Sie sind auf Seite 1von 28

Accepted Article

PROF. MRUDULA PATEL (Orcid ID : 0000-0002-8406-118X)

Article type : Review Article

Dental caries vaccine: Are we there yet?

*Patel M

Department of Oral Biological Sciences, School of Oral Health Sciences, Faculty of Health
Sciences, University of the Witwatersrand, Johannesburg, South Africa.

*Corresponding Author:

Mrudula Patel

Department of Oral Biological Sciences

Faculty of Health Sciences, University of the Witwatersrand

Private Bag 3, Wits

Johannesburg, 2050, South Africa.

E mail: Mrudula.patel@wits.ac.za

Telephone: +27 11 717 2110

Fax: +27 0865533020

Running title: Dental caries vaccine

This article has been accepted for publication and undergone full peer review but has not been through the
copyediting, typesetting, pagination and proofreading process, which may lead to differences between this
version and the Version of Record. Please cite this article as doi: 10.1111/lam.13218

This article is protected by copyright. All rights reserved


Accepted Article
Significance and Impact of study

Dental caries is an irreversible, multifactorial opportunistic infection. The treatment is costly,


making it a public health problem. Despite many years of promising laboratory research, animal
studies and clinical trials, there is no commercially available vaccine today. The research
objectives have become more refined from lessons learnt over the years. Multigenic
DNA/recombinant vaccines, using the best proved adjuvants with a delivery system for the nasal
or sublingual route, should be developed and researched with multicentre collaborative efforts. In
addition, new vaccine targets can be identified. To overcome the economic hurdle, funders and
public health interest should be stimulated.

Abstract

Dental caries, caused by Streptococcus mutans, is a common infection. Caries vaccine has been
under investigation for the last 40 years. Many in vitro and in vivo studies and some human
clinical trials have determined many pertinent aspects regarding vaccine development. The
virulence determinants of S. mutans, such as Ag I/II, responsible for adherence to surfaces,
glucosyltransferase (GTF), responsible for the production of glucan, and the glucan binding
protein (GBP), responsible for the attachment of glucan to surfaces, have been known to elicit an
antigen-specific immune response. It is also known that more than one antigen or a functional part
of the genome responsible for these virulence determinants provide a better host response
compared with the monogenic vaccine or complete genome of a specific antigen. To enhance the
host response, the use of adjuvants has been studied and the routes of antigen administration have
been investigated. In recent years, some promising vaccines such as pGJA-P/VAX, LT
derivative/Pi39-512, KFD2-rPAc and SBR/GBR-CMV-nirB have been developed and tested in
animals. New virulence targets need to be explored. Multicentre collaborative studies and human

This article is protected by copyright. All rights reserved


clinical trials are required and some interest from funders and public health experts should be
Accepted Article
generated to overcome this hurdle.

Keywords: Vaccine; Caries; Ag I/II; Glucan binding protein; Glucosyl transferase; PstS

This article is protected by copyright. All rights reserved


Introduction
Accepted Article
Dental caries, a multifactorial disease, is a very prevalent oral disease affecting 60-90% of
children in industrialised countries and 100% of the adult population worldwide (Petersen et al
2005). It is not a life-threatening disease but has a major impact on people’s daily lives and well-
being due to the pain and problems with eating, chewing and smiling. In addition, it restricts
activities at school and work causing millions of hours of absenteeism. Treatment is expensive and
has, therefore, become a public health problem (Petersen et al. 2005). Immunotherapy with caries
vaccines has also been explored because vaccines are good options for public health applications,
especially when there is a lack of a universally accessed healthcare system. In the development of
caries vaccines, the target has been the cariogenic bacteria Streptococcus mutans and S. sobrinus
(Smith DJ 2002).

Virulence properties of S. mutans and the targets for vaccine development

Both cariogenic bacteria are equally virulent with regard to dental caries (Conrads et al. 2014).
However, S. mutans is more frequently isolated from the oral cavity (Loesche 1986) and it is first
to adhere to teeth through adhesins such as Ag I/II. It also produces extracellular polysaccharides,
called glucan, from dietary sucrose through glucosyltransferases (GTFs). Glucan acts as a nutrient
and as an adherence moiety through glucan binding protein (GBP). It also has the ability to absorb
sugar rapidly through the phosphoenole pyruvate-mediated phosphotransferase sugar uptake
system (PEP-PTS) and produce cariogenic lactic acid through the glycolytic pathway. The
properties such as acidogenicity (producing acids) and aciduricity (tolerating acid) of S. mutans
allow them to continue with growth and metabolism, even at the cariogenic pH of 5.5 and below.
At this pH, demineralisation of enamel can occur and cause dental caries.

In the last 40 years, vaccines have been developed targeting the virulence antigens of S. mutans
(Robinette et al. 2011; Xu et al. 2005; Zhang et al. 2007; Ma et al. 1990). The most researched
antigens are adhesion Ag I/II, GTFs, and GBPs (Figure 1). Fewer studies have researched enolase
(Dinis et al. 2009; 2011). In recent years, a vaccine has been developed using phosphate-binding-
protein (Pst system) which also facilitates adhesion to abiotic surfaces (Luz et al. 2012, Ferreira et
al. 2016). All these proteins have antigenic properties and can, therefore, elicit specific antibodies

This article is protected by copyright. All rights reserved


which are known to block the particular function of the virulence factors (Lehner et al. 1980;
Accepted Article
Smith & Lehner 1981). Vaccines synthesised using these antigenic virulence factors can reduce
the number of S. mutans in the plaque and hence reduce the chances of developing dental caries.
In vitro and in vivo (animal and human) studies have established that these antibodies can raise the
levels of antigen-specific secretory IgA and reduce the number of S. mutans in dental plaque
(Koga et al. 2002). Secretory IgA can act against dental caries by blocking colonisation of S.
mutans on the tooth surface. Immunoglobulins G and M are less involved in the defence against
caries.

In the early years of research, vaccines generated using whole cells of either S. mutans or S.
sobrinus were developed. However, in vivo and in vitro studies revealed minimum benefits with
some side effects (McGhee et al. 1975; Talbman & Smith 1974; Hajishengallis and Michalek
1999; Ferretti et al. 1980). These studies will not be discussed here.

Passive immunisation

In passive immunisation, ready-made antibodies are administered which circulate in the body and
impart specific protection. For a caries vaccine, monoclonal antibodies specific to the target
antigen of S. mutans in vivo and in transgenic tobacco plants have been developed. Studies have
also used bovine milk and whey generated from immunised cows to control S. mutans. Mitoma et
al. (2002) developed antibodies against a surface antigen and GTF that significantly reduced the
colonisation of S. mutans when mixed in milk fed daily to rats. Monoclonal antibodies against Ag
I/II prepared in tobacco plants prevented the biofilm formation in humans; however, adequate
titres of antibodies could not be maintained in the oral cavity (Ma et al. 1990, 1998). Safe, passive
vaccines are less effective and require large quantities of repeated applications; active
immunisation has, therefore, become the major research focus.

