SciELO - Scientific Electronic Library Online

 
vol.18 issue2Dynamics of Neisseria meningitidis interactions with human cellular barriers and immune effectors author indexsubject indexarticles search
Home Pagealphabetic serial listing  

My SciELO

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

  • Have no similar articlesSimilars in SciELO

Share


Vaccimonitor

Print version ISSN 1025-028XOn-line version ISSN 1025-0298

Vaccimonitor vol.18 no.2 Ciudad de la Habana May-Aug. 2009

 

ARTICULOS ORIGINALES

 

Adjuvantes:Un componente esencial de las vacunas de Neisseria.

Adjuvants: an essential component of neisseria vaccines.

Reinaldo Acevedo1, Belkis Romeu1, Judith del Campo1, Elizabeth Gonzáles1, Julio Balboa1, Caridad Zayas1, Maribel Cuello1, Osmir Cabrera1, Miriam Lastre1, Valerie A. Ferro2, and Oliver Pérez1.

1Inmunology Department, Research Vice Presidency, Finlay Institute, P. O. Box 16017, Havana, Cuba.

2University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, 27 Taylor Street, Glasgow, G4 0NR, UK.

email: racevedo@finlay.edu.cu



Abstract

Adjuvants may be classified into delivery systems and immune potentiator or modulator molecules based on their mechanism of action. Neisseria vaccines containing traditional adjuvants such as aluminium salts have existed for long time, but meningitis caused by Neisseria meningitidis serogroups, particularly serogroup B, continues to be a global health problem. Novel strategies have applied in silico and recombinant technologies to develop "universal" antigens (e.g. proteins, peptides and plasmid DNA) for vaccines, but these antigens have been shown to be poorly immunogenic even when alum adjuvanted, implying a need for better vaccine design. In this work we review the use of natural, detoxified, or synthetic molecules in combination with antigens to activate the innate immune system and to modulate the adaptive immune responses. In the main, antigenic and imune potentiator signals are delivered using nano-, micro-particles, alum, or emulsions. The importance of interaction between adjuvants and antigens to activate and target dendritic cells, the bridge between the innate and adaptive immune systems, will be discussed. In addition, nasal vaccine strategies based on the development of mucosal adjuvants and Neisseria derivatives to eliminate the pathogen at the site of infection provide promising adjuvants effective not only against respiratory pathogens, but also against pathogens responsible for enteric and sexually transmitted diseases.

Keywords: Adjuvant, micro-particle, nano-particle, Neisseria, vaccine, delivery system, immune potentiator.


Introduction

Pathogenic Neisseria species are mainly responsible for meningococcal and gonorrhoeal disease. Approximately 62 million people get infected annually with gonorrhoea, while N. meningitidis is one of the main killers responsible for bacterial meningitis, principally in young children, and the only bacterium capable of generating epidemics taking more than 30, 000 lives each year (1). In this paper, we will focus mainly on the impact of adjuvants on N. meningitidis vaccine formulation.

Current meningococcal vaccines are administered by the parenteral route and adsorbed onto aluminium salts; early vaccines used native polysaccharides (Ps) from serogroups A, C, Y, and W135 but Ps vaccines are poorly immunogenic in young infants, fail to induce immunological memory and do not provide protection for more than 3-5 years (1). Immunogenicity of Ps was greatly improved when chemically conjugated to a protein carrier, while also inducing long term memory in adults and young infants. Conjugated vaccines have been shown to be very effective, but are too expensive for developing countries, nevertheless adjuvant strategies are being applied to reduce costs and increase immunogenicity. Furthermore, the development of vaccines against serogroup B, accounting for 2000-8000 deaths annually in developed and developing countries, has been vastly hindered because its' Ps is less immunogenic and cross reacts with sialylated proteins in human tissues. Thus, outer membrane vesicles (OMV) containing high amounts of surface protein antigens from the pathogen have been used for epidemic control e.g. in Cuba (2) and Norway (3). Despite the OMV being strain-specific, some level of cross reaction has been detected with the Cuban vaccine (Men B Finlay) (4) Novel strategies using reverse vaccinology (5) and DNA libraries constructed from bacterial genomes (6) have been investigated in an attempt to predict universal antigens to protect against B sero subtypes (7). However, these proteins, peptides or plasmid DNA are also proving to be poorly immunogenic and the traditional alum adjuvant is not sufficient to induce appropriate levels of protection. New adjuvant strategies are therefore being devised based on a combination of these antigens with immune potentiator, molecules and/or delivery systems capable of efficiently targeting immune response components such as dendritic cells (DC).

