulted in nonreplicating persistence even after prolonged culture in phosphate-buffered saline. Infection with either exponentially growing M. tuberculosis or nutrient-starved M. tuberculosis resulted in similar lung CFU levels in the first phase of the infection. However, between week 3 and 6 postinfection, there was a very pronounced increase in bacterial levels and associated lung pathology in nutrient-starved-M. tuberculosis-infected mice. This was associated with a shift from CD4 T cells that coexpressed gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α) or IFN-γ, TNF-α, and interleukin-2 to T cells that only expressed IFN-γ. Thus, nonreplicating M. tuberculosis induced through nutrient starvation promotes a bacterial form that is genetically identical to exponentially growing M. tuberculosis yet characterized by a differential impact on the immune system that may be involved in undermining host antimycobacterial immunity and facilitate increased pathology and transmission.
Collapse
|
25
|
Mothé BR, Lindestam Arlehamn CS, Dow C, Dillon MBC, Wiseman RW, Bohn P, Karl J, Golden NA, Gilpin T, Foreman TW, Rodgers MA, Mehra S, Scriba TJ, Flynn JL, Kaushal D, O'Connor DH, Sette A. The TB-specific CD4(+) T cell immune repertoire in both cynomolgus and rhesus macaques largely overlap with humans. Tuberculosis (Edinb) 2015; 95:722-735. [PMID: 26526557 DOI: 10.1016/j.tube.2015.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/23/2015] [Accepted: 07/26/2015] [Indexed: 01/05/2023]
Abstract
Non-human primate (NHP) models of tuberculosis (TB) immunity and pathogenesis, especially rhesus and cynomolgus macaques, are particularly attractive because of the high similarity of the human and macaque immune systems. However, little is known about the MHC class II epitopes recognized in macaques, thus hindering the establishment of immune correlates of immunopathology and protective vaccination. We characterized immune responses in rhesus macaques vaccinated against and/or infected with Mycobacterium tuberculosis (Mtb), to a panel of antigens currently in human vaccine trials. We defined 54 new immunodominant CD4(+) T cell epitopes, and noted that antigens immunodominant in humans are also immunodominant in rhesus macaques, including Rv3875 (ESAT-6) and Rv3874 (CFP10). Pedigree and inferred restriction analysis demonstrated that this phenomenon was not due to common ancestry or inbreeding, but rather presentation by common alleles, as well as, promiscuous binding. Experiments using a second cohort of rhesus macaques demonstrated that a pool of epitopes defined in the previous experiments can be used to detect T cell responses in over 75% of individual monkeys. Additionally, 100% of cynomolgus macaques, irrespective of their latent or active TB status, responded to rhesus and human defined epitope pools. Thus, these findings reveal an unexpected general repertoire overlap between MHC class II epitopes recognized in both species of macaques and in humans, showing that epitope pools defined in humans can also be used to characterize macaque responses, despite differences in species and antigen exposure. The results have general implications for the evaluation of new vaccines and diagnostics in NHPs, and immediate applicability in the setting of macaque models of TB.
Collapse
Affiliation(s)
- Bianca R Mothé
- Department of Biology, CSUSM, San Marcos, CA 92096, USA; La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA.
| | | | - Courtney Dow
- Department of Biology, CSUSM, San Marcos, CA 92096, USA
| | - Myles B C Dillon
- La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Roger W Wiseman
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Patrick Bohn
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Julie Karl
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Nadia A Golden
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Trey Gilpin
- Department of Biology, CSUSM, San Marcos, CA 92096, USA
| | - Taylor W Foreman
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15216, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, LA 70803, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town 7925, South Africa
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15216, USA
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| |
Collapse
|
26
|
Luabeya AKK, Kagina BMN, Tameris MD, Geldenhuys H, Hoff ST, Shi Z, Kromann I, Hatherill M, Mahomed H, Hanekom WA, Andersen P, Scriba TJ, Schoeman E, Krohn C, Day CL, Africa H, Makhethe L, Smit E, Brown Y, Suliman S, Hughes EJ, Bang P, Snowden MA, McClain B, Hussey GD. First-in-human trial of the post-exposure tuberculosis vaccine H56:IC31 in Mycobacterium tuberculosis infected and non-infected healthy adults. Vaccine 2015; 33:4130-40. [PMID: 26095509 DOI: 10.1016/j.vaccine.2015.06.051] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/07/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND H56:IC31 is a candidate tuberculosis vaccine comprising a fusion protein of Ag85B, ESAT-6 and Rv2660c, formulated in IC31 adjuvant. This first-in-human, open label phase I trial assessed the safety and immunogenicity of H56:IC31 in healthy adults without or with Mycobacterium tuberculosis (M.tb) infection. METHODS Low dose (15 μg H56 protein in 500 nmol IC31) or high dose (50 μg H56, 500 nmol IC31) vaccine was administered intramuscularly thrice, at 56-day intervals. Antigen-specific T cell responses were measured by intracellular cytokine staining and antibody responses by ELISA. RESULTS One hundred and twenty-six subjects were screened and 25 enrolled and vaccinated. No serious adverse events were reported. Nine subjects (36%) presented with transient cardiovascular adverse events. The H56:IC31 vaccine induced antigen-specific IgG responses and Th1 cytokine-expressing CD4(+) T cells. M.tb-infected vaccinees had higher frequencies of H56-induced CD4(+) T cells than uninfected vaccinees. Low dose vaccination induced more polyfunctional (IFN-γ(+)TNF-α(+)IL-2(+)) and higher frequencies of H56-specific CD4(+) T cells compared with high dose vaccination. A striking increase in IFN-γ-only-expressing CD4(+) T cells, displaying a CD45RA(-)CCR7(-) effector memory phenotype, emerged after the second high-dose vaccination in M.tb-infected vaccinees. TNF-α(+)IL-2(+) H56-specific memory CD4(+) T cells were detected mostly after low-dose H56 vaccination in M.tb-infected vaccinees, and predominantly expressed a CD45RA(-)CCR7(+) central memory phenotype. Our results support further clinical testing of H56:IC31.