Active immunisation

Active immunisation is much more effective and long-lasting due to the involvement of host
response. In the development of a caries vaccine, many studies have been based on active
immunisation using target antigens. Major cell surface adhesins such as Ag I/II (or PAc or P1)
from S. mutans and SpaA (or PAg) from S. sobrinus have been identified and characterised. In
vitro and in vivo studies have shown that antibodies developed against these adhesins prevent the

This article is protected by copyright. All rights reserved


adherence of S. mutans to the saliva coated tooth. Immunisation with intact Ag I/II can protect
Accepted Article
rodents, primates or humans from dental caries caused by S. mutans (Lehner et al. 1981).
Similarly, immunisation with S. sobrinus SpaA protected rats from caries caused by S. sobrinus
(Redman et al. 1996). Animal studies have shown that antibodies produced against GTF can
interfere with plaque development (Smith & Taubman 1990). Subsequently, these authors showed
that oral and topical GTF vaccine administration in humans after a thorough oral hygiene,
interfered with the establishment of resident flora due to the induction of SIgA (Smith & Taubman
1987; 1990). Glucan binding protein B has also been shown to generate an immune response and
protect against experimental caries if applied through either subcutaneous or salivary gland
injections or by an intranasal route (Smith & Taubman 1996; Smith et al. 1997).

Subunit vaccines

Subunit vaccines containing the functional part of the genome responsible for the production of
Ag I/II or GTFs or GBP have also been constructed and studied. These vaccines contain single or
multiple copies (multivalent) of functional epitopes associated with either of the virulence proteins
(monomeric) or in combinations (dimeric) (Cao et al. 2016; Oishi et al. 2001). These subunit
vaccines can target multiple virulence properties, have enhanced the production of desired
antibodies and eliminated some of the unwanted immune responses. For example, N-terminal
saliva-binding region (SBR) on S. mutans surface antigen I/II is important in the initial adherence
of this organism to the tooth surface. When the vaccine containing SBR with a C-terminal
structural region of Ag I/II, Ag II and whole Ag I/II were administered to rats intranasally , they
induced salivary IgA anti-Ag I/II antibodies. However, protection against caries was better with
SBR compared to Ag II (Hajishengallis et al. 1998). Similarly, vaccines containing a 22-mer
sequence from the catalytic domain of GTF showed an immunogenic response in rats and
protection against S. mutans and S. sobrinus infection (Smith et al. 2005).

Synthetic vaccines

Synthetic vaccines are constructed with synthetically produced functional region target antigens.
For example, subcutaneous immunisation with a synthetic peptide derived from the alanine-rich
region of Ag I/II from S. mutans, induced a high level of IgG antibody (Takahashi et al. 1991).

This article is protected by copyright. All rights reserved


Fusion proteins containing saliva-binding alanine-rich region (PAcA) of adhesion PAc with the
Accepted Article
glucan binding (GB) domain of GTF-I, an enzyme catalysing the synthesis of water-insoluble
glucan from sucrose, also inhibited sucrose-dependent adhesion of S. mutans to a hydroxyapatite
surface (Yu et al. 1997).

A vaccine was developed by Ferreira et al. (2016) using PstS protein. These authors used E. coli to
clone and express the pstS gene and purified recombinant PstS immunogenic protein. When PstS
protein was applied to mice sublingually, the results showed a significant increase in serum anti-
PstS IgG, particularly with the adjuvant. An in vitro study on sera collected from the animals
prevented the adherence of S. mutans and in the oral cavities of animals when vaccinated and
thereafter challenged with S. mutans.

Recombinant and DNA vaccines

These DNA vaccines have become the trend in caries vaccine research because they are safe and
the expression of antigenic protein is stable with stronger antigenicity (Table 1). Recombinant
vaccines are prepared by isolating the functional genome/s responsible for the target antigens and
linking them to vectors such as attenuated S. typhimurium, S. typhi, E. coli or plasmid. This
recombinant mutant vector would produce the respective proteins (chimeric) or, on application,
would elicit an immune response (DNA vaccine). For example, oral immunisation with
recombinant S. typhimurium expressing SpaA of S. sobrinus, was able to induce a persistent
mucosal immune response protecting rats against cariogenic S. sobrinus (Redman et al. 1994;
1995). Jespersgaard et al. (1999) immunised mice with an E. coli- expressed recombinant GTF-
glucan-binding domain sequence and the chimeric protein, and both conferred protection against
experimental S. mutans infection and resulting caries. Recombinant plasmid pCIA-P construct
containing A-P region of pac gene when administered to gnotobiotic rats, showed high levels of
PAc-specific salivary IgA and serum IgG antibody (Fan et al. 2002). These investigators also
showed a two-fold improvement in PAc-specific IgG antibody responses in serum with pCIA-
P/bupivacaine and reduction in the development of carious lesions (Jia et al. 2004). This vaccine
was subsequently improved by adding the glucan binding domain of S. mutans gtfB gene, the
signal peptide and extracellular regions of human CTLA4 gene and the hinge and Fc regions of
human Iggamma1 gene (pGJA-P), showing increased serum and salivary antibody response and
fewer caries (Xu et al. 2005).

This article is protected by copyright. All rights reserved


Research has also been conducted to improve the efficacy and the delivery of caries vaccines
Accepted Article
using nanotechnology. Cao et al. (2017) successfully generated self-assembling nanoparticles by
linking the Glucan-binding region of glucosyltransferase (GLU) to the N-terminal domain of
ferritin (GLU-FTH) which significantly increased the level of GLU-specific antibodies and
produced lower caries scores compared to GLU in mice. Similar results were found by Li et al.
(2016) using trimethyl chitosan nanoparticles. Their pVAX1-wapA/trimethyl chitosan vaccine
elicited greater immune response and the rats had fewer carious lesions compared to the animals
immunised with naked pVAX1-wapA.

