Вы находитесь на странице: 1из 3

PERSPECTIVE

D.J. Smith
Prospects in Caries Vaccine
The Forsyth Institute, 245 First Street, Cambridge, MA 02146, USA; dsmith@
forsyth.org Development
J Dent Res 91(3):225-226, 2012

KEY WORDS: mucosal immunity, Streptococcus mutans,


vaccine(s), adjuvants, dental caries, DNA vaccines.

V accines have long been attractive for broad-based pub-


lic health coverage of infectious diseases, of which dental
caries is one of the most pervasive. We are now in the seventh
antibody by including Salmonella-derived flagellin protein, a
ligand of Toll-like receptor 5, at the time of pGJA-P/VAX admin-
istration. The S. mutans-specific sequences of this construct code
decade of vaccine approaches intended to prevent or arrest den- for the glucan-binding domain (GLU) of GtfB and the A-P adhe-
tal caries. In the 1940s, Lactobacilli were among the principal sin-associated fragment of the Pac surface protein. Addition of
bacterial targets. In the latter half of the 20th century, attention recombinant flagellin to this intranasally administered DNA vac-
was focused on mutans streptococci (Streptococcus mutans and cine enhanced both serum IgG and salivary IgA antibody levels to
S. sobrinus), given their repeated association with dental caries, the A-P fragment. Anti-GLU antibody responses were not reported.
especially that of early childhood. Although more recent use of These authors suggested that flagellin-enhanced antibody
molecular probes has implicated additional bacteria (e.g., bifi- responses to the adhesin were responsible for greater caries reduc-
dobacteria) in the development of early lesions (Kanasi et al., tions than had been seen with chitosan or bupivacaine, based on
2010), most of the immunologic interest remains with S. mutans. comparison with earlier studies. Direct adjuvant comparison in
Protein, recombinant or synthetic peptide, protein-carbohydrate follow-up studies could confirm this suggestion.
conjugate, or DNA-based active vaccines—each has been suc- Investigators have sought to enhance antigen uptake to muco-
cessful in experimental systems. However, no vaccines have sal lymphatic tissues by combining adjuvants with acceptable
been brought to market. Although there are several reasons for delivery vehicles. Delivery systems such as PLGA microparticles,
this, including financial risk/reward for development, the ability liposomes, or ISCOMs have resulted in generally modest
to induce and maintain high levels of protective antibody in oral increases in salivary IgA antibody activity, although such increases
fluids has been a significant challenge. Several strategies have have sometimes been associated with protection in animal models
been pursued to identify powerful and safe adjuvants that can be for dental caries. Additional chitosan incorporation into, or coat-
used in the mucosal immune context. In the present issue of the ing on, microparticles has been reported to be beneficial to muco-
JDR, Shi and co-workers have evaluated the use of recombinant sal immune induction, possibly by promoting M-cell uptake
flagellin protein as an adjuvant for the intranasal delivery of an (Borges et al., 2007). New methods to target M-cell uptake of
anti-caries DNA vaccine (Shi et al., 2012). antigens may offer enhancement of mucosal IgA antibody forma-
DNA vaccine approaches for dental caries have had a history tion sufficient to sustain protection (Azizi et al., 2010).
of success in animal models. Incorporating message for one or Heat-labile enterotoxins (HLT) from Vibrio cholera (LT-I) or
more of the known S. mutans virulence antigens, investigators Escherichia coli (LT- II) induce vigorous mucosal (and sys-
have used such constructs to induce systemic IgG and/or mucosal temic) immune responses when used as adjuvants for protein
IgA antibody responses to S. mutans glucosyltransferases and antigens. However, their inherent toxicity precludes the use of
adhesins. These responses were associated with reductions in car- such holotoxins in human vaccines. Even genetically detoxified
ies lesions in rodents. Since DNA vaccines are relatively weak mutants of E. coli LT-II have been associated with neurological
immunogens, there has been significant effort, especially by the issues following their use with intranasally delivered inactivated
Fan group, to identify compounds and conditions that would influenza vaccine (Lewis et al., 2009), likely a function of LT
increase antibody levels which could also improve the protective binding to neuronal gangliosides. However, work continues to
effect. Useful adjuvants appropriate for intranasal (IN) vaccine identify safe HLT subcomponents which retain significant
delivery have been sought, in part because the IN route has been immunomodulatory characteristics that can be used at the pre-
shown to enhance regional mucosal antibody levels, including ferred intranasal site (Liang et al., 2009).
salivary antibody. This group has had some success using bupiva- Unlike active vaccines, passive applications of antibody do not
caine (Jia et al., 2004) and chitosan (Xu et al., 2006) adjuvants to require adjuvants. Experimentally successful passive vaccines for
increase salivary IgA antibody responses. Shi and co-workers (Shi dental caries have taken many forms, including mouse monoclonal
et al., 2012) sought to further enhance salivary IgA and serum IgG IgG, bovine or simian IgG, chicken IgY, and tobacco-plant-derived
transgenic SIgA/G antibody (reviewed in Smith and Mattos-
DOI: 10.1177/0022034511425928 Graner, 2008). The aforementioned transgenic vaccine also showed
some promise in small clinical trials in adults (Ma et al., 1998).
Received September 13, 2011; Accepted September 15, 2011
Most of these vaccines were directed to the same glucan-generating
© International & American Associations for Dental Research or adhesin targets as their active counterparts. Since passive vac-
225
226  Smith J Dent Res 91(3) 2012

