It is the cache of ${baseHref}. It is a snapshot of the page. The current page could have changed in the meantime.
Tip: To quickly find your search term on this page, press Ctrl+F or ⌘-F (Mac) and use the find bar.

Dental caries vaccine Shivakumar K M, Vidya S K, Chandu G N - Indian J Dent Res
Indian Journal of Dental ResearchIndian Journal of Dental ResearchIndian Journal of Dental Research
HOME | ABOUT US | EDITORIAL BOARD | AHEAD OF PRINT | CURRENT ISSUE | ARCHIVES | INSTRUCTIONS | SUBSCRIBE | ADVERTISE | CONTACT
Indian Journal of Dental Research   Login   |  Users online: 90

Home Bookmark this page Print this page Email this page Small font sizeDefault font size Increase font size         

 


 
REVIEW ARTICLE Table of Contents   
Year : 2009  |  Volume : 20  |  Issue : 1  |  Page : 99-106
Dental caries vaccine


1 Department of Preventive and Community Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Medical Sciences (Deemed University), Sawangi (Meghe), Wardha Maharashtra, 442 004, India
2 Department of Oral and Maxillofacial Pathology, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Medical Sciences (Deemed University), Sawangi (Meghe), Wardha Maharashtra, 442 004, India
3 Department of Preventive and Community Dentistry, College of Dental Sciences, Davangere, Karnataka - 577 004, India

Click here for correspondence address and email

Date of Submission 26-Oct-2007
Date of Decision 27-Oct-2007
Date of Acceptance 09-Aug-2008
 

   Abstract  

Dental caries is one of the most common diseases in humans. In modern times, it has reached epidemic proportions. Dental caries is an infectious microbiologic disease of the teeth that results in localized dissolution and destruction of the calcified tissue. Dental caries is a mulitifactorial disease, which is caused by host, agent, and environmental factors. The time factor is important for the development and progression of dental caries. A wide group of microorganisms are identified from carious lesions of which S. mutans , Lactobacillus acidophilus , and Actinomyces viscosus are the main pathogenic species involved in the initiation and development of dental caries. In India, surveys done on school children showed caries prevalence of approximately 58%. Surveys among the U.S. population showed an incidence of 45.3% in children and 93.8% in adults with either past or present coronal caries. Huge amounts of money and time are spent in treating dental caries. Hence, the prevention and control of dental caries is the main aim of public health, eventually the ultimate objective of public health is the elimination of the disease itself. Recently, dental caries vaccines have been developed for the prevention of dental caries. These dental caries vaccines are still in the early stages.

Keywords: Dental caries, dental caries vaccine, streptococcus mutans

How to cite this article:
Shivakumar K M, Vidya S K, Chandu G N. Dental caries vaccine. Indian J Dent Res 2009;20:99-106

How to cite this URL:
Shivakumar K M, Vidya S K, Chandu G N. Dental caries vaccine. Indian J Dent Res [serial online] 2009 [cited 2014 Mar 6];20:99-106. Available from: http://www.ijdr.in/text.asp?2009/20/1/99/49066
Dental caries is an infectious microbiologic disease of the teeth that results in localized dissolution and destruction of the calcified tissue. [1],[2] Dental caries is one of the most common diseases in humans. [3],[4] In modern times, it has reached epidemic proportions. The prevalence of dental caries in developed countries varies greatly and can reach over 90%. The rate of Caries has been increasing in developing countries with the increase in the popularity of highly refined sugars. The development of dental caries requires the presence of cariogenic bacteria that are capable of producing acid and a sugar present in the diet which favors the colonization of these bacteria to form acid. [3],[5] Dental caries appears to be a major public health problem which if left untreated can cause considerable pain, discomfort, and treatment costs are very high. Dental caries results from the interaction between the host, the host's diet, and the microflora on the tooth surface bounded by the time factor. [6] A wide group of microorganisms are identified from carious lesions of which  Streptococcus mutans Scientific Name Search utans), Lactobacillus acidophilus, and Actinomyces viscosus are the main pathogenic species involved in the initiation and development of dental caries. [7],[8] S. mutans has been implicated as a causative organism of dental caries. [4],[9] S. mutans accounts for seven distinct species isolated from animals and humans; Streptococcus cricetus, Streptococcus ferus, Streptococcus macacae, Streptococcus rattus, Streptococcus downey, S. mutans, and Streptococcus sobrinus. S. mutans and  Streptococcus sobrinus Scientific Name Search e exclusively isolated from humans and S. mutans is the most prevalent species. [10]

The traditional way of managing dental caries was by a surgical approach of "drill and fill". This approach has slowly evolved into a more conservative mode. Various preventive measures have been implicated for the prevention of dental caries, among which is immunization of the population against the disease. [7] Many studies have been conducted on the development of an effective vaccine to prevent the occurrence of dental caries. A review of literature pertaining to a dental caries vaccine is presented.