Routes of application and adjuvants

A major immune constituent of the major and minor salivary glands is salivary secretory IgA
which can be induced by the mucosal application of a caries vaccine. Intranasal, tonsillar, oral and
rectal routes of caries vaccine delivery have also been explored. Topical application to the
intranasal, tonsillar regions and lower lip area has shown some promising results. The intranasal
application of S. mutans Ag I/II and GBPB vaccines has shown protection against S. mutans
(Smith et al. 1997b). In rabbits, topical application of formalin-killed S. sobrinus cells in the
tonsillar region elicited an immune response which significantly reduced the development of
caries (Fukuizumi et al. 1999). In one study, the indigenous Streptococcal flora was reduced in
young human adults after the application of GTF vaccine onto the lower lip when compared with a
placebo group (Smith & Taubman 1990). These mucosal route vaccines often require adjuvants to
enhance the immune response. Adjuvants accelerate, prolong or enhance antigen-specific immune
responses when used in combination with specific vaccine antigens. As adjuvants, the use of
enterotoxins from E. coli, V. cholera and S. typhimurium have been successfully investigated in
caries vaccines (Zhao et al. 2011; Saito et al. 2001; Hajishengallis et al. 1998; Batista et al. 2017).
However, toxin-related adjuvants may cause adverse effects on intranasal administration, therefore
non-toxic derivatives of the labile-toxin (LTK4R) produced by enterotoxigenic E. coli have been
investigated with promising results (Batista et al. 2017).

Lessons learned and where are we today?

Much research has been done in animals with some promising results which have been extensively
summarised in the literature (Da Silva et al. 2014; Koga et al. 2002). Limited research has been
done in humans (Table 2) and most of these studies were conducted in the early years and showed

This article is protected by copyright. All rights reserved


only short term protection. There was one passive vaccine generated in transgenic plants that
Accepted Article
showed a two-year protection which was promising, but it had problems of multiple doses and
recolonisation of S. sanguis and Veillonella (Ma et al. 1990; 1998). So what have we learnt from
40 years of research in caries vaccine? An era of research mainly from the Forsyth Dental centre
in Boston and the University of Alabama at Birmingham, Alabama, established pertinent aspects
of caries vaccines. The use of immunogenic proteins was identified. It was also established that a
multigenic vaccine provided better protection in animal studies. The requirement of adequate
adjuvants and the routes of administration were investigated. Liposomes (phospholipid membrane
vesicles), alum, bacterial flagellin and enterotoxins as adjuvants also proved to be better in
eliciting an immune response (Batista et al. 2014; Childers et al. 1999; Yang et al. 2017).

Based on these findings, research expanded to DNA/recombinant vaccines. The centres that have
been active for approximately the last 15 years are Wuhan University and the Chinese academy of
Science, Wuhan, China, and, for a short time, Shandong University in China and the University of
Florida collaborating with the University of Sao Paulo. Caries vaccine research has focused down
to some important advancements. A pGJA-P/VAX vaccine containing cytotoxic T lymphocyte-
associated antigen 4 gene, Fc gene, S. mutans gtfB and pac gene fragments has been constructed
and it has shown promising results in rats, rabbits and monkeys (Jia et al. 2006; Xu et al. 2007;
Niu et al. 2009). Recently, a vaccine trial in mice using LT derivative (detoxified) as an adjuvant
with the subunit of Pi39-512 and PstS protein has also been shown to elicit antigen specific antibody
production and a reduction in the adherence of S. mutans (Ferreira et al. 2016; Batista et al. 2017).

Yang et al. (2017) have also shown that a monomeric vaccine, KFD2-rPAc, which contains an
alanine-rich to proline-rich region fragment of PAc from S. mutans, and a partial length flagellin
attached with HIV-1 p24, produced significant amounts of rPAc specific serum IgG, serum IgA
and salivary IgA compared with the rPAC alone when administered to rats. In an in vitro study,
this serum and saliva also significantly inhibited biofilm formation. In addition, the caries
inhibition ratio improved from 18% (rPAc) to 50% (KFD2-rPAc). These results were improved by
adding a fragment of the GTF gene (Jiang et al. 2017). These researchers used the saliva-binding
region of PAc, the glucan-binding region of GTF-I and attached a dual promoter nirB-CMV to
enhance the immune response. This was delivered using Salmonella typhimurium which, on its
own, acts an adjuvant. In a mouse model using this vaccine, the immune response was enhanced
and S. mutans colonisation reduced. Perhaps, with new technology such as nano delivery systems

This article is protected by copyright. All rights reserved


and increasing knowledge, these vaccines can be improved further and taken into human clinical
Accepted Article
trials (Li et al. 2016; Cao et al. 2017).

Future direction

For future studies, two paths can be suggested. Firstly, to search for new target virulence genes or
antigenic proteins, develop vaccines, use the best historically proved adjuvant and administration
technique and enhance further using nanotechnology. For example, Ferreira et al. (2016) studied a
completely new protein, PstS, which showed promising results. Secondly, existing best-proved
animal trial vaccines can be improved to the required level. Multi-expert multi-centre studies are
required where the different vaccines can be discussed and compared. For example, monomeric vs
dimeric, type of adjuvants and immune promoters. Collective efforts should be placed on the most
promising vaccine rather than researchers working in isolation. In animal studies, the outcome
measures should also be standardised, such as serum and salivary antibody measurement and their
efficacy in the prevention of S. mutans adherence in vivo and in vitro. In addition, caries scores
should be measured. This will allow comparison of vaccines from different centres. Further to
that, human trials and further research can be conducted to establish efficacy, dosage and the
protection time period.

Although the use of fluoride through water fluoridation has been implemented by some countries
to reduce the prevalence of dental caries, it has been controversial due to ethical and public health
issues (Spencer & Do 2016; Peckham & Awofeso 2014). Reductions in starch and sugar
consumption, advocated for lifestyle illnesses, may assist oral health, but are unlikely to reduce
caries rates because of differences in socioeconomic status, especially in developing countries. A
caries vaccine, therefore, can still be the long term most cost-effective solution to the oral health
problem of dental caries.

In conclusion, dental caries is an irreversible, multifactorial, opportunistic infection. The treatment


is costly, making it a public health problem. Despite many years of promising laboratory research
and animal studies and some human clinical trials, there is no commercially available vaccine
today. This does not mean there will not be one tomorrow. The research objectives have become
more refined from lessons learnt over the years. Scientific hurdles have been overcome.
Multigenic DNA/recombinant vaccines, using the best proved adjuvants with a delivery system for
the nasal or sublingual route can be developed and researched with multicentre collaborative

This article is protected by copyright. All rights reserved


efforts. Animal studies and human clinical trials can be performed. Perhaps new vaccine targets
Accepted Article
can be identified. To overcome the economic hurdle, funders and public health interest should be
stimulated.

Acknowledgements

Author thanks Bruce Conradie and Lynne McNamara for their valuable comments and the
proofreading of the manuscript.

Conflict of interest

None to declare.