Collapse
Affiliation(s)
- Angelique Kany Kany Luabeya
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa.
| | - Benjamin M N Kagina
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa; Vaccines for Africa Initiative, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Michele D Tameris
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Hennie Geldenhuys
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Soren T Hoff
- Statens Serum Institut (SSI), Copenhagen, Denmark
| | | | | | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Hassan Mahomed
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa; Western Cape Government and Stellenbosch University, Cape Town, South Africa
| | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Elisma Schoeman
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Colleen Krohn
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Cheryl L Day
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa; Department of Global Health, Rollins School of Public Health, Atlanta, GA, USA; Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Hadn Africa
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Lebohang Makhethe
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Yolande Brown
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Sara Suliman
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - E Jane Hughes
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa
| | - Peter Bang
- Statens Serum Institut (SSI), Copenhagen, Denmark
| | | | | | - Gregory D Hussey
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine (IDM), School of Child and Adolescent Health, University of Cape Town, University of Cape Town, Cape Town, South Africa; Vaccines for Africa Initiative, Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| |
Collapse
|
27
|
Vaccines for TB: Lessons from the Past Translating into Future Potentials. J Immunol Res 2015; 2015:916780. [PMID: 26146643 PMCID: PMC4469767 DOI: 10.1155/2015/916780] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/11/2015] [Accepted: 05/18/2015] [Indexed: 01/20/2023] Open
Abstract
Development of vaccines for infectious diseases has come a long way with recent advancements in adjuvant developments and discovery of new antigens that are capable of eliciting strong immunological responses for sterile eradication of disease. Tuberculosis (TB) that kills nearly 2 million of the population every year is also one of the highlights of the recent developments. The availability or not of diagnostic methods for infection has implications for the control of the disease by the health systems but is not related to the immune surveillance, a phenomenon derived from the interaction between the bacteria and their host. Here, we will review the immunology of TB and current vaccine candidates for TB. Current strategies of developing new vaccines against TB will also be reviewed in order to further discuss new insights into immunotherapeutic approaches involving adjuvant and antigens combinations that might be of potential for the control of TB.
