A Caries Vaccine?

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Caries Res 2004;38:230–235

DOI: 10.1159/000077759

A Caries Vaccine?
The State of the Science of Immunization against Dental Caries

Michael W. Russell a Noel K. Childers b Suzanne M. Michalek c


Daniel J. Smith d Martin A. Taubman d
a Departments of Oral Biology and Microbiology and Immunology, University at Buffalo, Buffalo, N.Y.,

Departments of b Oral Biology and c Microbiology, University of Alabama at Birmingham, Birmingham, Ala., and
d Department of Immunology, Forsyth Institute, Boston, Mass., USA

Key Words The concept of vaccination against dental caries has


Mutans streptococci W Salivary IgA antibodies W Vaccine existed almost from the time that this disease was recog-
antigen W Mucosal immunization nized to result from colonization of the teeth by acidogen-
ic bacteria, even though the etiological agents were origi-
nally thought to be lactobacilli. Since then, Streptococcus
Abstract mutans and Streptococcus sobrinus and their relatives,
Studies performed in numerous laboratories over sever- collectively known as mutans streptococci, have become
al decades have demonstrated the feasibility of immuniz- recognized as the principal organisms responsible for ini-
ing experimental rodents or primates with protein anti- tiating caries in humans [Loesche, 1986], and consider-
gens derived from Streptococcus mutans or Streptococ- able progress has been made in elucidating the factors
cus sobrinus against oral colonization by mutans strep- involved in their pathogenic activity, culminating recent-
tococci and the development of dental caries. Protec- ly in the sequencing of the entire S. mutans genome [Ajdic
tion has been attributed to salivary IgA antibodies which et al., 2002]. Likewise, enormous strides have been made
can inhibit sucrose-independent or sucrose-dependent in comprehending the workings of the mucosal immune
mechanisms of streptococcal accumulation on tooth sur- system by which secretory IgA (S-IgA) antibodies are gen-
faces according to the choice of vaccine antigen. Strate- erated in saliva and other secretions [Ogra et al., 1999].
gies of mucosal immunization have been developed to This system is functional in newborn infants, and al-
induce high levels of salivary antibodies that can persist though at birth salivary IgA levels are almost zero, infants
for prolonged periods and to establish immune memory. promptly develop salivary IgA antibodies concomitantly
Studies in humans show that salivary antibodies to mu- with oral microbial colonization [Smith and Taubman,
tans streptococci can be induced by similar approaches, 1992; Smith et al., 1998]. The mechanisms of action of
and that passively applied antibodies can also suppress salivary IgA antibodies against mutans streptococci in-
oral re-colonization by mutans streptococci. Progress clude interference with their sucrose-independent and
towards practical vaccine development requires evalua- sucrose-dependent attachment to, and accumulation on,
tion of candidate vaccines in clinical trials. Promising tooth surfaces, as well as possible inhibition of their meta-
strategies of passive immunization also require further bolic activities [Russell et al., 1999]. The goal of immuniz-
clinical evaluation. ing infants and young children against colonization by
Copyright © 2004 S. Karger AG, Basel mutans streptococci and hence diminishing the develop-