Immune potentiator, modulator molecules and delivery systems

The innate immune system utilizes multiple receptors (Pattern Recognition Receptors, PRR) of fixed specificity to recognize an enormously diverse array of ligands on microbes known also as Pathogen-Associated Molecular Patterns (PAMPs) (8). The most important PRR studied are the toll-like receptors (TLR) which are transmembrane proteins that recognize proteins and Malyala et al. (19) recently confirmed these results.

Neisseria derivatives as vaccine adjuvants

Neisseria derivatives have been used to adjuvant many antigens and advances in this field of study are perhaps as important as the development of Neisseria vaccines themselves. The Adjuvant Finlay PL 1 (AFPL1) is a Neisseria derived PL that contains several PAMPs such as LPS, porins, and DNA traces. Ovalbumin incorporated in AFPL1 is very immunogenic when administered by the parenteral route, as are allergen antigens co-adsorbed onto alum with the AFPL1; inducing a Th1 response (20). Alternativly, the mucosally administered Adjuvant Finlay Cochleate 1 (AFCo1) is a microtubular structure derived from Neisseria PL interaction with calcium (20). It is more stable and immunogenic than AFPL1 and has also been used to adjuvant parenteral administered antigens from Leishmania and malaria (21-22). Intranasal immunization of AFCo1 with incorporated or co-administered ovalbumin has been shown to induce strong systemic and mucosal immune responses (20).

Protollin™ is the commercial name of a proprietary adjuvant from ID Biomedical Corporation (subsequently GSK). This is a non-covalent complex between Neisseria proteosomes and Shigella flexneri 2ª LPS used mainly as a mucosal adjuvant. Protollin™ is a safe formulation administered to humans inducing mucosal and systemic immune responses against Shigella (23) and has also been shown to protect mice from respiratory syncytial virus (24).

Nasal route for Neisseria Vaccines

The respiratory tract is the site of entry and colonization of N. meningitidis. In many cases non symptomatic individuals can transmit the pathogen to others (25). Parenteral immunization of the current Neisseria vaccines is effective in inducing systemic immune responses, however to protect against infection, the induction of immune responses at mucosal surfaces is required (26). Conjugate Ps vaccines induce some level of mucosal immune response and it has been suggested that one of the most important successes of this vaccine relies on the induction of herd immunity through mucosal stimulation (27). Nevertheless, some formulations using liposomes encapsulating serogroup C meningococcal Ps conjugated to Escherichia coli heat labile enterotoxin mutant, LTK63 have also been shown to induce potent mucosal and systemic immune response when administered intranasally (28). Similarly, when, the conjugated vaccine Menjugate C was reformulated with chitosan, instead of alum, and intranasally administered to humans, it showed similar systemic immune responses and enhanced mucosal immune responses compared with parenteral administration of the vaccine (29).

OMV from N. meningitidis B have also been used in clinical trials; but the nasal immunization required 10 fold more antigen per dose than the injectable form to induce similar PAMPs like: lipopolysaccharides (LPS, TLR4), lipopeptides (TLR1 and 6), flagellin (TLR5) and nucleic acids (TLR7 or 8, ssRNA; TLR9, unmethylated CpG) from pathogens. To date 13 TLR have been identified in mammals (9).

More than 60 million doses of the Cuban VA-MENGOC-BC® Neisserial vaccine have been administered and it has shown a good safety profile. It is composed of OMV which are nano proteoliposome that contain important porin antigens (PorA and PorB) and native LPS that stimulate DC through TLR4, inducing IL-12 and gIFN cytokines characteristic of a Th1 pattern (10). One of the most important features of neisserial proteoliposomes are their ability to deliver antigenic and immune activating signals to DC (11).