Collapse
|
28
|
Rosales-Mendoza S, Ríos-Huerta R, Angulo C. An overview of tuberculosis plant-derived vaccines. Expert Rev Vaccines 2015; 14:877-89. [PMID: 25683476 DOI: 10.1586/14760584.2015.1015996] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tuberculosis (TB) is a leading fatal infectious disease to which the current BCG vaccine has a questionable efficacy in adults. Thus, the development of improved vaccines against TB is needed. In addition, decreasing the cost of vaccine formulations is required for broader vaccination coverage through global vaccination programs. In this regard, the use of plants as biofactories and delivery vehicles of TB vaccines has been researched over the last decade. These studies are systematically analyzed in the present review and placed in perspective. It is considered that substantial preclinical trials are still required to address improvements in expression levels as well as immunological data. Approaches for testing additional antigenic configurations with higher yields and improved immunogenic properties are also discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | | | | |
Collapse
|
29
|
Pepponi I, Diogo GR, Stylianou E, van Dolleweerd CJ, Drake PMW, Paul MJ, Sibley L, Ma JKC, Reljic R. Plant-derived recombinant immune complexes as self-adjuvanting TB immunogens for mucosal boosting of BCG. PLANT BIOTECHNOLOGY JOURNAL 2014; 12:840-50. [PMID: 24629003 DOI: 10.1111/pbi.12185] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 02/12/2014] [Accepted: 02/18/2014] [Indexed: 06/03/2023]
Abstract
Progress with protein-based tuberculosis (TB) vaccines has been limited by poor availability of adjuvants suitable for human application. Here, we developed and tested a novel approach to molecular engineering of adjuvanticity that circumvents the need for exogenous adjuvants. Thus, we generated and expressed in transgenic tobacco plants the recombinant immune complexes (RICs) incorporating the early secreted Ag85B and the latency-associated Acr antigen of Mycobacterium tuberculosis, genetically fused as a single polypeptide to the heavy chain of a monoclonal antibody to Acr. The RICs were formed by virtue of the antibody binding to Acr from adjacent molecules, thus allowing self-polymerization of the complexes. TB-RICs were purified from the plant extracts and shown to be biologically active by demonstrating that they could bind to C1q component of the complement and also to the surface of antigen-presenting cells. Mice immunized with BCG and then boosted with two intranasal immunizations with TB-RICs developed antigen-specific serum IgG antibody responses with mean end-point titres of 1 : 8100 (Acr) and 1 : 24 300 (Ag85B) and their splenocytes responded to in vitro stimulation by producing interferon gamma. 25% of CD4+ proliferating cells simultaneously produced IFN-γ, IL-2 and TNF-α, a phenotype that has been linked with protective immune responses in TB. Importantly, mucosal boosting of BCG-immunized mice with TB-RICs led to a reduced M. tuberculosis infection in their lungs from log10 mean = 5.69 ± 0.1 to 5.04 ± 0.2, which was statistically significant. We therefore propose that the plant-expressed TB-RICs represent a novel molecular platform for developing self-adjuvanting mucosal vaccines.
Collapse
Affiliation(s)
- Ilaria Pepponi
- St George's University of London, London, UK; Jenner Institute, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Darrah PA, Bolton DL, Lackner AA, Kaushal D, Aye PP, Mehra S, Blanchard JL, Didier PJ, Roy CJ, Rao SS, Hokey DA, Scanga CA, Sizemore DR, Sadoff JC, Roederer M, Seder RA. Aerosol vaccination with AERAS-402 elicits robust cellular immune responses in the lungs of rhesus macaques but fails to protect against high-dose Mycobacterium tuberculosis challenge. THE JOURNAL OF IMMUNOLOGY 2014; 193:1799-811. [PMID: 25024382 DOI: 10.4049/jimmunol.1400676] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Development of a vaccine against pulmonary tuberculosis may require immunization strategies that induce a high frequency of Ag-specific CD4 and CD8 T cells in the lung. The nonhuman primate model is essential for testing such approaches because it has predictive value for how vaccines elicit responses in humans. In this study, we used an aerosol vaccination strategy to administer AERAS-402, a replication-defective recombinant adenovirus (rAd) type 35 expressing Mycobacterium tuberculosis Ags Ag85A, Ag85B, and TB10.4, in bacillus Calmette-Guérin (BCG)-primed or unprimed rhesus macaques. Immunization with BCG generated low purified protein derivative-specific CD4 T cell responses in blood and bronchoalveolar lavage. In contrast, aerosolized AERAS-402 alone or following BCG induced potent and stable Ag85A/b-specific CD4 and CD8 effector T cells in bronchoalveolar lavage that largely produced IFN-γ, as well as TNF and IL-2. Such responses induced by BCG, AERAS-402, or both failed to confer overall protection following challenge with 275 CFUs M. tuberculosis Erdman, although vaccine-induced responses associated with reduced pathology were observed in some animals. Anamnestic T cell responses to Ag85A/b were not detected in blood of immunized animals after challenge. Overall, our data suggest that a high M. tuberculosis challenge dose may be a critical factor in limiting vaccine efficacy in this model. However, the ability of aerosol rAd immunization to generate potent cellular immunity in the lung suggests that using different or more immunogens, alternative rAd serotypes with enhanced immunogenicity, and a physiological challenge dose may achieve protection against M. tuberculosis.