© 2004 S. Karger AG, Basel Michael W. Russell, PhD


ABC Department of Microbiology, Farber 138
Fax + 41 61 306 12 34 University at Buffalo, 3435 Main Street
E-Mail karger@karger.ch Accessible online at: Buffalo, NY 14214 (USA)
www.karger.com www.karger.com/cre Tel. +1 716 829 2790, Fax +1 716 829 2169, E-Mail russellm@buffalo.edu
ment of caries might be accomplished by applying new ing of protein antigens by recombinant DNA technology
strategies of mucosal vaccination that would induce sali- as well as the construction of synthetic peptides repre-
vary IgA antibodies without the complications of paren- senting identified antigenic epitopes have been pursued
teral injection. A large body of experimental work over [Jespersgaard et al., 1999; Smith et al., 2003; Takahashi et
several decades has demonstrated the feasibility of induc- al., 1991; Taubman et al., 1995; Zhang et al., 2002].
ing protective immunity against mutans streptococci and Numerous experiments in a variety of animal models
the subsequent development of dental caries in animal comprising rodents and primates have demonstrated the
models. Information has also accrued from several small- induction of salivary S-IgA and circulating IgG antibodies
scale trials in adult volunteers attesting to the applicabili- to mutans streptococcal antigens by oral or intranasal
ty of these approaches to humans. For other recent immunization with AgI/II, GTF or glucan-binding pro-
reviews of this subject, see Childers et al. [2002], Koga et teins [reviewed in Childers et al., 2002; Koga et al., 2002;
al. [2002], Russell et al. [1999], Russell [2001] and Smith Russell et al., 1999; Russell, 2001; Smith, 2002]. Upon
[2002]. subsequent oral challenge with virulent mutans strepto-
cocci and the institution of a high-sucrose diet, these mod-
els have further demonstrated reductions in colonization
Current Approaches and Findings in Active and diminished development of dental caries lesions.
Immunization Despite these successes, rodent models in particular have
limitations in predicting applicability of findings to the
Although over the years numerous surface or secreted human situation for a variety of reasons, including the
products of mutans streptococci have been proposed as short duration of the experiments compared with the time
vaccine antigen candidates, attention has become focused scale of caries development in humans. Thus, it is impor-
on three protein antigens: the surface fibrillar adhesins tant that the generation of salivary IgA antibodies by
known as AgI/II (synonyms: antigen B, P1, SpaP, PAc, immunization procedures developed in rodents has been
SpaA, PAg), the glucosyltransferases (GTF) and the glu- achieved in primates [Russell et al., 1996] and in human
can-binding proteins, all of which have demonstrable experiments (see below).
associations with virulence and the process of tooth sur- An important aspect of mucosal immunity centers
face colonization [Jenkinson and Lamont, 1997]. While around the question of immunological memory and the
some early efforts utilized parenteral injection which was recall of responses upon subsequent exposure to antigens.
successful in rodent and primate models [Lehner et al., Most studies of memory have focused on systemic anti-
1976; Russell et al., 1982] probably because of gingival body and cellular responses, and indeed earlier concepts,
transudation of circulating antibodies [Challacombe et especially those founded upon experiments using simple
al., 1978], most authorities have long recognized that methods of oral immunization with killed microorgan-
mucosal routes of immunization, designed to stimulate isms or purified protein antigens, held that memory was
the common mucosal immune system and induce potent poorly developed in the mucosal immune system. More
salivary S-IgA antibodies, will not only be more effica- effective strategies of mucosal immunization, especially
cious but also be more acceptable and circumvent some those exploiting the extraordinary immunogenicity and
concerns over safety. This and other vaccine goals have adjuvanticity of cholera and related enterotoxins, how-
driven the development of novel strategies for effectively ever, have shown that memory can be induced and
stimulating mucosal immune responses [Russell, 2003]. recalled by mucosal immunization [Harrod et al., 2001;
Several of these have been applied to mutans streptococ- Vajdy and Lycke, 1993]. While many details of the cellu-
cal antigens, including the delivery of immunogens in lar and regulatory mechanisms underlying this remain to
liposomes and other microparticles, co-administration of be elucidated, this finding has important implications for
mucosal adjuvants such as enterobacterial enterotoxins the development of vaccines against many mucosal infec-
and their detoxified mutants, coupling of immunogens to tions including caries. Particularly in this case, it may be
the nontoxic B subunits of enterotoxins and the expres- desirable that a salivary antibody response should be
sion of mutans streptococcal antigens in attenuated Sal- induced and sustained throughout the ‘window of infec-
monella strains [Eastcott et al., 2002; Hajishengallis et al., tivity’, the period from approximately 18 to 32 months of
1995; Harokopakis et al., 1997; Huang et al., 2001; Mar- age when infants are most likely to become infected with
tin et al., 2000; Michalek et al., 1992; Russell and Wu, mutans streptococci [Caufield et al., 1993]. It may also be
1991; Smith et al., 2000]. In addition, molecular engineer- desirable that responses should be recallable either by