Since LPS has also been described as toxic endotoxin, some groups have worked on detoxified forms of it such as the 3-0-desacyl-4'-monophosphoryl lipid A (MPL) that comes from LPS of the Gram-negative Salmonella minnesota R595 or synthetic LPS analogs such as RC529, which are less toxic than native LPS. MPL and RC529 interact with TLR4 inducing a Th1 response similar to native LPS, but have failed to induce long term memory of the stimulated CD4+ T cell subset (12). When these structures have been encapsulated in poly(lactide-co-glycolide) (PLG) microparticles an enhanced immune response has been elicited against N. meningitidis B antigens adsorbed on the microparticle surface (13).

MPL adsorbed onto alum is a GlaxoSmithKline Biologicals (GSK) adjuvant used in humans, known as AS04 (14). It has been used with several outer proteins from Neisseria and in addition to the depot effect of alum, co-administered MPL has been shown to redirect the classic Th2 pattern induced by alum alone to a mixed Th1/Th2 response, which favours the induction of protective immune responses. Emulsions have also been formulated with Neisseria antigens, including: MF59 a safe oil-in-water adjuvant used in humans (14) and Titermax for experimental use only (15). Lucila et al. (15) demonstrated that formulations using meningococcal C Ps (PsC) conjugated to OMV from N. meningitidis B were very efficient in inducing immune responses and long lasting memory in a neonatal mice model. Co-encapsulation of Ps on liposomes with immune potentiator or modulator molecules such as CpG and CD40 is being studied as a non-covalent alternative to conjugated Ps vaccines (16-17) and probably offers a less expensive option to developing countries. OMV have also been used to co-adjuvant the immune response to plasmid DNA (6).

CpG is the ligand for TLR9 and activation leads to an enhanced humoral and cell-mediated immune response through B-cell stimulation to produce more immunoglobulin, as well as promotion of a Th1 pattern and cytokine secretion by DC (18). PLG anionic microparticles uploaded with CpG have been described by Singh et al.(13) as a potent delivery system for co-administered Neisseria meningitidis B recombinant systemic immune responses. However, these studies did not evaluate mucosal immune responses (30). We too have found that IN immunization with different OMV induce greater mucosal immune responses than parenteral administration (31). The IN route has also been used to test vaccine candidates against N. gonorrhoea (32). Recombinant proteins from this pathogen adjuvanted with cholera toxin subunit B induced high immune responses at the genital tract, showing that this route can also be used to stimulate distal mucosal responses.

Commentary

Neisserial vaccine progression is very much related to adjuvant development. Firstly, because Neisseria derivatives are being used to adjuvant parenteral and mucosal vaccines candidates from other microorganisms and secondly, novel formulations based on combinations of delivery systems and immune potentiator or modulator molecules are emerging to face the global meningitis problem.

A "universal" B meningococcal vaccine strategy must be accompanied by the selection of the right adjuvants, and enables a number of adjuvant formulations to be examined head-to-head. This would further remove the problem of making the wrong choice of adjuvant or discarding good candidates as a result of selecting a poor adjuvant combination. Conjugated Ps vaccines have represented a huge advance in protecting against Neisseria pathogens; however they are too expensive, particularly for the developing world. Current adjuvants could lead to the improvement of new ways to formulate less expensive and equally or more immunogenic antigens as an alternative to conjugated Ps vaccines.

Development of better mucosal adjuvants is another approach to obtain more effective vaccines against Neisseria, for the induction of mucosal, as well as systemic immune response, which could potentially protect vaccinees from the pathogen and the population from pathogen spread. We predict that over the next few years, this field will see a plethora of combined current and novel adjuvant technologies directed towards the mucosal route of administration.