Collapse
Affiliation(s)
- Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Diane L Bolton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Andrew A Lackner
- Tulane National Primate Research Center, Covington, LA 70433; and
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433; and
| | - Pyone Pyone Aye
- Tulane National Primate Research Center, Covington, LA 70433; and
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433; and
| | | | - Peter J Didier
- Tulane National Primate Research Center, Covington, LA 70433; and
| | - Chad J Roy
- Tulane National Primate Research Center, Covington, LA 70433; and
| | - Srinivas S Rao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
31
|
Yuk JM, Jo EK. Host immune responses to mycobacterial antigens and their implications for the development of a vaccine to control tuberculosis. Clin Exp Vaccine Res 2014; 3:155-67. [PMID: 25003089 PMCID: PMC4083068 DOI: 10.7774/cevr.2014.3.2.155] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/01/2014] [Accepted: 04/07/2014] [Indexed: 01/17/2023] Open
Abstract
Tuberculosis (TB) remains a worldwide health problem, causing around 2 million deaths per year. Despite the bacillus Calmette Guérin vaccine being available for more than 80 years, it has limited effectiveness in preventing TB, with inconsistent results in trials. This highlights the urgent need to develop an improved TB vaccine, based on a better understanding of host-pathogen interactions and immune responses during mycobacterial infection. Recent studies have revealed a potential role for autophagy, an intracellular homeostatic process, in vaccine development against TB, through enhanced immune activation. This review attempts to understand the host innate immune responses induced by a variety of protein antigens from Mycobacterium tuberculosis, and to identify future vaccine candidates against TB. We focus on recent advances in vaccine development strategies, through identification of new TB antigens using a variety of innovative tools. A new understanding of the host-pathogen relationship, and the usefulness of mycobacterial antigens as novel vaccine candidates, will contribute to the design of the next generation of vaccines, and to improving the host protective immune responses while limiting immunopathology during M. tuberculosis infection.
Collapse
Affiliation(s)
- Jae-Min Yuk
- Department of Microbiology and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
32
|
Trentini MM, de Oliveira FM, Gaeti MPN, Batista AC, Lima EM, Kipnis A, Junqueira-Kipnis AP. Microstructured liposome subunit vaccines reduce lung inflammation and bacterial load after Mycobacterium tuberculosis infection. Vaccine 2014; 32:4324-32. [PMID: 24951861 DOI: 10.1016/j.vaccine.2014.06.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/04/2014] [Accepted: 06/06/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Tuberculosis is a disease affecting millions of people throughout the world. One of the main problems in controlling the disease is the low efficacy of the Bacillus Calmette-Guérin (BCG) vaccine in protecting young adults. The development of new vaccines that induce a long-lasting immune response or that stimulate the immunity induced by BCG may improve the control of tuberculosis. METHODS The use of microstructured liposomes containing HspX, with or without MPL or CpG DNA adjuvants, as vaccines for tuberculosis was evaluated. The HspX-specific humoral and cellular immune responses to the different vaccine formulations were compared. RESULTS All vaccines containing liposome microparticles and HspX were immunogenic. Vaccines formulated with CpG DNA and HspX induced the strongest humoral and cellular immune responses, mainly by inducing interferon-γ and tumor necrosis factor-α expression by both CD4(+) and CD8(+) T cells. HspX and MPL mainly induced CD8(+) T-cell activation and specific humoral responses. When evaluated the protective efficacy of the formulations against Mycobacterium tuberculosis challenge, the microstructured liposome containing L-HspX and L-HspX-CPG DNA reduced both lung inflammatory lesions and the bacterial load. CONCLUSION We have thus demonstrated, for the first time, the use of microstructured liposomes as an adjuvant and delivery system for a vaccine formulation against tuberculosis.
Collapse
Affiliation(s)
- Monalisa Martins Trentini
- Laboratório de Imunopatologia das Doenças Infecciosas, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Brazil
| | - Fábio Muniz de Oliveira
- Laboratório de Bacteriologia Molecular, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Brazil
| | | | - Aline Carvalho Batista
- Laboratório de Patologia, Faculdade de Odontologia, Universidade Federal de Goiás, Brazil
| | - Eliana Martins Lima
- Laboratório de Nanotecnologia Farmacêutica - FarmaTec, Faculdade de Farmácia, Universidade Federal de Goiás, Brazil
| | - André Kipnis
- Laboratório de Bacteriologia Molecular, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Brazil
| | - Ana Paula Junqueira-Kipnis
- Laboratório de Imunopatologia das Doenças Infecciosas, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Brazil.