A Caries Vaccine? Caries Res 2004;38:230–235 231


booster immunization or by natural exposure to mutans fere with mutans streptococcal colonization (table 2). Hu-
streptococci, if further opportunities for infection arise at man volunteers immunized orally with S. sobrinus GTF
later times, such as when children enter school or their packaged in enteric capsules (14 young adults, compared
permanent teeth erupt. Thus, we have found that salivary with 11 placebo controls) developed increased levels of
IgA responses to AgI/II induced by mucosal immuniza- parotid salivary IgA antibodies to GTF and showed
tion with AgI/II coupled to cholera toxin B subunit or delayed reaccumulation of mutans streptococci in their
expressed in recombinant Salmonella can persist for up to oral microbiota [Smith and Taubman, 1987]. In a further
1 year in mice (i.e. for half their normal life-span; table 1) study on 23 young adults, topical application of GTF to
and are amenable to prompt recall by booster immuniza- the lower lip intended to stimulate local antibody produc-
tion even after 2 years [Hajishengallis et al., 1996; Haro- tion in the minor salivary glands also delayed oral re-
kopakis et al., 1997; Harrod et al., 2001; Russell and Wu, colonization with mutans streptococci although antibody
1991; Wu et al., 2000]. levels were not significantly increased [Smith and Taub-
man, 1990]. Oral immunization with preparations of S.
mutans GTF that also contained a truncated form of AgI/
Human Trials II in enteric capsules was also successful in elevating sali-
vary IgA antibodies to the antigen preparation [Childers
Several small-scale human trials in adults have shown et al., 1994]. When similar antigen preparations were
that it is feasible to increase levels of salivary S-IgA anti- administered intranasally or by topical application to the
bodies to mutans streptococci, and in some cases to inter- tonsils, either in soluble form or incorporated in lipo-
somes, salivary IgA antibodies were likewise increased
[Childers et al., 1997, 1999, 2002, 2003; Li et al., 2003].
Table 1. Persistence of serum and salivary antibodies to AgI/II in
These studies now need to be extended into progressively
mice after intranasal immunization with AgI/II conjugated to cholera younger age groups in controlled trials aimed at estab-
toxin B subunit lishing whether equivalent responses can be induced in
children and whether the responses obtained can suppress
Time after Serum Saliva oral colonization by mutans streptococci.
immunization
IgG, Ìg/ml IgA, Ìg/ml IgA (Ab/Ig), %

Before 0.25 1.42 0 Passive Immunization – An Alternative


!/&2.31 !/&1.38
Approach
4 months 571 43.1 60.5
!/&1.84 !/&1.50 !/&1.41
8 months 175 16.1 13.6 An alternative approach lies in the development of
!/&1.58 !/&1.69 !/&1.76 antibodies suitable for passive oral application against
12 months 136 24.2 21.6 dental caries. This has considerable potential advantage
!/&1.83 !/&1.40 !/&1.54
in that it completely avoids any risks that might arise
Geometric mean !/& SD, n = 5; from Wu et al. [2000]. from active immunization. Conversely, in the absence of
any active response on the part of the recipient, there is no

Table 2. Trials in adult humans: active immunization with S. mutans protein antigens

Antigen Route n Predominant antibody response (protective effect) Reference

GTF oral 25 increased salivary IgA antibody Smith and Taubman [1987]
(delayed reaccumulation of indigenous S. mutans)
topical (MSG) 23 (delayed reaccumulation of indigenous S. mutans) Smith and Taubman [1990]
GTF oral 7 increased salivary IgA2 antibody (n.t.) Childers et al. [1994]
(+ AgI/II) nasal 5, 21 increased nasal IgA1, salivary IgA1 and IgA2 antibodies (n.t.) Childers et al. [1997, 1999]
nasal or tonsillar (topical) 21 IgA1 nasal and salivary antibodies in nasal group (n.t.) Childers et al. [2002]
nasal 12 salivary IgA1 antibodies (n.t.) Li et al. [2003]
nasal 26 IgA1 nasal and salivary antibodies (n.t.) Childers et al. [2003]

MSG = Minor salivary glands ; n.t. = not tested.