References

1. State of the art of new vaccine and development. Immunization, Vaccines and Biologicals. WHO 2006.01

2. Sierra G, Campa HC, Varcacel NM, Izquierdo PL, Sotolongo PF, Casanueva GV, et al. Vaccine against group B Neisseria meningitidis: Protection trial and mass vaccination results in Cuba. NIPH. Ann. 1991;14:195-210

3. Bjune G, Hoiby EA, Gronnesby JK, Arnesen O, Fredriksen JH, Halstensen A, et al. Effect of outer membrane vesicle vaccine against group meningococcal disease in Norway. Lancet 1991;338:1093-6


4. Boutriau D, Poolman J, Borrow R, et al. Immunogenicity and safety of three doses of a bivalent (B:4:p1.19,15 and B:4:p1.7-2,4) meningococcal outer membrane vesicle vaccine in healthy adolescents. Clin. Vaccine Immunol 2005;14(1):65-73

5. Kelly DF, Rappuoli R. Reverse vaccinology and vaccines for serogroup B Neisseria meningitidis. Adv Exp Med Biol 2005;568:217-223

6. Yero D, Pajón R, Caballero E, González S, Cobas K, Fariñas M, et al. A novel method to screen genomic libraries that combines genomic immunization with the prime-boost strategy. FEMS Immunol Med Mic 2007;50(3):430-3

7. Holst J. Strategies for development of universal vaccines against meningococcal serogroup B disease: the most promising options and the challenges evaluating them.Hum Vaccin 2007;3(6):290-4

8. Hargreaves DC and Medzhitov R. Innate Sensors of Microbial Infection.Journal of Clinical Immunology.2005;25;6:503-10

9. Lahiri A, Das P, Chakravortty D. Engagement of TLR signaling as adjuvant: Towards smarter vaccine and beyond.. Vaccine 2008;26:6777-83

10. Pérez O, Lastre M, Lapinet J, et al. Immune Response Induction and New Effector Mechanisms Possibly Involved in Protection of Cuban Anti-Meningococcal BC Vaccine. Infect Immun 2001; 69:4502-08

11. Rodríguez T, Pérez O, Menager N, Ugrinovic S, Bracho G, Mastroeni P. Interactions of proteoliposome from serogroup B Neisseria meningitidis with bone marrow-derived dendritic cells and macrophages: adjuvants and antigen delivery. Vaccine 2005;23:1312-21

12. Thompson BS, Chilton PM, Ward JR, Evans JT and Mitchell TC. The low toxicity versions of LPS, MPL adjuvant and RC529, are efficient adjuvants for CD4+ Tcells. J Leukoc Biol 2005;78:1273-80

13. Singh M, Kazzaz J, Chesko J, Soenawan E, Ugozzoli M, Giuliani M, Pizza M, Rappouli R, O'hagan DT. Anionic Microparticles are a Potent Delivery System for Recombinant Antigens from Neisseria meningitidis Serotype B. J Pharm Sci. 2004;93:273-82.

14. Tagliabue A, Rappuoli R. Vaccine adjuvants: the dream becomes real. Hum Vaccin 2008;4(5):566-73

15. Kazzaz J, Singh M, Ugozzoli M, Chesko J, Soenawan E, O'Hagan DT. Encapsulation of the immune potentiators MPL and RC529 in PLG microparticles enhances their potency. J Control Release 2006;110(3):566-73.

16. Lucila O. Fukasawa, Waldely O. Dias, Rocilda P.F. Schenkman, I. Raw, Martha M. Tanizaki. Adjuvant can improve protection induced by OMV vaccine against Neisseria meningitidis serogroups B/C in neonatal mice. FEMS Immunology and Medical Microbiology 2004;41:205-10

17. Hatzifoti C, Bacon A, Marriott H, Laing P, Heath AW. Liposomal Co-Entrapment of CD40mAb Induces Enhanced IgG Responses against Bacterial Polysaccharide and Protein. PLoS ONE 3 2008;(6):e2368.