| |
Collapse
|
33
|
Junqueira-Kipnis AP, Marques Neto LM, Kipnis A. Role of Fused Mycobacterium tuberculosis Immunogens and Adjuvants in Modern Tuberculosis Vaccines. Front Immunol 2014; 5:188. [PMID: 24795730 PMCID: PMC4005953 DOI: 10.3389/fimmu.2014.00188] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 04/09/2014] [Indexed: 11/13/2022] Open
Abstract
Several approaches have been developed to improve or replace the only available vaccine for tuberculosis (TB), BCG (Bacille Calmette Guerin). The development of subunit protein vaccines is a promising strategy because it combines specificity and safety. In addition, subunit protein vaccines can be designed to have selected immune epitopes associated with immunomodulating components to drive the appropriate immune response. However, the limited antigens present in subunit vaccines reduce their capacity to stimulate a complete immune response compared with vaccines composed of live attenuated or killed microorganisms. This deficiency can be compensated by the incorporation of adjuvants in the vaccine formulation. The fusion of adjuvants with Mycobacterium tuberculosis (Mtb) proteins or immune epitopes has the potential to become the new frontier in the TB vaccine development field. Researchers have addressed this approach by fusing the immune epitopes of their vaccines with molecules such as interleukins, lipids, lipoproteins, and immune stimulatory peptides, which have the potential to enhance the immune response. The fused molecules are being tested as subunit vaccines alone or within live attenuated vector contexts. Therefore, the objectives of this review are to discuss the association of Mtb fusion proteins with adjuvants; Mtb immunogens fused with adjuvants; and cytokine fusion with Mtb proteins and live recombinant vectors expressing cytokines. The incorporation of adjuvant molecules in a vaccine can be complex, and developing a stable fusion with proteins is a challenging task. Overall, the fusion of adjuvants with Mtb epitopes, despite the limited number of studies, is a promising field in vaccine development.
Collapse
Affiliation(s)
- Ana Paula Junqueira-Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| | - Lázaro Moreira Marques Neto
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia , Brazil
| |
Collapse
|
34
|
Fox CB, Kramer RM, Barnes V L, Dowling QM, Vedvick TS. Working together: interactions between vaccine antigens and adjuvants. THERAPEUTIC ADVANCES IN VACCINES 2014; 1:7-20. [PMID: 24757512 DOI: 10.1177/2051013613480144] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of vaccines containing adjuvants has the potential to enhance antibody and cellular immune responses, broaden protective immunity against heterogeneous pathogen strains, enable antigen dose sparing, and facilitate efficacy in immunocompromised populations. Nevertheless, the structural interplay between antigen and adjuvant components is often not taken into account in the published literature. Interactions between antigen and adjuvant formulations should be well characterized to enable optimum vaccine stability and efficacy. This review focuses on the importance of characterizing antigen-adjuvant interactions by summarizing findings involving widely used adjuvant formulation platforms, such as aluminum salts, emulsions, lipid vesicles, and polymer-based particles. Emphasis is placed on the physicochemical basis of antigen-adjuvant associations and the appropriate analytical tools for their characterization, as well as discussing the effects of these interactions on vaccine potency.
Collapse
|
35
|
Singh S, Saraav I, Sharma S. Immunogenic potential of latency associated antigens against Mycobacterium tuberculosis. Vaccine 2014; 32:712-6. [DOI: 10.1016/j.vaccine.2013.11.065] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 10/26/2022]
|
36
|
Fraser CK, Diener KR, Brown MP, Hayball JD. Improving vaccines by incorporating immunological coadjuvants. Expert Rev Vaccines 2014; 6:559-78. [PMID: 17669010 DOI: 10.1586/14760584.6.4.559] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.
Collapse
Affiliation(s)
- Cara K Fraser
- Experimental Therapeutics Laboratory, Hanson Institute, and School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Australia.
| | | | | | | |
Collapse
|
37
|
Mutwiri G, Gerdts V, van Drunen Littel-van den Hurk S, Auray G, Eng N, Garlapati S, Babiuk LA, Potter A. Combination adjuvants: the next generation of adjuvants? Expert Rev Vaccines 2014; 10:95-107. [DOI: 10.1586/erv.10.154] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Lingnau K, Riedl K, von Gabain A. IC31®and IC30, novel types of vaccine adjuvant based on peptide delivery systems. Expert Rev Vaccines 2014; 6:741-6. [DOI: 10.1586/14760584.6.5.741] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Acosta J, Carpio Y, Valdés I, Velázquez J, Zamora Y, Morales R, Morales A, Rodríguez E, Estrada MP. Co-administration of tilapia alpha-helical antimicrobial peptides with subunit antigens boost immunogenicity in mice and tilapia (Oreochromis niloticus). Vaccine 2014; 32:223-9. [DOI: 10.1016/j.vaccine.2013.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/06/2013] [Indexed: 10/26/2022]
|
40
|
Hamborg M, Kramer R, Schanté CE, Agger EM, Christensen D, Jorgensen L, Foged C, Middaugh CR. The physical stability of the recombinant tuberculosis fusion antigens h1 and h56. J Pharm Sci 2013; 102:3567-78. [PMID: 23873630 DOI: 10.1002/jps.23669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 06/03/2013] [Accepted: 06/24/2013] [Indexed: 01/13/2023]
Abstract
The recombinant fusion proteins hybrid 1 [H1 (Ag85B-ESAT-6)] and hybrid 56 [H56 (Ag85B-ESAT-6-Rv2660c)] derived from Mycobacterium tuberculosis are promising antigens for subunit vaccines against tuberculosis. Both antigens are early batches of antigens to be enrolled in human clinical trials and it is therefore important to characterize their conformational stability in solution as well as upon interaction with adjuvants. In this study, the physical stability of the two antigens was characterized using a number of biophysical techniques. Dynamic light scattering and sodium dodecyl sulfate-polyacrylamide gel electrophoresis analyses demonstrated that both antigens exist as a distribution of multimeric states under nonstressed conditions. Their conformational stability was monitored as a function of pH and temperature and visualized in three-index empirical phase diagrams. Both antigens showed a gradual loss of secondary as well as tertiary structure as a function of temperature, with no cooperative transitions observed. Preformulation studies with the Th1-inducing cationic adjuvant CAF01 showed that the antigens were almost completely surface adsorbed. Upon adsorption, the liposome size increased; however, the physical stabilities of the bound and the unbound antigens were comparable. This study provides important information about the biophysical properties of H1 and H56 and highlights the analytical challenges of characterizing complex vaccine formulations.