232 Caries Res 2004;38:230–235 Russell/Childers/Michalek/Smith/Taubman


induction of immunological memory, and the adminis- maintain protection over a prolonged time remains a
tered antibodies can persist in the mouth for only a few major challenge. Although new technologies for antibody
hours at most or up to 3 days in plaque [Ma et al., 1990]. engineering and production in animals or especially in
Strategies include the development of antibodies to mu- plants (‘plantibodies’) offer the prospect of reducing the
tans streptococcal antigens in cow’s milk and hen’s eggs costs sufficiently to enable these materials to be incorpo-
and the genetic engineering of human-like S-IgA anti- rated into products for daily use, such as mouthwashes
bodies in plants [Hamada et al., 1991; Hatta et al., 1997; and dentifrices, long-term efficacy has yet to be reliably
Loimaranta et al., 1998; Ma et al., 1995; Mitoma et al., demonstrated.
2002]. Animal experiments have been encouraging: for
example, the administration of chicken egg IgY anti-
bodies to glucan-binding proteins diminished the devel- Future Prospects and Potential Impact
opment of caries lesions in a rat model [Smith et al.,
2001]. Mouse monoclonal antibodies to AgI/II applied Given that dental caries usually develops slowly and
topically inhibited oral colonization by mutans strepto- can occur throughout life, it may be anticipated that
cocci and development of caries in monkeys for at least 1 immune protection would need to be similarly long-last-
year [Lehner et al., 1985]. Similar treatment, after exten- ing. Thus, the duration and anamnestic recall of salivary
sive oral prophylaxis, of a small number of human adult antibody responses are important factors. While it is now
volunteers with this IgG, or with engineered ‘human’ S- clear that mucosal immune responses can persist and that
IgA antibodies derived from the same monoclonal anti- memory is established if the priming stimulus is suffi-
body, also suppressed the re-emergence of mutans strep- cient, relatively little is known about the parameters that
tococci for up to 2 years or 4 months, respectively [Ma et govern memory in the mucosal immune system. The
al., 1990, 1998]. The plausible though unproven explana- characteristics of specific mucosal memory cells, their
tion offered for these findings was that once mutans strep- location, and how they can be recalled and directed to par-
tococci had been displaced by prophylaxis, passive appli- ticular effector sites such as the salivary glands to produce
cation of antibody prevented their immediate re-coloni- IgA antibodies for transport into the secretion are impor-
zation so that their oral ‘niche’ became occupied by other tant subjects for investigation. Although current under-
species with the result that their re-emergence was sup- standing holds that oral colonization with mutans strepto-
pressed for far longer than the antibody persisted in the cocci mainly occurs during a ‘window of infectivity’ at
mouth. Unfortunately, further experiments on larger around 2 years of age after primary teeth begin to erupt, it
numbers of adults have not consistently demonstrated is unclear whether further opportunities for colonization
equivalent long-term reductions in colonization [Wein- exist, for example when children enter school and mix
traub et al., 2001]. Whether a similar application of anti- socially with a much larger group of their peers, or when
bodies to young infants might inhibit subsequent oral the permanent teeth erupt. Two corollaries arise from
colonization by mutans streptococci remains to be deter- such considerations: (i) that it would be necessary to
mined. However, in spite of these disappointments, col- immunize infants or young children in order to provide
lectively these studies clearly demonstrate the potential of immune protection prior to initial colonization with mu-
antibodies to interfere with the ability of mutans strepto- tans streptococci; (ii) that booster immunization to recall
cocci to colonize teeth and to inhibit caries development. responses might be desirable to forestall colonization at
The key question then becomes: how can such anti- later time points. As the transmission of mutans strepto-
bodies be effectively delivered orally in caries-susceptible cocci appears to be primarily from mother to infant [Li
individuals and maintained at a protective level for the and Caufield, 1995], a third possibility is that young
required length of time? Active vaccination has the ad- mothers might be immunized actively or passively with
vantage of inducing the endogenous production of sali- the objective of reducing their oral load of mutans strepto-
vary antibodies and the establishment of immune memo- cocci (possibly in combination with conventional prophy-
ry but requires a commitment to performing the human laxis or other interventions), thereby diminishing the
trials necessary to establish safety and efficacy. Passive probability and extent of transmission to their infants. If
administration of preformed exogenous antibodies offers the transferred bacteria are coated with maternal salivary
the advantage of evading risks, however small, that are antibodies, this would likely reduce their capacity to
inherent in any active immunization procedure, but the colonize the infant’s mouth. It has been suggested that
need to provide a continuous source of antibodies to immunization of young mothers to induce the generation