18. Bacon AD, Gregoriadis G, Laing P. Liposomal Vaccine Compositions Comprising a Polysaccharide Antigen and a Protein Adjuvant. US20080317838 A1, 2008:

19. Klinman DM Adjuvant activity of CpG oligodeoxynucleotides. Int Rev Immunol 2006;25(3-4):135-54.

20. Malyala P, Chesko J, Ugozzoli M, Goodsell A, Zhou F, Vajdy M, O'Hagan DT, Singh M. The potency of the adjuvant, CpG oligos, is enhanced by encapsulation in PLG microparticles.. J Pharm Sci. 2008; 97(3):1155-64.

21. Pérez O, Lastre M, Cabrera O et al. New Vaccines Require Potent Adjuvants like AFPL1 and AFCo1. Scandinavian Journal of Immunology 2007;66: 271-277.

22. Bracho G, Zayas C, Wang L, Coppel R, Perez O and Petrovsky N. AFCo1, a meningococcal B-derived cochleate adjuvant, strongly enhances antibody and T-cell immunity against Plasmodium falciparum merozoite surface protein 4 and 5. Malaria Journal 2009;8(35) in press

23. Pérez O, Bracho G, Lastre M, et al. Novel adjuvant based on a proteoliposome derived cochleate structure containing native lipopolysaccharide as a pathogen associated molecular pattern. Immunology and Cell Biology 2004; 82:603-10

24. Fries LF, Montemarano AD, Mallett CP, Taylor DN, Hale TL, Lowell GH. Safety and immunogenicity of a proteosome-Shigella flexneri2a lipopolysaccharide vaccine administered intranasally to healthy adults. Infect Immun 2001;69:4545-53

25. Cyr SL, Jones T, Stoica-Popescu I, Burt D, Ward BJ. C57Bl/6 mice are protected from respiratory syncytial virus (RSV) challenge and IL-5 associated pulmonary eosinophilic infiltrates following intranasal immunization with Protollin-eRSV vaccine. Vaccine 2007; 25: 3228-32.

26. Yazdankhah SP and Caugant DA. Neisseria meningitidis: an overview of the carriage state. J Med Microbiol 53, 2004:821-32


27. Sarno ML, Mattos AP, Lee P. Intranasal vaccines for protection against respiratory and systemic bacterial infection. Expert Rev. Vaccines 2007;6: 419-429.

28. Maiden MC, Ibarz-Pavón AB, Urwin R, Gray SJ, Andrews NJ, Clarke SC, et al. Impact of meningococcal serogroup C conjugate vaccines on carriage and herd immunity. J Infect Dis 2008;197:737-43.

29. Baudner BC, Balland O, Giuliani MM, et al. Enhancement of protective efficacy following intranasal immunization with vaccine plus a nontoxic LTK63 mutant delivered with nanoparticles. Infect Immun 2002;70:4785-90.

30. Huo Z, Sinha R, McNeela EA, Borrow R, Giemza R, Cosgrove C, et al. Induction of Protective Serum Meningococcal Bactericidal and Diphtheria-Neutralizing Antibodies and Mucosal Immunoglobulin A in Volunteers by Nasal Insufflations of the Neisseria meningitidis Serogroup C Polysaccharide-CRM197 Conjugate Vaccine Mixed with Chitosan. Infect Immun 2005;73: 8256-65.

31. Haneberg B, Dalseg R, Wedege E, Høiby EA, Haugen IL, Oftung F, et al. Intranasal Administration of a Meningococcal Outer Membrane Vesicle Vaccine Induces Persistent Local Mucosal Antibodies and Serum Antibodies with Strong Bactericidal Activity in Humans. Infect Immun 1998; 66:1334-41

32. del Campo J, Lastre M, Bracho G, et al. Immunological evaluation of bacterial derived Cochleate and Proteoliposome as mucosal adjuvants Vaccine 2006;24 (S2):50-1

33. Price GA, Russell MW and Cornelissen CN. Intranasal Administration of Recombinant Neisseria gonorrhoeae Transferrin Binding Proteins A and B Conjugated to the Cholera Toxin B Subunit Induces Systemic and Vaginal Antibodies in Mice. Inf. Imm 2005;73(7):3945-53

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License