   Vaccines   Top


Vaccines are an immuno-biological substance designed to produce specific protection against a given disease. It stimulates the production of a protective antibody and other immune mechanisms. Vaccines are prepared from live modified organisms, inactivated or killed organisms, extracted cellular fractions, toxoids, or a combination thereof. [11]


   The Immune Response   Top


a) The primary response

When an antigen is administered for the first time to an animal or human, there is a latent period of induction of 3 to 10 days before antibodies appear in the blood. The antibody that is elicited first is entirely of the IgM type. The IgM antibody titer rises steadily during the next 2 to 3 days, reaches a peak level, and then declines almost as fast as it developed. Meanwhile, if the antigenic stimulus was sufficient, the IgG antibody appears in a few days. IgG reaches a peak in 7 to 10 days and then gradually falls over a period of weeks or months. An important outcome of the primary antigenic challenge is the education of the reticulo-endothelial system of the body. Both B and T lymphocytes produce what are known as "memory cells" or primed cells. These cells are responsible for the immunological memory that is established after immunization. [11],[12]

b) Secondary (Booster response)

The response to a booster dose differs in a number of ways from the primary response. The secondary response also involves the production of IgM and IgG antibodies. A collaboration between B and T cells is necessary to initiate a secondary response. There is a brief production of the IgM antibody and a much larger and more prolonged production of the IgG antibody. This accelerated response is attributed to immunological memory. The immune response (primary and secondary) and immunological memory are the basis of vaccination and revaccination. [11],[12]


   Mechanism of Action of Vaccine   Top


Saliva contains approximately 1-3% of immunoglobulin concentration, a majority of which is secretary IgA. However, saliva also contains the humoral immunoglobulin IgG and IgM from the gingival sulcular fluid. In addition, cellular components of the immune system such as lymphocytes, macrophages, and neutrophils are also present in gingival sulcus. Some of the possible ways antibodies might control bacterial growth are listed below: [13],[14]

  1. The salivary immunoglobulin may act as a specific agglutinin interacting with the bacterial surface receptors and inhibiting colonization and subsequent caries formation. They might also inactivate surface glucosyltransferase, which would then reduce the synthesis of extra cellular glucans resulting in reducing plaque formation.
  2. The salivary glands produce secretory IgA antibodies by direct immunization of the gut associated lymphoid tissue (GALT), from where sensitized B-cells may be home to the salivary glands. The salivary IgA antibodies have, of course, direct access to the tooth surface. They may prevent S. mutans from adhering to the enamel surface or they may prevent formation of dextran by inhibiting the activity of glucosyltransferase (GTF). [3]
  3. The gingival crevicular mechanism involves all the humoral and cellular components of the systemic immune system, which may exert its function at the tooth surface. There is now sufficient evidence to postulate what may happen after subcutaneous immunization with S. mutans. The organism is phagocytosed and undergoes antigenic processing by macrophages. In the lymphoid tissue, T and B-lymphocytes are sensitized by the macrophages preventing the antigen HLA Class-II complex and releasing IL-I. This induces the CD-4 helper and CD-8 cytotoxic suppressor cell response with the activation of IL-2 receptors and the release of IL2. The interaction between the cells play an essential part in modulating the formation of IgG, IgA, and IgM classes of antibodies and B-lymphocytes. [3],[15],[16]



   Studies   Top


a) Animal studies

Rodents are attractive as experimental laboratory animals because they are inexpensive and easy to maintain. Rapid decay of their teeth can be induced by S. mutans when present during the provision of a sugar containing diet. The ability to establish large experimental groups and to arrive at an accurate diagnosis of caries by examination of the tooth surface also makes rodents a good choice for laboratory animals. [17] Immunization experiments with cells of S. mutans both in rats and monkeys have consistently resulted in a significant decrease in dental caries. Purified components of S. mutans have been used only to a limited extent. Protection has been induced by immunization with GTF in rats. Recently, immunization with purified protein antigen I/II, which resides in the cell wall of S. mutans, has induced protection against caries. The latter utilizes only one subcutaneous injection of the antigen with adjuvant, unlike all other experiments, which have not been performed in rhesus monkeys and have used from 5 to 15 injections. Immunization with whole cells of S. mutans or with purified I/II antigen produces a reduction of about 70% in both smooth surface and fissure caries when compared with controls. [18] In gnotobiotic rats, ingestion of whole S. mutans selectively produces S-IgA. The appearance of S-IgA correlated with a reduced incidence of the caries vaccine. [17],[19]