Collapse
Affiliation(s)
- Mette Hamborg
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Szabo A, Gogolak P, Pazmandi K, Kis-Toth K, Riedl K, Wizel B, Lingnau K, Bacsi A, Rethi B, Rajnavolgyi E. The two-component adjuvant IC31® boosts type i interferon production of human monocyte-derived dendritic cells via ligation of endosomal TLRs. PLoS One 2013; 8:e55264. [PMID: 23405128 PMCID: PMC3566214 DOI: 10.1371/journal.pone.0055264] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 12/20/2012] [Indexed: 11/26/2022] Open
Abstract
The aim of this study was to characterize and identify the mode of action of IC31®, a two-component vaccine adjuvant. We found that IC31® was accumulated in human peripheral blood monocytes, MHC class II positive cells and monocyte-derived DCs (moDCs) but not in plasmacytoid DCs (pDCs). In the presence of IC31® the differentiation of inflammatory CD1a+ moDCs and the secretion of chemokines, TNF-α and IL-6 cytokines was inhibited but the production of IFNβ was increased. Sustained addition of IC31® to differentiating moDCs interfered with IκBα phosphorylation, while the level of phospho-IRF3 increased. We also showed that both IC31® and its KLK component exhibited a booster effect on type I IFN responses induced by the specific ligands of TLR3 or TLR7/8, whereas TLR9 ligand induces type I IFN production only in the presence of IC31® or ODN1. Furthermore, long term incubation of moDCs with IC31® caused significantly higher expression of IRF and IFN genes than a single 24 hr treatment. The adjuvant activity of IC31® on the IFN response was shown to be exerted through TLRs residing in the vesicular compartment of moDCs. Based on these results IC31® was identified as a moDC modulatory adjuvant that sets the balance of the NF-κB and IRF3 mediated signaling pathways to the production of IFNβ. Thus IC31® is emerging as a potent adjuvant to increase immune responses against intracellular pathogens and cancer in future vaccination strategies.
Collapse
Affiliation(s)
- Attila Szabo
- Department of Immunology, Medical and Health Science Centre, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
García-Arévalo C, Bermejo-Martín JF, Rico L, Iglesias V, Martín L, Rodríguez-Cabello JC, Arias FJ. Immunomodulatory Nanoparticles from Elastin-Like Recombinamers: Single-Molecules for Tuberculosis Vaccine Development. Mol Pharm 2013; 10:586-97. [DOI: 10.1021/mp300325v] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Carmen García-Arévalo
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | - Jesús F. Bermejo-Martín
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Lucia Rico
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Verónica Iglesias
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Laura Martín
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | | | - F. Javier Arias
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| |
Collapse
|
43
|
|
44
|
Nasal and skin delivery of IC31(®)-adjuvanted recombinant HSV-2 gD protein confers protection against genital herpes. Vaccine 2012; 30:4361-8. [PMID: 22682292 DOI: 10.1016/j.vaccine.2012.02.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 12/28/2011] [Accepted: 02/06/2012] [Indexed: 11/21/2022]
Abstract
Genital herpes caused by herpes simplex virus type 2 (HSV-2) remains the leading cause of genital ulcers worldwide. Given the disappointing results of the recent genital herpes vaccine trials in humans, development of novel vaccine strategies capable of eliciting protective mucosal and systemic immune responses to HSV-2 is urgently required. Here we tested the ability of the adjuvant IC31(®) in combination with HSV-2 glycoprotein D (gD) used through intranasal (i.n.), intradermal (i.d.), or subcutaneous (s.c.) immunization routes for induction of protective immunity against genital herpes infection in C57BL/6 mice. Immunization with gD plus IC31(®) through all three routes of immunization developed elevated gD-specific serum antibody responses with HSV-2 neutralizing activity. Whereas the skin routes promoted the induction of a mixed IgG2c/IgG1 isotype profile, the i.n. route only elicited IgG1 antibodies. All immunization routes were able to induce gD-specific IgG antibody responses in the vaginas of mice immunized with IC31(®)-adjuvanted gD. Although specific lymphoproliferative responses were observed in splenocytes from mice of most groups vaccinated with IC31(®)-adjuvanted gD, only i.d. immunization resulted in a significant splenic IFN-γ response. Further, immunization with gD plus IC31(®) conferred 80-100% protection against an otherwise lethal vaginal HSV-2 challenge with amelioration of viral replication and disease severity in the vagina. These results warrant further exploration of IC31(®) for induction of protective immunity against genital herpes and other sexually transmitted infections.