References

Bai, G., Tian, Y., Wu, J., Gu, Y., Chen, Z., Zeng, F. and Liu, J. (2019). Construction of a fusion
anti-caries DNA vaccine in transgenic tomato plants for PAcA gene and cholera toxin B subunit.
Biotechnol Appl Biochem Aug 21. doi: 10.1002/bab.1806.

Batista, M.T., Souza, R.D., Ferreira, E.L., Robinette, R., Crowley, P.J., Rodrigues, J.F., Brady,
L.J., Ferreira, L.C. and Ferreira, R.C. (2014). Immunogenicity and in vitro and in vivo protective
effects of antibodies targeting a recombinant form of the Streptococcus mutans P1 surface protein.
Infect Immun 82, 4978-4988. doi: 10.1128/IAI.02074-14. Epub 2014 Sep 15.

Batista, M.T., Ferreira, E.L., Pereira, G.S., Stafford, P., Maeda, D.L.N.F., Rodrigues, J.F., Brady,
L.J., Johnston, S.A., Ferreira, L.C.S. and Ferreira, R.C.C. (2017). LT adjuvant modulates epitope
specificity and improves the efficacy of murine antibodies elicited by sublingual vaccination with
the N-terminal domain of Streptococcus mutans P1. Vaccine, 35,7273-7282.

Cao, X.X., Fan, J., Chen, J., Li, Y.H. and Fan, M.W. (2016). Immunogenicity and prediction of
epitopic region of antigen Ag I/II and glucosyltransferase from Streptococcus mutans. J Huazhong
Univ Sci Technolog Med Sci 36, 416-421. doi: 10.1007/s11596-016-1602-y. Epub 2016 Jul 5.

Cao, X.X., Li, Y.H., Ye, Q.L., Hu, X., Wang, T.F. and Fan, M.W. (2017). Self-assembling
anticaries mucosal vaccine containing ferritin cage nanostructure and glucan-binding region of S.

This article is protected by copyright. All rights reserved


mutans glucosyltransferase effectively prevents caries formation in rodents. Hum Vaccin
Accepted Article
Immunother 13, 2332-2340.

Chen, L., Zhu, J., Li, Y., Lu, J., Gao, L., Xu, H., Fan, M. and Yang X. (2013). Enhanced nasal
mucosal delivery and immunogenicity of anti-caries DNA vaccine through incorporation of
anionic liposomes in chitosan/DNA complexes. PLoS One 8, e71953. doi:
10.1371/journal.pone.0071953.

Childers, N.K., Zhang, S.S. and Michalek, S.M. (1994). Oral immunization of humans with
dehydrated liposomes containing Streptococcus mutans glucosyltransferase induces salivary
immunoglobulin A2 antibody responses. Oral Microbiol Immunol 9, 146-153.

Childers, N.K., Tong, G. and Michalek, S.M. (1997). Nasal immunization of humans with
dehydrated liposomes containing Streptococcus mutans antigen. Oral Microbiol Immunol 12, 329-
335.

Childers, N.K., Tong, G., Mitchell, S., Kirk, K., Russell, M.W. and Michalek, S.M. (1999). A
controlled clinical study of the effect of nasal immunization with a Streptococcus mutans antigen
alone or incorporated into liposomes on induction of immune responses. Infect Immun 67, 618-
623.

Childers, N.K., Tong, G., Li, F., Dasanayake, A.P., Kirk, K. and Michalek, S.M. (2002). Humans
immunized with Streptococcus mutans antigens by mucosal routes. J Dental Res 81, 48-52.

Childers, N.K., Li, F., Dasanayake, A.P., Li, Y., Kirk, K. and Michalek, S.M. (2006). Immune
response in humans to a nasal boost with Streptococcus mutans antigens. Oral Microbiol Immunol
21, 309-313.

Cole, M.F., Emilson, C.G., Hsu, S.D., Li, S.H. and Bowen, W.H. (1984). Effect of peroral
immunization of humans with Streptococcus mutans on induction of salivary and serum
antibodies and inhibition of experimental infection. Infect Immun 46, 703-709.

This article is protected by copyright. All rights reserved


Accepted Article
Conrads, G., de Soet, J.J., Song, L., Henne, K., Sztajer, H., Wagner-Döbler, I. and Zeng, A.P.
(2014). Comparing the cariogenic species Streptococcus sobrinus and S. mutans on whole genome
level. J Oral Microbiol 6,26189. doi: 10.3402/jom.v6.26189. eCollection 2014.

da Silva, D.R., da Silva, A.C.B., Filho, R.M., Verli, F.D. and Marinho, S.A. (2014). Vaccine
against Dental Caries: An Update. Adv Microbiol 4, 925-933.

Dinis, M., Tavares, D., Veiga-Malta I., Fonseca A. J. M. M., Bonifácio Andrade E., Trigo G.,
Ribeiro A., Videira A., M A., Cabrita S. and Ferreira P. (2009). Oral therapeutic vaccination with
Streptococcus sobrinus recombinant Enolase confers protection against Dental Caries in Rats. J
Infect Dis 199, 116-123.

Dinis, M., Trigo, G., Chaves, N., Fonseca, A.J., Ribeiro, A., Tavares, D., Cabrita, A.M, and
Ferreira, P. (2011). rEnolase maternal immunization confers caries protection on offspring. J Dent
Res 90, 325-330. doi: 10.1177/0022034510391793.

Fan, M.W., Bia, Z., Peng, Z.X., Zhong, Y., Chen, Z., Peng, B. and Jia, R. (2002). A DNA vaccine
encoding a cell-surface protein antigen of Streptococcus mutans protects gnotobiotic rats from
caries. J Dent Res 81, 784-787.

Ferreira, E.L., Batista, M.T., Cavalcante, R.C., Pegos, V.R., Passos, H.M., Silva, D.A., Balan,
A., Ferreira, L.C. and Ferreira, R.C. (2016). Sublingual immunization with the phosphate-binding-
protein (PstS) reduces oral colonization by Streptococcus mutans. Mol Oral Microbiol 31, 410-
422. doi: 10.1111/omi.12142. Epub 2015 Dec 2.

Ferretti, J.J., Shea, C. and Humphrey, M.W. (1980). Cross-reactivity of Streptococcus mutans
antigens and human heart tissue. Infect Immun 30, 69-73.

This article is protected by copyright. All rights reserved


Gahnberg, L. and Krasse, B. (1983). Salivary immunoglobulin A antibodies and recovery from
Accepted Article
challenge of Streptococcus mutans after oral administration of Streptococcus mutans vaccine in
humans. Infect Immun 39, 514-519.

Gregory, R.L. and Filler, S.J. (1987). Protective secretory immunoglobulin A antibodies in
humans following oral immunization with Streptococcus mutans. Infect Immun 55, 2409-2415.