cines do not induce a continuous antibody-secreting host response, References


effective delivery of the passive reagents is key to their success.
Azizi A, Kumar A, Diaz-Mitoma F, Mestecky J (2010). Enhancing oral
Intriguingly, single-chain antibody (scFv), directed against a vaccine potency by targeting intestinal M cells. PLoS Pathog 6:
S. mutans adhesin and expressed on the surface of Lactobacilli, has e1001147.
been associated with dental caries reductions in a rat model (Kruger Borges O, Tavares J, de Souza A, Borchard G, Jungingee HE, Cordeiro-
et al., 2005). This approach would offer sustained delivery of anti- da-Silva A (2007). Evaluation of the immune response following
body as long as the probiotic colonizes the host. a short oral vaccination schedule with hepatitis B antigen encapsu-
lated into alginate-coated chitosan nanoparticles. Eur J Pharm Sci 32:
Some of the uncertainty about caries vaccine effectiveness 278-290.
stems from our limited understanding of the influence which the Jia R, Guo JH, Fan MW, Bian Z, Chen Z, Peng B, et al. (2004). Mucosal
natural mucosal immune response has on the entry of cariogenic immunization against dental caries with plasmid DNA encoding pac
mutans streptococci into the oral biofilm. Attempts to correlate gene of Streptococcus mutans in rats. Vaccine 22:2511-2516.
Kanasi E, Dewhirst FE, Chalmers NI, Kent R, Moore A, Hughes CV, et al.
host antibody and caries levels have been problematic. Recently,
(2010). Clonal analysis of the microbiota of severe early childhood car-
Nogueira and co-workers (2005) provided suggestive evidence ies. Caries Res 44:485-497.
that salivary IgA antibody to certain virulence antigens delayed S. Kruger C, Hultberg A, van Dollenweerd C, Marcotte H, Hammarström
mutans colonization in very young children. These potentially L (2005). Passive immunization by lactobacilli expressing single-
protective responses theoretically could have been provoked chain antibodies against Streptococcus mutans. Mol Biotechnol 31:
221-231.
through antigenic stimulation by similar epitopes on earlier-colo- Lewis DJ, Huo Z, Barnett S, Kromann I, Giemza R, Galiza E, et al. (2009).
nizing oral streptococci. Transient facial nerve paralysis (Bell’s palsy) following intranasal
Dental caries vaccines, directed to key components of mutans delivery of a genetically detoxified mutant of Escherichia coli heat
streptococcal colonization and enhanced by safe and effective labile toxin. PLoS ONE 4:e6999.
adjuvants and optimal delivery vehicles, are likely to be forth- Liang S, Hosur KB, Nawar HF, Russell MW, Connell TD, Hajishengallis G
(2009). In vivo and in vitro adjuvant activities of the B subunit of Type
coming. Will these vaccines lower dental caries risk in children? IIb heat-labile enterotoxin (LT-IIb-B5) from Escherichia coli. Vaccine
Effectiveness may be tied to immunization timing and a reduc- 27:4302-4308.
tion in environmental risk factors. For example, one strategy Ma JK, Hikmat BY, Wycoff K, Vine ND, Chargelegue D, Yu L, et al. (1998).
may be to immunize prior to permanent acquisition of cario- Characterization of a recombinant plant monoclonal secretory antibody
and preventive immunotherapy in humans. Nat Med 4:601-606.
genic S. mutans early in childhood. Ultimately, clinical trials of
Nogueira RD, Alves AC, Napimoga MH, Smith DJ, Mattos-Graner RO
immunologically superior dental caries vaccine formulations (2005). Characterization of salivary IgA responses in children heavily
will determine their usefulness for public health applications. exposed to the oral bacteria Streptococcus mutans: influence of specific
Although scientific, regulatory, and economic hurdles need to be antigen recognition in infection. Infect Immun 73:5675-5684.
cleared to reach this goal, the potential benefit continues to Shi W, Liu F, Yang J, Zhou D, Chen Y, Zhang Y, et al. (2012). Flagellin
enhances saliva IgA response and protection of anti-caries DNA vac-
make the race worth running.
cine. J Dent Res 91:249-254.
Smith DJ, Mattos-Graner RO (2008). Secretory immunity following mutans
Acknowledgments streptococcal infection or immunization. Curr Top Microbiol Immunol
319:131-156.
The author received no financial support and declares no poten- Xu QA, Yu F, Fan MW, Bian Z, Chen Z, Fan B, et al. (2006). Immunogenicity
tial conflicts of interest with respect to the authorship and/or and persistence of a targeted anti-caries DNA vaccine. J Dent Res
publication of this article. 85:915-918.
Copyright of Journal of Dental Research is the property of Sage Publications Inc. and its content may not be
copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written
permission. However, users may print, download, or email articles for individual use.

Вам также может понравиться