Collapse
|
45
|
Kamath AT, Mastelic B, Christensen D, Rochat AF, Agger EM, Pinschewer DD, Andersen P, Lambert PH, Siegrist CA. Synchronization of dendritic cell activation and antigen exposure is required for the induction of Th1/Th17 responses. THE JOURNAL OF IMMUNOLOGY 2012; 188:4828-37. [PMID: 22504654 DOI: 10.4049/jimmunol.1103183] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The dendritic cell (DC) targeting/activation patterns required to elicit Th1/Th17 responses remain undefined. One postulated requirement was that of a physical linkage between Ags and immunomodulators. Accordingly, the separate same-site administration of Ag85B-ESAT-6 (hybrid-1 protein; H1), a mycobacterial fusion Ag, and the CAF01 liposome-based adjuvant induced similar Ab and weak Th2 responses as those of coformulated H1/CAF01 but failed to elicit Th1/Th17 responses. Yet, this separate same-site injection generated the same type and number of activated Ag(+)/adjuvant(+) DCs in the draining lymph nodes (LN) as that of protective H1/CAF01 immunization. Thus, targeting/activating the same DC population by Ag and adjuvant is not sufficient to elicit Th1/Th17 responses. To identify the determinants of Th1/Th17 adjuvanticity, in vivo tracking experiments using fluorescently labeled Ag and adjuvant identified that a separate same-site administration elicits an additional early Ag(+)/adjuvant(-) DC population with a nonactivated phenotype, resulting from the earlier targeting of LN DCs by H1 than by CAF01 molecules. This asynchronous targeting pattern was mimicked by the injection of free H1 prior to or with, but not after, H1/CAF01 or H1/CpG/ aluminum hydroxide immunization. The injection of soluble OVA similarly prevented the induction of Th1 responses by OVA/CAF01. Using adoptively transferred OT-2 cells, we show that the Ag targeting of LN DCs prior to their activation generates nonactivated Ag-pulsed DCs that recruit Ag-specific T cells, trigger their initial proliferation, but interfere with Th1 induction in a dose-dependent manner. Thus, the synchronization of DC targeting and activation is a critical determinant for Th1/Th17 adjuvanticity.
Collapse
Affiliation(s)
- Arun T Kamath
- World Health Organization Collaborating Center for Vaccinology and Neonatal Immunology, Department of Pathology-Immunology, Medical Faculty of the University of Geneva, Geneva 1211, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lin PL, Dietrich J, Tan E, Abalos RM, Burgos J, Bigbee C, Bigbee M, Milk L, Gideon HP, Rodgers M, Cochran C, Guinn KM, Sherman DR, Klein E, Janssen C, Flynn JL, Andersen P. The multistage vaccine H56 boosts the effects of BCG to protect cynomolgus macaques against active tuberculosis and reactivation of latent Mycobacterium tuberculosis infection. J Clin Invest 2011; 122:303-14. [PMID: 22133873 DOI: 10.1172/jci46252] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that one-third of the world's population is infected with Mycobacterium tuberculosis. Infection typically remains latent, but it can reactivate to cause clinical disease. The only vaccine, Mycobacterium bovis bacillus Calmette-Guérin (BCG), is largely ineffective, and ways to enhance its efficacy are being developed. Of note, the candidate booster vaccines currently under clinical development have been designed to improve BCG efficacy but not prevent reactivation of latent infection. Here, we demonstrate that administering a multistage vaccine that we term H56 in the adjuvant IC31 as a boost to vaccination with BCG delays and reduces clinical disease in cynomolgus macaques challenged with M. tuberculosis and prevents reactivation of latent infection. H56 contains Ag85B and ESAT-6, which are two of the M. tuberculosis antigens secreted in the acute phase of infection, and the nutrient stress-induced antigen Rv2660c. Boosting with H56/IC31 resulted in efficient containment of M. tuberculosis infection and reduced rates of clinical disease, as measured by clinical parameters, inflammatory markers, and improved survival of the animals compared with BCG alone. Boosted animals showed reduced pulmonary pathology and extrapulmonary dissemination, and protection correlated with a strong recall response against ESAT-6 and Rv2660c. Importantly, BCG/H56-vaccinated monkeys did not reactivate latent infection after treatment with anti-TNF antibody. Our results indicate that H56/IC31 boosting is able to control late-stage infection with M. tuberculosis and contain latent tuberculosis, providing a rationale for the clinical development of H56.