The first report of successful immunization of monkeys against caries was by Bowen who injected whole cells S. mutans into Macaca fascicularis monkeys. [7] Rats and monkeys have been largely used in immunization studies. The principal design in most of the experiments has been to first immunize the animals with an antigen from S. mutans incorporated in an adjuvant as frequently as is necessary to attain high antibody levels, and follow this by implanting the same organism in the mouth and placing the animals on a high sucrose diet. Some of the experiments have been designed to aim for salivary immunity and others have been designed to aim for systemic immunity. [3] There have been reports of the successful use of GTF as an anticaries vaccine in rats and hamsters, but neither crude preparations, nor a highly purified GTF conferred any protection on monkeys. [20]

b) Human studies

As dental caries fulfills the criteria of an infectious disease, the possibility of preventing it by vaccination has been pursued. The rationale is that immunization with S. mutans should induce an immune response, which might prevent the organism from colonizing the tooth surface, and thereby prevent decay. As a vaccine would be administered before the deciduous dentition has erupted at about 6 months of age, it would prevent the disease in children who show the greatest incidence of caries. The vaccine could be given at the same time as the vaccines against diphtheria and tetanus. Immunity could be boosted at intervals thereafter to provide life-long protection. The existing delivery system of immunization could be used, without any financial burden being incurred. [3] Currently, clinical trials are underway to test the use of a pill of S. mutans for control of caries. There have been some conflicting results thus far in human studies. Some workers have actually reported a negative correlation between S-IgA and caries prevalence. However, this result could be the result of the experimental design. It has been shown that ingestion of capsules containing S. mutans stimulates the production of S-IgA. Stimulation of serum immunoglobulin in humans has also produced mixed results and no correlation could be made between caries experience and serum immunoglobulin stimulation. [8],[13]


   Antigenic Components of Streptococcus Mutans   Top


S. mutans posses various cell surface substances including adhesins, GTFs, and glucan binding proteins (GBP). These substances are used for vaccine preparation. Most of the recent experimental efforts have been directed toward these compounds. [21]

Adhesins: Adhesins form the two principal human pathogens of S. mutans (variously identified as antigens I/II, Pac, or P1 and Streptococcus sobrinus, Spa-A or Pag) and has been purified. Antigens I/II (Ag I/II) are found in the culture supernatant as well as in the S. mutans cell surface. This 185-KDa protein composed of a single polypeptide chain of approximately 1600 residues. Ag I/II contains an alanine-rich tandem-repeating region in the N-terminal third and a proline rich repeat region in the center of the molecule. These regions have been associated with the adhesin activity of Ag I/II. The proline-rich central portion contains an adhesin epitope basing their conclusions on adhesin inhibition assays involving the recombinant fragment of Ag I/II. The antibody directed to the intact Ag I/II molecule or to its salivary binding domain blocked adherence of S. mutans of saliva-coated hydroxyapatite. Immunization of mice with synthetic peptide (residue 301-319) from the alanine rich region of Ag I/II suppressed tooth colonization with S. mutans. [14],[22]


   Glucosyltransferase   Top


S. mutans has three forms of glucosyltransferases (GTFs):

  • Water insoluble glucan synthesizing enzyme: GTF-I
  • Water insoluble and water-soluble glucan synthesizingenzymes: GTF-S-I
  • Water-soluble glucan synthesizing enzymes: GTF-S


The genes encoding GTF-I, GTF-SI, and GTF-S are called the GTF-B, GTF-C, and GTF-D genes, respectively. All three GTF genes are important for smooth surface caries formation in the pathogen-free rat model system. Streptococcus sobrinus produces a water insoluble glucan-synthesizing enzyme GTF-S. The GTF-I gene encoding GTF-I and the GTF-S and GTF-T genes encoding two GTF-S enzymes have been cloned. [21],[23],[24] S. mutans and Streptococcus sobrinus each synthesize several GTFs. [25]

Glucan binding protein (GBP):
S. mutans secretes at least three distinct proteins with glucan binding activity: GBP-A, GBP-B, and GBP-C. Of the three S. mutans GBPs, only GBP-B has been shown to induce a protective immune response to experimental dental caries. GBP-A has a sequence of 563 amino acids. The molecular weight is 59.0 Kda. The carboxy-terminal 2/3 rd of GBP-A sequence has significant homology with a putative glucan binding region of S. mutans GTFs. The C-terminal region contains 16 repeating units, which represent the full glucan-binding domain of this protein. GBP-A has a greater affinity for water soluble glucan than for water insoluble glucan.