Guo, J.H., Jia, R., Fan, M.W., Bian, Z., Chen, Z. and Peng B. (2004). Construction and
immunogenic characterization of a fusion anti-caries DNA vaccine against PAc and
glucosyltransferase I of Streptococcus mutans. J Dent Res 83, 266-270.

Hajishengallis, G., Russell, M.W. and Michalek, S.M. (1998). Comparison of an adherence
domain and a structural region of Streptococcus mutans antigen I/II in protective immunity
against dental caries in rats after intranasal immunization. Infect Immun 66, 1740-1743.

Hajishengallis, G. and Michalek, S.M. (1999). Current status of a mucosal vaccine against dental
caries. Oral Microbiol Immunol 14, 1-20. Review.

Han, T.K. and Dao ML. (2007). Enhancement of salivary IgA response to a DNA vaccine against
Streptococcus mutans wall-associated protein A in mice by plasmid-based adjuvants. J Med
Microbiol 56(Pt 5), 675-680.

Han, T.K. and Dao, M.L. (2005). Differential immunogenicity of a DNA vaccine containing the
Streptococcus mutans wall-associated protein A gene versus that containing a truncated derivative
antigen A lacking in the hydrophobic carboxyterminal region. DNA Cell Biol 24, 574-581.

Huang, Y., Hajishengallis, G. and Michalek, S.M. (2001). Induction of


protective immunity against Streptococcus mutans colonization after mucosal immunization with
attenuated Salmonella enterica serovar typhimurium expressing an S. mutans adhesin under the
control of in vivo-inducible nirB promoter. Infect Immun 69, 2154-2161.

This article is protected by copyright. All rights reserved


Accepted Article
Huang, L., Xu, Q.A., Liu, C., Fan, M.W. and Li, Y.H. (2013). Anti-caries DNA vaccine-induced
secretory immunoglobulin A antibodies inhibit formation of Streptococcus mutans biofilms in
vitro. Acta Pharmacol Sin 34, 239-246. doi: 10.1038/aps.2012.145.

Jespersgaard, C., Hajishengallis, G., Huang, Y., Russell, M.W., Smith, D.J. and Michalek, S.M.
(1999). Protective immunity against Streptococcus mutans infection in mice after intranasal
immunization with the glucan-binding region of S. mutans glucosyltransferase. Infect Immun 67,
6543-6549.

Jia, R., Guo, J.H., Fan, M.W., Bian, Z., Chen, Z., Peng, B. and Fan, B. (2004). Mucosal
immunization against dental caries with plasmid DNA encoding pac gene of Streptococcus mutans
in rats. Vaccine 22, 2511-2516.

Jia, R., Guo, J.H., Fan, M.W., Bian, Z., Chen, Z., Fan, B., Yu, F. and Xu, Q.A. (2006).
Immunogenicity of CTLA4 fusion anti-caries DNA vaccine in rabbits and monkeys. Vaccine
24,5192-5200.

Jiang, H., Hu, Y., Yang, M., Liu, H. and Jiang, G. (2017). Enhanced immune response to a dual-
promoter anti-caries DNA vaccine orally delivered by attenuated Salmonella typhimurium.
Immunobiology 222,730-737.

Koga, T., Oho, T., Shimazaki, Y. and Nakano, Y. (2002). Immunization against dental caries.
Vaccine 20, 2027-2044.

Lehner, T., Russell, M.W. and Caldwell, J. (1980). Immunisation with a purified protein from
Streptococcus mutans against dental caries in rhesus monkeys. Lancet 1, 995-996.

Lehner, T., Russell, M.W., Caldwell, J. and Smith, R. (1981). Immunization with purified protein
antigens from Streptococcus mutans against dental caries in rhesus monkeys. Infect Immun 34,
407-415.

This article is protected by copyright. All rights reserved


Accepted Article
Li, F., Michalek, S.M., Dasanayake, A.P., Li, Y., Kirk, K. and Childers, N.K. (2003). Intranasal
immunization of humans with Streptococcus mutans antigens. Oral Microbiol Immunol 8, 271-
277.

Li, H., Lu, Y., Xiang, J., Jiang, H., Zhong, Y. and Lu, Y. (2016). Enhancement of immunogenic
response and protection in model rats by CSTM nanoparticles anticaries DNA vaccine.
Nanomedicine (Lond) 11, 1407-1416.

Liu, C., Fan, M., Bian, Z., Chen, Z. and Li, Y. (2008). Effects of targeted fusion anti-caries DNA
vaccine pGJA-P/VAX in rats with caries. Vaccine 26, 6685-9. doi:
10.1016/j.vaccine.2008.08.041.

Liu, G.X., Xu, Q.A., Jin, J., Li, Y.H., Jia, R., Guo, .JH. and Fan, M.W. (2009). Mucosal and
systemic immunization with targeted fusion anti-caries DNA plasmid in young rats. Vaccine 27,
2940-2947. doi: 10.1016/j.vaccine.2009.03.009.

Loesche, W.J. (1986). The identification of bacteria associated with periodontal disease and dental
caries by enzymatic methods. Oral Microbiol Immunol 1, 65-72.

Loimaranta, V., Laine, M., Söderling, E., Vasara, E., Rokka, S., Marnila, P., Korhonen,
H., Tossavainen, O. and Tenovuo, J. (1999). Effects of bovine immune and non-immune whey
preparations on the composition and pH response of human dental plaque. Eur J Oral Sci 107,
244-250.

Luz, D.E., Nepomuceno, R.S., Spira, B., Ferreira, R.C., Luz, D.E., Nepomuceno, R.S., Spira, B.
and Ferreira, R.C. (2012). The Pst system of Streptococcus mutans is important for phosphate
transport and adhesion to abiotic surfaces. Mol Oral Microbiol 27, 172-181. doi: 10.1111/j.2041-
1014.2012.00641.x. Epub 2012 Feb 18.

This article is protected by copyright. All rights reserved


Ma, J.K., Hunjan, M., Smith, R., Kelly, C. and Lehner, T. (1990). An investigation into the
Accepted Article
mechanism of protection by local passive immunization with monoclonal antibodies against
Streptococcus mutans. Infect Immun 58, 3407-3414.

Ma, J.K., Hikmat, B.Y., Wycoff, K., Vine, N.D., Chargelegue, D., Yu, L., Hein, M.B. and Lehner,
T. (1998). Characterization of a recombinant plant monoclonal secretory antibody and preventive
immunotherapy in humans. Nat Med 4, 601-606.