A Caries Vaccine? Caries Res 2004;38:230–235 233


of antibodies to mutans streptococcci in breast-milk could achieve a sufficiently high prevalence of immunity in a
be exploited to provide passive immunity against caries to population that the chain of transmission is broken and
their infants. However, it seems unlikely that this strategy the pathogen cannot sustain itself in the community.
would have significant impact at least in Western socie- However, the biology of caries is different from that of
ties, where breast-feeding, if given, usually terminates acute infections, and as with other modalities of interven-
well before the ‘window of infectivity’ for mutans strepto- tion, it is conceivable that immunization will not attain
cocci opens. complete effectiveness. Nevertheless, efficacy as low as
Regardless of the mechanism by which immune pro- 50% could have significant impact on the burden of dis-
tection against dental caries is achieved, further advances ease, and the social and economic costs associated with it.
to make immunization against caries practicable will Given that the bulk of dental caries occurs among a high-
depend upon clinical trials aimed at establishing whether risk sector of the population (at least in the USA), target-
the findings from animal experiments can be transferred ing an effective vaccine to such individuals would in-
to humans. Particular goals for such studies include deter- crease its impact.
mining whether appropriate immune responses can be
safely generated in humans, especially in the susceptible
age groups, and whether such responses will afford desir- Acknowledgements
able levels of protection.
The authors’ studies have been supported by USPHS grants
The goals for vaccination against most other, mainly DE06746, DE09846, DE07026, DE08182, DE09081, DE04733 and
acute, infectious diseases are usually to provide near-com- DE06153 from the National Institute of Dental and Craniofacial
plete protection of the individual against infection, and to Research.