Collapse
Affiliation(s)
- Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Novel protein-based pneumococcal vaccines administered with the Th1-promoting adjuvant IC31 induce protective immunity against pneumococcal disease in neonatal mice. Infect Immun 2011; 80:461-8. [PMID: 22025519 DOI: 10.1128/iai.05801-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Streptococcus pneumoniae is responsible for many vaccine-preventable deaths, annually causing around 1 million deaths in children younger than 5 years of age. A new generation of pneumococcal vaccines based on conserved proteins is being developed. We evaluated the immunogenicities and protective efficacies of four pneumococcal protein vaccine candidates, PcsB, StkP, PsaA, and PspA, in a neonatal mouse model. Mice were immunized three times and challenged intranasally with virulent pneumococci. All four proteins were immunogenic in neonatal mice, and antibody (Ab) responses were significantly enhanced by the novel adjuvant IC31, which consists of an antibacterial peptide (KLKL5KLK) and a synthetic oligodeoxynucleotide, ODN1a, that signals through Toll-like receptor 9 (TLR9). Two single proteins, StkP and PspA, combined with IC31 significantly reduced pneumococcal bacteremia but had no effects on lung infection. Three proteins, PcsB, StkP, and PsaA, were evaluated with alum or IC31. IC31 enhanced Ab responses and avidity to all three proteins, whereas alum enhanced Ab responses and avidity to StkP and PsaA only. Mice receiving the trivalent protein formulation with IC31 had significantly reduced bacteremia and lung infection compared to unvaccinated mice, but the level of protection was dependent on the dose of IC31. When PspA was added to the trivalent protein formulation, the dose of IC31 needed to obtain protective immunity could be reduced. These results demonstrate that a novel pneumococcal protein-based vaccine is immunogenic at an early age of mice and emphasize the benefits of using a combination of conserved proteins and an effective adjuvant to elicit potent protective immunity against invasive pneumococcal disease.
Collapse
|
48
|
Grover A, Troudt J, Arnett K, Izzo L, Lucas M, Strain K, McFarland C, Hall Y, McMurray D, Williams A, Dobos K, Izzo A. Assessment of vaccine testing at three laboratories using the guinea pig model of tuberculosis. Tuberculosis (Edinb) 2011; 92:105-11. [PMID: 21962569 DOI: 10.1016/j.tube.2011.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/06/2011] [Accepted: 09/10/2011] [Indexed: 11/24/2022]
Abstract
The guinea pig model of tuberculosis is used extensively in different locations to assess the efficacy of novel tuberculosis vaccines during pre-clinical development. Two key assays are used to measure protection against virulent challenge: a 30 day post-infection assessment of mycobacterial burden and long-term post-infection survival and pathology analysis. To determine the consistency and robustness of the guinea pig model for testing vaccines, a comparative assessment between three sites that are currently involved in testing tuberculosis vaccines from external providers was performed. Each site was asked to test two "subunit" type vaccines in their routine animal model as if testing vaccines from a provider. All sites performed a 30 day study, and one site also performed a long-term survival/pathology study. Despite some differences in experimental approach between the sites, such as the origin of the Mycobacterium tuberculosis strain and the type of aerosol exposure device used to infect the animals and the source of the guinea pigs, the data obtained between sites were consistent in regard to the ability of each "vaccine" tested to reduce the mycobacterial burden. The observations also showed that there was good concurrence between the results of short-term and long-term studies. This validation exercise means that efficacy data can be compared between sites.
Collapse
Affiliation(s)
- Ajay Grover
- Colorado State University, Department of Microbiology, Immunology & Pathology, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Locht C, Rouanet C. Nouveaux vaccins antituberculeux. Arch Pediatr 2011; 18:1023-7. [DOI: 10.1016/j.arcped.2011.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 04/22/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
|
50
|
Tyagi AK, Nangpal P, Satchidanandam V. Development of vaccines against tuberculosis. Tuberculosis (Edinb) 2011; 91:469-78. [DOI: 10.1016/j.tube.2011.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 01/14/2011] [Accepted: 01/16/2011] [Indexed: 12/20/2022]
|