McGhee, J.R., Michalek, S.M., Webb, J., Navia, J.M., Rahman, A.F. and Legler, D.W. (1975).
Effective immunity to dental caries: protection of gnotobiotic rats by local immunization with
Streptococcus mutans. J Immunol 114, 300-305.

Mestecky, J., McGhee, J.R., Arnold, R.R., Michalek, S.M., Prince, S.J. and Babb, J.L. (1978).
Selective induction of an immune response in human external secretions by ingestion of bacterial
antigen. J Clin Invest 61, 731-737.

Mitoma, M., Oho, T., Michibata, N., Okano, K., Nakano, Y., Fukuyama, M. and Koga, T. (2002).
Passive immunization with bovine milk containing antibodies to a cell surface protein antigen-
glucosyltransferase fusion protein protects rats against dental caries. Infect Immun 70, 2721-2724.

Niu, Y., Sun, J., Fan, M., Xu, Q.A., Guo, J., Jia, R. and Li, Y. (2009). Construction of a new
fusion anti-caries DNA vaccine. J Dent Res 88, 455-460.

Peckham, S. and Awofeso, N. (2014). Water Fluoridation: A critical review of the physiological
effects of ingested fluoride as a public health intervention. Sci World J
http://dx.doi.org/10.1155/2014/293019

Petersen, .PE., Bourgeois, D., Ogawa, H., Estupinan-Day, S. and Ndiaye, C. (2005). The global
burden of oral diseases and risks to oral health. Bull WHO 83, 661-669.

Redman, T.K., Harmon, C.C. and Michalek, S.M. (1996). Oral immunization with recombinant
Salmonella typhimurium expressing surface protein antigen A (SpaA) of Streptococcus sobrinus:

This article is protected by copyright. All rights reserved


effects of the Salmonella virulence plasmid on the induction of protective and sustained humoral
Accepted Article
responses in rats. Vaccine 14, 868-878.

Redman, T.K., Harmon, C.C. and Michalek, S.M. (1994). Oral immunization with recombinant
Salmonella typhimurium expressing surface protein antigen A of Streptococcus sobrinus:
persistence and induction of humoral responses in rats. Infect Immunity 62, 3162-3171.

Redman, T.K., Harmon, C.C., Lallone, R.L. and Michalek, S.M. (1995). Oral immunization with
recombinant Salmonella typhimurium expressing surface protein antigen A of Streptococcus
sobrinus: dose response and induction of protective humoral responses in rats. Infect Immun 63,
2004-2011.

Robinette, R.A., Oli, M.W., McArthur, W.P. and Brady, L.J. (2011). A therapeutic anti-
Streptococcus mutans monoclonal antibody used in human passive protection trials influences the
adaptive immune response. Vaccine 29, 6292-300.

Salam, M.A., Katz, J., Zhang, P., Hajishengallis, G. and Michalek SM. (2006). Immunogenicity of
Salmonella vector vaccines expressing SBR of Streptococcus mutans under the control of a T7-
nirB (dual) promoter system. Vaccine 24, 5003-5015.

Shimazaki, Y., Mitoma, M., Oho, T., Nakano, Y., Yamashita, Y., Okano, K., Nakano,
Y., Fukuyama, M., Fujihara, N., Nada, Y. and Koga, T. (2001). Passive immunization with milk
produced from an immunized cow prevents oral recolonization by Streptococcus mutans. Clin
Diagn Lab Immunol 8, 1136-1139.

Smith, R. and Lehner, T. (1981). A radioimmunoassay for serum and gingival crevicular fluid
antibodies to a purified protein of Streptococcus mutans. Clin Exp Immunol 43, 417-424.

Smith, D.J. and Taubman, M.A. (1987). Oral immunization of humans with Streptococcus
sobrinus glucosyltransferase. Infect Immun 55, 2562-2569.

This article is protected by copyright. All rights reserved


Smith, D.J. and Taubman, M.A. (1990). Effect of local deposition of antigen on salivary immune
Accepted Article
responses and reaccumulation of mutans streptococci. J Clin Immunol 10, 273-281.

Smith, D.J. and Taubman, M.A. (1996). Experimental immunization of rats with a Streptococcus
mutans 59-kilodalton glucan-binding protein protects against dental caries. Infect Immun 64, 3069-
3073.

Smith, D.J., Heschel, R.L., Melvin, J., King, W.F., Pereira, M.B.B. and Taubman, M.A. (1997).
Streptococcus mutans glucan binding proteins as dental caries vaccines. In: Mucosal solutions.
Advances in mucosal immunology. Vol. 2. Husband AJ, Beagley KW, Clancy RL, Collins AM,
Cripps AW, Emery DL, editors. Sydney, Australia: University of Sydney Press, pp. 367-377.

Smith, D.J. (2002). Dental caries vaccines: prospects and concerns. Crit Rev Oral Biol Med 13,
335-349.

Smith, D.J., King, W.F., Rivero, J. and Taubman, M.A. (2005). Immunological and protective
effects of diepitopic subunit dental caries vaccines. Infect Immun 73, 2797-2804.

Spencer, A.J. and Do, L.G. (2016). Caution needed in altering the 'optimum' fluoride
concentration in drinking water. Community Dent Oral Epidemiol 44, 101-108.

Su, L.K., Yu, F., Li, Z.F., Zeng, C., Xu, Q.A. and Fan, M.W. (2014). Intranasal co-delivery of IL-
6 gene enhances the immunogenicity of anti-caries DNA vaccine. Acta Pharmacol Sin 35, 592-
598. doi: 10.1038/aps.2013.184.

Sun, J., Yang, X., Xu, Q.A., Bian, Z., Chen, Z. and Fan, M. (2009). Protective efficacy of two new
anti-caries DNA vaccines. Vaccine 27, 7459-7466. doi: 10.1016/j.vaccine.2009.05.007.

Sun, Y., Shi, W., Yang, J.Y., Zhou, D.H., Chen, Y.Q., Zhang, Y., Yang, Y., He, B.X., Zhong,
M.H., Li, Y.M., Cao, Y., Xiao, Y., Li, W., Yu, J., Li, Y.H., Fan, M.W. and Yan, H.M. (2012).

This article is protected by copyright. All rights reserved


Flagellin-PAc fusion protein is a high-efficacy anti-caries mucosal vaccine. J Dent Res 91, 941-
Accepted Article
947.

Sun, Y., Yang, Y., Zhou, D., Cao, Y., Yu, J., Zhao, B., Zhong, M., Li, Y., Yang, J. and Yan, H.
(2016). Flagellin-rPAc vaccine inhibits biofilm formation but not proliferation of S. mutans. Hum
Vaccin Immunother 12, 2847-2854.