References

Ajdic D, McShan WM, McLaughlin RE, Savic G, Childers NK, Tong G, Mitchell S, Kirk K, Russell Harokopakis E, Hajishengallis G, Greenway TE,
Chang J, Carson MB, Primeaux C, Tian RY, MW, Michalek SM: A controlled clinical study Russell MW, Michalek SM: Mucosal immuno-
Kenton S, Jia HG, Lin SP, Qian YD, Li SL, of the effect of nasal immunization with a genicity of a recombinant Salmonella typhimu-
Zhu H, Najar F, Lai HS, White J, Roe BA, Fer- Streptococcus mutans antigen alone or incorpo- rium-cloned heterologous antigen in the ab-
retti JJ: Genome sequence of Streptococcus mu- rated into liposomes on induction of immune sence or presence of co-expressed cholera toxin
tans UA159, a cariogenic dental pathogen. responses. Infect Immun 1999;67:618–623. A2/B subunits. Infect Immun 1997;65:1445–
Proc Natl Acad Sci USA 2002;99:14434– Childers NK, Zhang SS, Michalek SM: Oral immu- 1454.
14439. nization of humans with dehydrated liposomes Harrod T, Martin M, Russell MW: Long-term per-
Caufield P, Cutter G, Dasanayake A: Initial acqui- containing Streptococcus mutans glucosyltrans- sistence and recall of immune responses in
sition of mutans streptococci by infants: Evi- ferase induces salivary immunoglobulin A2 an- aged mice after mucosal immunization. Oral
dence for a discrete window of infectivity. J tibody responses. Oral Microbiol Immunol Microbiol Immunol 2001;16:170–177.
Dent Res 1993;72:37–45. 1994;9:146–153. Hatta H, Tsuda K, Ozeki M, Kim M, Yamamoto T,
Challacombe SJ, Russell MW, Hawkes JE, Berg- Eastcott JW, Orr N, Smith DJ, Hayden TL, Taub- Otake S, Hirasawa M, Katz J, Childers NK,
meier LA, Lehner T: Passage of immunoglobu- man MA: Expression and delivery of GTF pep- Michalek SM: Passive immunization against
lins from plasma to the oral cavity in rhesus tides in Salmonella enterica (abstract 3886). J dental plaque formation in humans: Effect of a
monkeys. Immunology 1978;35:923–931. Dent Res 2002;81(special issue A). mouth rinse containing egg yolk antibodies
Childers NK, Li F, Kirk K, Dasanayake AP, Kim Hajishengallis G, Hollingshead SK, Koga T, Rus- (IgY) specific to Streptococcus mutans. Caries
J-G, Michalek SM: Nasal but not tonsillar im- sell MW: Mucosal immunization with a bacte- Res 1997;31:268–274.
munization of humans with Streptococcus mu- rial protein antigen genetically coupled to cho- Huang Y, Hajishengallis G, Michalek SM: Induc-
tans antigens primes for responses 2 years after lera toxin A2/B subunits. J Immunol 1995;154: tion of protective immunity against Streptococ-
an initial immunization (abstract 1721). J Dent 4322–4332. cus mutans colonization after mucosal immu-
Res 2003;82(special issue). Hajishengallis G, Michalek SM, Russell MW: Per- nization with attenuated Salmonella enterica
Childers NK, Tong G, Li F, Dasanayake AP, Kirk sistence of serum and salivary antibody re- serovar typhimurium expressing an S. mutans
K, Michalek SM: Humans immunized with sponses after oral immunization with a bacte- adhesin under the control of in vivo-inducible
Streptococcus mutans antigens by mucosal rial protein antigen genetically linked to the nirB promoter. Infect Immun 2001;69:2154–
routes. J Dent Res 2002;81:48–52. A2/B subunits of cholera toxin. Infect Immun 2161.
Childers NK, Tong G, Michalek SM: Nasal immu- 1996;64:665–667. Jenkinson HF, Lamont RJ: Streptococcal adhesion
nization of humans with dehydrated liposomes Hamada S, Horikoshi T, Minami T, Kawabata S, and colonization. Crit Rev Oral Biol Med
containing Streptococcus mutans antigen. Oral Hiraoka J, Fujiwara T, Ooshima T: Oral pas- 1997;8:175–200.
Microbiol Immunol 1997;12:329–335. sive immunization against dental caries in rats
by use of hen egg yolk antibodies specific for
cell-associated glucosyltransferase of Strepto-
coccus mutans. Infect Immun 1991;59:4161–
4167.