Takahashi, I., Okahashi, N., Matsushita, K., Tokuda, M., Kanamoto, T., Munekata, Russell, M.W.,
and Koga, T. (1991). Immunogenicity and protective effect against oral colonization by
Streptococcus mutans of synthetic peptides of a streptococcal surface protein antigen. J Immunol
146, 332-336.

Talbman, M.A. and Smith, D.J. (1974). Effects of local immunization with Streptococcus mutans
on induction of salivary immunoglobulin A antibody and experimental dental caries in rats. Infect
Immun 9, 1079-1091.

Xu, Q.A., Yu, F., Fan, M.W., Bian, Z., Chen, Z., Peng, B., Jia, R. and Guo, J.H. (2007). Protective
efficacy of a targeted anti-caries DNA plasmid against cariogenic bacteria infections. Vaccine 25,
1191-1195.

Xu, Q.A., Yu, F., Fan, M., Bian, Z., Guo, J., Jia, R., Chen, Z., Peng, B. and Fan, B. (2005).
Immunogenicity and protective efficacy of a targeted fusion DNA construct against dental caries.
Caries Res 39, 422-431.

Yan, Y.H., Qi, S.C., Su, L.K., Xu, Q.A. and Fan, M.W. (2013). Co-delivery of CCL19 gene
enhances anti-caries DNA vaccine pCIA-P immunogenicity in mice by increasing dendritic cell
migration to secondary lymphoid tissues. Acta Pharmacol Sin 34, 432-440. doi:
10.1038/aps.2012.153.

This article is protected by copyright. All rights reserved


Yan, Y.H., Yu, F., Zeng, C., Cao, L.H., Zhang, Z. and Xu, Q.A. (2016). CCL17 combined with
Accepted Article
CCL19 as a nasal adjuvant enhances the immunogenicity of an anti-cariesDNA vaccine in rodents.
Acta Pharmacol Sin 37, 1229-1236. doi: 10.1038/aps.2016.73.

Yang, Y.P., Li, Y.H., Zhang, A.H., Bi, L. and Fan, M.W. (2009). Good Manufacturing Practices
production and analysis of a DNA vaccine against dental caries. Acta Pharmacol Sin 30, 1513-
1521. doi: 10.1038/aps.2009.152.

Yang, J., Sun, Y., Bao, R., Zhou, D., Yang, Y., Cao, Y., Yu, J., Zhao, B., Li, Y., Yan, H. and
Zhong, M. (2017). Second-generation flagellin-rPAc fusion protein, KFD2-rPAc, shows high
protective efficacy against dental caries with low potential side effects. Scientific Reports, 7,
11191. doi: 10.1038/s41598-017-10247-8.

Yu, H., Nakano, Y., Yamashita, Y., Oho, T. and Koga, T. (1997). Effects of antibodies against cell
surface protein antigen PAc-glucosyltransferase fusion proteins on glucan synthesis and cell
adhesion of Streptococcus mutans. Infect Immun 65, 2292-2298.

Zhang, F., Li, Y.H., Fan, M.W., Jia, R., Xu, Q.A., Guo, J.H., Yu, F. and Tian, Q.W. (2007).
Enhanced efficacy of CTLA-4 fusion anti-caries DNA vaccines in gnotobiotic hamsters. Acta
Pharmacologica Sinica 28, 1236-1242.

Table 1 Development in caries DNA vaccines

Antigen (Vaccine Study type Efficacy References


type)
SBR region of Intranasal and intragastric Anti-SBR antibodies detected in Huang et al.
AgI/II with immunisation of mice with serum and saliva after 3 weeks 2001
cholera toxin in S. single dose and booster after
typhimurium 17 weeks. Animals challenged Booster enhanced immune
(SBR-CTA2B) with virulent S. mutans response

This article is protected by copyright. All rights reserved


Accepted Article Significant reduction in the S.
mutans in plaque
SBR-CTA2B under the T7 and nirB promoters induced a Salam et al.
control of both the T7 and high and persistent mucosal and 2006
nirB promoters systemic anti-SBR antibody
response
Intranasal immunisation in
mice at 1, 18 & 320 days
Cell wall Intranasal 2 doses to mice, 3 Reduction in the S. mutans Han and Dao,
associated protein weeks apart adherence 2005
A (pCDNA-
wapA) pCDNA-wapA with DMRIE- Proved to be a weak adjuvant Han and Dao,
C a lipid adjuvant 2007
GLU region of Targeted salivary gland High level of Pac-specific salivary Fan et al. 2002
GTF+ two high- IgA with fewer carious lesions
conservative An administration to rats
regions of Pac Subcutaneous injections to Fewer carious lesions Guo et al.
(pGLUA-P-pCIA- rats 2004
P) Intranasal immunisation with High salivary s-IgA mucosal Jia et al. 2004
pCIA-P/bupivacaine DNA antibody response
complexes
Co-delivery of gene encoding Serum Pac-specific IgG increased Yan et al.
C-C chemokine ligand-19 from 4 to 14 weeks 2013
with pCIA-P to mice
Co-delivery of IL-6 high antigen specific IgG in serum Su et al. 2014
expressing plasmid pCI-IL-6 and sIgA in saliva
with pCIA-P
Lower level of caries
Mice immunised on day 0 and
14
Co-delivery of CCL17 and high antigen specific IgG and IgA Yan et al.
CCL19 with in serum 2016
pCIA-P
inhibition in the colonization of S.
Mice intranasal immunised mutans
and orally challenged with S.

This article is protected by copyright. All rights reserved


Accepted Article mutans
GLU region of pGJA P/VAX fused with T Accelerated and increased Jia et al. 2006
GTF+ two high- lymphocyte antigen-4 (CTLA- antibody response in serum and
conservative 4) Monkeys and rabbits were saliva in rabbits and monkeys
regions of Pac immunised
(pGJA P/VAX) Intranasal administration to Increase in antigen specific Xu et al. 2007
mice antibodies and significant
reduction in caries

Protection against S. mutans and


S. sobrinus
Hamsters were immunised High antigen specific antibodies Zhang et al.
with pGJA P/VAX - CTLA-4 and fewer caries 2007
Mice immunised with pGJA Widespread distribution of Liu et al. 2008
P/VAX intranasal/ plasmid with high expression in
intramuscular and in vivo fate the inoculated tissue and draining
of plasmid was assessed lymph nodes
5 day old Mice immunised Significant increase in serum IgG Liu et al. 2009
with pGJA P/VAX intranasal/ and IFN-gamma
intramuscular
Mice and rats were vaccinated High serum IgG and salivary Yang et al.
with Clinical- grade pGJA SIgA 2009
P/VAX with chitosan Impurities were under the limits of
set specification
Mice immunised with pGJA Increase in antigen specific Sun et al. 2009
P/VAX containing two new antibodies and reduction in caries
catalytic regions of GTF,
intranasal/ intramuscular
Rats were immunised with At 10 weeks SIgA reached its Huang et al.
pGJA P/VAX and peak which inhibited adherence 2013
S. mutans biofilm was Reduction in the thickness of
examined biofilm and the bacterial counts
Anionic liposomes and High transfection efficacy and Chen et al.
chitosan incorporated pGJA longer residence time of the DNA 2013
P/VAX (AL/CS/DNA) at nasal mucosal surface
In vivo and in vitro cellular High level of SIgA