234 Caries Res 2004;38:230–235 Russell/Childers/Michalek/Smith/Taubman


Jespersgaard C, Hajishengallis G, Huang Y, Russell Michalek SM, Childers NK, Katz J, Dertzbaugh M, Smith DJ, Taubman MA: Oral immunization of
MW, Smith DJ, Michalek SM: Protective im- Zhang S, Russell MW, Macrina FL, Jackson S, humans with Streptococcus sobrinus glucosyl-
munity against Streptococcus mutans infection Mestecky J: Liposomes and conjugate vaccines transferase. Infect Immun 1987;55:2562–
in mice after intranasal immunization with for antigen delivery and induction of mucosal 2569.
the glucan-binding region of S. mutans gluco- immune responses. Adv Exp Med Biol 1992; Smith DJ, Taubman MA: Effect of local deposition
syltransferase. Infect Immun 1999;67:6543– 327:191–198. of antigen on salivary immune responses and
6549. Mitoma M, Oho T, Michibata N, Okano K, Naka- reaccumulation of mutans streptococci. J Clin
Koga T, Oho T, Shimazaki Y, Nakano Y: Immuni- no Y, Fukuyama M, Koga T: Passive immuni- Immunol 1990;10:273–281.
zation against dental caries. Vaccine 2002;20: zation with bovine milk containing antibodies Smith DJ, Taubman MA: Ontogeny of immunity
2027–2044. to a cell surface protein antigen-glucosyltrans- to oral microbiota in humans. Crit Rev Oral
Lehner T, Caldwell J, Smith R: Local passive im- ferase fusion protein protects rats against den- Biol Med 1992;3:109–133.
munization by monoclonal antibodies against tal caries. Infect Immun 2002;70:2721–2724. Smith DJ, Trantolo DJ, King WF, Gusek EJ, Fackl-
streptococcal antigen I/II in the prevention of Ogra PL, Mestecky J, Lamm ME, Strober W, Bie- er PH, Gresser JD, De Souza VL, Wise DL:
dental caries. Infect Immun 1985;50:796–799. nenstock J, McGhee JR (eds): Mucosal Immu- Induction of secretory immunity with bioadhe-
Lehner T, Challacombe SJ, Caldwell J: Immuno- nology. San Diego, Academic Press, 1999. sive poly(D,L-lactide-co-glycolide) microparti-
logical basis for vaccination against dental car- Russell MW: Potential for vaccines in the pre- cles containing Streptococcus sobrinus glucosyl-
ies in rhesus monkeys. J Dent Res 1976;55(spe- vention of caries lesions. Oper Dent 2001;suppl transferase. Oral Microbiol Immunol 2000;15:
cial issue C):166. 6:51–60. 124–130.
Li F, Michalek SM, Dasanayake AP, Li Y, Kirk K, Russell MW: Mucosal immunity; in Ellis RW, Takahashi I, Okahashi N, Matsushita K, Tokuda
Childers NK: Intranasal immunization of hu- Brodeur BR (eds): New Bacterial Vaccines. M, Kanamoto T, Munekata E, Russell MW,
mans with Streptococcus mutans antigens: Low Georgetown, Landes Bioscience, 2003. Koga T: Immunogenicity and protective effect
dose differentiates responses to soluble versus Russell MW, Hajishengallis G, Childers NK, Mi- against oral colonization by Streptococcus mu-
liposomal antigens. Oral Microbiol Immunol chalek SM: Secretory immunity in defense tans of synthetic peptides of a streptococcal
2003;18:271–277. against cariogenic mutans streptococci. Caries surface protein antigen. J Immunol 1991;146:
Li Y, Caufield PW: The fidelity of initial acquisi- Res 1999;33:4–15. 332–336.
tion of mutans streptococci by infants from Russell MW, Moldoveanu Z, White PL, Sibert GJ, Taubman MA, Holmberg CJ, Smith DJ: Immuni-
their mothers. J Dent Res 1995;74:681–685. Mestecky J, Michalek SM: Salivary, nasal, ge- zation of rats with synthetic peptide constructs
Loesche WJ: Role of Streptococcus mutans in hu- nital, and systemic antibody responses in mon- from the glucan-binding or catalytic region of
man dental decay. Microbiol Rev 1986;50: keys immunized intranasally with a bacterial mutans streptococcal glucosyltransferase pro-
353–380. protein antigen and the cholera toxin B sub- tects against dental caries. Infect Immun 1995;
Loimaranta V, Carlén A, Olsson J, Tenovuo J, unit. Infect Immun 1996;64:1272–1283. 63:3088–3093.
Syväoja EL, Korhonen H: Concentrated bo- Russell MW, Wu H-Y: Distribution, persistence, Vajdy M, Lycke N: Stimulation of antigen-specific
vine colostral whey proteins from Streptococ- and recall of serum and salivary antibody re- T- and B-cell memory in local as well as sys-
cus mutans/Strep. sobrinus immunized cows sponses to peroral immunization with protein temic lymphoid tissues following oral immuni-
inhibit the adherence of Strep. mutans and pro- antigen I/II of Streptococcus mutans coupled to zation with cholera toxin adjuvant. Immunolo-
mote the aggregation of mutans streptococci. J the cholera toxin B subunit. Infect Immun gy 1993;80:197–203.
Dairy Res 1998;65:599–607. 1991;59:4061–4070. Weintraub JA, Hilton JF, White JM, Hoover C,
Ma JKC, Hiatt A, Hein M, Vine ND, Wang F, Sta- Russell RRB, Beighton D, Cohen B: Immunization Pelino JE, Tran K, Wycoff K, Larrick JW, Yu
bila P, Van Dolleweerd C, Mostov K, Lehner of monkeys (Macaca fascicularis) with antigens L, Featherstone JDB: Results of a plant-de-
T: Generation and assembly of secretory anti- purified from Streptococcus mutans. Br Dent J rived mutans streptococci antibody clinical
bodies in plants. Science 1995;268:716–719. 1982;152:81–84. trial (abstract 201). J Dent Res 2001;80(special
Ma JKC, Hikmat BY, Wycoff K, Vine ND, Char- Smith DJ: Dental caries vaccines: Prospects and issue).
gelegue D, Yu L, Hein MB, Lehner T: Charac- concerns. Crit Rev Oral Biol Med 2002;13: Wu HY, Abdu S, Stinson D, Russell MW: Genera-
terization of a recombinant plant monoclonal 335–349. tion of female genital tract antibody responses
secretory antibody and preventive immuno- Smith DJ, King WF, Akita H, Taubman MA: Asso- by local or central (common) mucosal immuni-
therapy in humans. Nat Med 1998;4:601–606. ciation of salivary immunoglobulin A antibody zation. Infect Immun 2000;68:5539–5545.
Ma JKC, Hunjan M, Smith R, Kelly C, Lehner T: and initial mutans streptococcal infection. Oral Zhang P, Jespersgaard C, Lamberty-Mallory L,
An investigation into the mechanism of protec- Microbiol Immunol 1998;13:278–285. Katz J, Huang Y, Hajishengallis G, Michalek
tion by local passive immunization with mono- Smith DJ, King WF, Barnes LA, Peacock Z, Taub- SM: Enhanced immunogenicity of a genetic
clonal antibodies against Streptococcus mutans. man MA: Immunogenicity and protective im- chimeric protein consisting of two virulence
Infect Immun 1990;58:3407–3414. munity induced by synthetic peptides associat- antigens of Streptococcus mutans and protec-
Martin MH, Metzger DJ, Michalek SM, Connell ed with putative immunodominant regions of tion against infection. Infect Immun 2002;70:
TD, Russell MW: Comparative analysis of the Streptococcus mutans glucan-binding protein 6779–6787.
mucosal adjuvanticity of the type II heat-labile B. Infect Immun 2003;71:1179–1184.
enterotoxins, LT-IIa and LT-IIb. Infect Immun Smith DJ, King WF, Godiska R: Passive transfer of
2000;68:281–287. immunoglobulin Y antibody to Streptococcus
mutans glucan binding protein B can confer
protection against experimental dental caries.
Infect Immun 2001;69:3135–3142.

A Caries Vaccine? Caries Res 2004;38:230–235 235


Copyright: S. Karger AG, Basel 2004. Reproduced with the permission of S. Karger AG, Basel. Further
reproduction or distribution (electronic or otherwise) is prohibited without permission from the copyright
holder.

You might also like