This article is protected by copyright. All rights reserved


Accepted Article uptake was studied
GLU of GTF+ Pac Intramuscular and intra nasal Peak response seen after 8-10 Niu et al. 2009
with catalytic administration to mice on day weeks with high serum IgG anti-
fragment of S. 0 and 14 Pac and anti-GLU
sobrinus gtf-I (two
plasmids – High salivary anti-CAT
pGJGAC/VAX)
Flagellin derived Intranasal immunisation to High rPAc-specific antibodies in Sun et al. 2012
from E. coli (KF) rats serum and saliva
fused with antigen
PAc containing A- 8.5 µg KF-rPAc achived 64.2%
P fragment (rPAc) reduction in caries
(KF-rPAc) Biofilm inhibition by serum Inhibition of biofilm formation by Sun et al. 2016
and saliva of KF-rPAc serum and saliva
immunised rats was studied
SBR+GBR of Intragastrointestinal After 3 weeks anti-SBR and anti- Jiang et al.
GTF cloned in S. administration to mice GBR antibodies detected in serum 2017
typhimurium and saliva, booster enhanced
(SBR+GBR+CM antibodies
V and nirB
promoter) S. mutans challenged mice
showed reduction in S. mutans in
plaque
Pac with cholera Integrated in tomato genomes Gene expression was confirmed Bai et al. 2019
toxin (Pac-CTXB)
No animal studies reported

Table 2 Efficacy of active and passive immunisation against dental caries in humans

Antigen No. Route Efficacy Reference


(vaccine subjects
type)
Whole 4 Oral, capsule 3 months study period: Increase in Mestecky

This article is protected by copyright. All rights reserved


Killed S.
Accepted Article administrations SIgA during treatment and et al. 1978
mutans over 14 days thereafter decline, No effect on
serum antibody
Whole 11 Oral rinse and 53 days study period: No increase Gahnberg
Killed S. ingestion, 4 in SIgA, reduction in S. mutans and Krasse
mutans applications over 1983
35 days
Whole 8 1 oral application 360 days study period: No increase Cole et al.
Killed S. and ingestion for in SIgA, reduction in S. mutans 1984
mutans 3 days
Whole 6 Oral, capsule 180 days study period: Increase in Gregory
Killed S. administrations SIgA and decrease in salivary and and Filler
mutans over 10 days plaque S. mutans 1987
GTF 25 Oral, 18 capsule 42 days study period: Parotid saliva Smith &
(S. administrations IgA and anti-GTF increased Taubman
sobrinus) over 3 months Reduced recolonization 1987
GTF 23 Topical lower 6 weeks study: No effect on anti- Smith &
(S. lip, daily for 5 GTF IgA, Reduced recolonization Taubman
sobrinus) days 1990
Dehydrated 7 Oral, Ingested 8 weeks study: Increase in IgA1, Childers et
GTF in for 3 consecutive IgA2 and anti-GTF, No IgG and al. 1994
liposomes days and repeat IgM response
(S. mutans) after 28 days
Crude GTF 5 Intranasal, twice, 6 weeks study: Increase in IgA1 Childers et
in 7 days interval anti-GTF in nasal wash but less in al., 1997
liposomes saliva, Increased serum IgM and
(S. mutans) IgA-anti-GTF
C-GTF 21 Intranasal, 28 days study: Increased IgA in Childers et
(S. mutans) previously nasal wash, Increased IgG and IgA al. 1999
immunised anti-C-GTF in serum
group, 2

This article is protected by copyright. All rights reserved


Accepted Article administrations
over 7 days
E-GTF 21 Nasal spray two 3 months study: Increased anti-E- Childers et
(S. mutans) doses, one week GTF in saliva and nasal wash al. 2002
apart
Soluble and 12 Intranasal, 2 3 months study: Increased salivary Li et al.
liposomal applications over IgA anti-E-GTF 2003
E-GTF 7 days Increased IgA in nasal wash
Increased Serum IgG
E-GTF 26 Intranasal 3 months study: Increased mucosal Childers et
Tonsillar, and serum IgA-anti-GTF in both al. 2006
immunised twice routes, Increased IgA-anti-E-GTF
over 7 days in nasal wash and serum in nasal
route
PAc and 94 Not available Study period: not available, No Cao et al.
gtfB difference in total IgA, anti-PAc s 2016
IgA and anti-GLU in children with
or without caries
Ag I/II 15 Oral, 6 Two years of protection against Ma et al.
monoclonal applications over recolonisation by S. mutans but 1990
antibodies 3 weeks S. sanguis and Veillonella colonised
sites
Ag I/II 6 oral Reduced S. mutans colonisation for Ma et al.
monoclonal applications after 3.5 months 1998
antibodies CHX treatment
Cows 4 Milk Mouth 14 days study: Reduced Shimazaki
immunised rinse, twice a day recolonisation of S. mutans in saliva et al. 2001
with PAc and plaque
and GB
domain of
GTF-I

This article is protected by copyright. All rights reserved


Colostral
Accepted Article 9 3 times a day for Reduced S. mutans relative to Loimaranta
products 3 days mouth plaque flora et al. 1999
from rinsing
immunised
cows with
S. mutans

This article is protected by copyright. All rights reserved


lam_13218_f1.pdf

Figure 1 Virulence properties of S. mutans and the target antigens for caries vaccine
Accepted Article
development

Caries

Cariogenic
bacteria

Streptococcus mutans * Biofilm (plaque)


(Biofilm forming, Acidogenic & aciduric) formation
* * *
Glucosyltransferases Glucan binding Adhesin protein
(GTFs) protein (GBP) (Ag I/II/PAc/P1)
Adherence & glucan Adhesion of glucan to Adhesion to other
production bacteria & surfaces bacteria & surfaces

Extracellular polysaccharides (Glucan)


Adherence and nutrient

*: Virulence factors and target antigens for vaccine development to reduce the adherence of
bacteria

This article is protected by copyright. All rights reserved

Das könnte Ihnen auch gefallen