Dextranases: Dextran is an important constituent of early dental plaque. Dextranase is an enzyme produced by mutans Streptococcus. They destroy dextran and thus the bacteria can invade dextran-rich early dental plaque. Dextranase, when used as an antigen, can prevent the colonization of the organism in early dental plaque. [7]


   Routes of Immunization   Top


In general, 4 routes of immunization have been used with S. mutans:

  1. Oral
  2. Systemic (subcutaneous)
  3. Active gingivo-salivary
  4. Passive dental immunization


Common mucosal immune system

Mucosal applications of dental caries vaccines are generally preferred for the induction of secretory IgA antibodies in the salivary compartment, since this immunoglobulin constitutes the major immune component of major and minor salivary gland secretions. Many investigators have shown that exposure of an antigen to a mucosally associated lymphoid tissue in the gut, nasal, bronchial, or rectal site can give rise to immune responses not only in the region of induction but also in remote locations. This has given rise to the notion of a "common mucosal immune system". Consequently, several mucosal routes have been used to induce protective immune responses to dental caries vaccine antigens. [22],[26]

1. Oral route

Many of the earlier studies relied on oral induction of immunity in the GALT to elicit protective salivary IgA antibody responses. In these studies, an antigen was applied by oral feeding, gastric intubation, or in vaccine containing capsules or liposome. [22] Killed S. mutans was administered to germ-free rats in drinking water for 45 days before implantation of live S. mutans and then throughout the experimental period. A significant reduction in caries was related to an increased level of salivary IgA antibodies to S. mutans, as the serum antibody titer was minimal. Oral immunization with S. mutans did not induce significant secretory IgA in monkeys. Daily administration of 10 [11] cells of S. mutans in capsules produced a small increase in secretory IgA. The oral route failed to reduce caries significantly, as compared with subcutaneous immunization. The rise in secretory antibodies produced was small and of short duration, even after secondary immunization. Experiments in humans of the ingestion of S. mutans in gelatins capsules resulted in an increase in secretory IgA antibodies in saliva, although for a limited time only. Immunological memory in secretory IgA responses is rather limited and this may curtail the value of oral immunization. [3] Although the oral route was not ideal for reasons including the detrimental effects of stomach acidity on antigen, or because inductive sites were relatively distant, experiments with this route established that induction of mucosal immunity alone was sufficient to change the course of infection with S. mutans and disease in animal models and in humans. [22],[26],[27],[28]

2. Intranasal route

More recently, attempts have been made to induce protective immunity in mucosal inductive sites that are in closer anatomical relationship to the oral cavity. Intranasal installation of the antigen, the nasal associated lymphoid tissue (NALT), has been used to induce immunity to many bacterial antigens including those associated with mutans Streptococcal colonization and accumulation. Protective immunity after infection with cariogenic mutans streptococci could be induced in rats by the intranasal route with many S. mutans antigens or functional domains associated with these components. Protection could be demonstrated with S. mutans Ag I/II, the SBR of Ag I/II, a 19-mer sequence within the SBR, the glucan binding domain of S. mutans, GBP-B, and fimbrial preparations from S. mutans with antigen alone or combined with mucosal adjuvants. [22],[23],[27]

3. Tonsillar route

The ability of tonsillar application of antigens to induce immune responses in the oral cavity is of great interest. The tonsillar tissue contains the required elements of immune induction of secretory IgA responses although IgG, rather than IgA, response characteristics are dominant in this tissue. Nonetheless, the palatine tonsils, and especially the nasopharyngeal tonsils, have been suggested to contribute precursor cells to mucosal effector sites, such as the salivary glands. In this regard, the experiments have shown that topical application of formalin-killed Streptococcus sobrinus cells in rabbits can induce a salivary immune response, which can significantly decrease the consequences of infection with cariogenic Streptococcus sobrinus. Interestingly, repeated tonsillar application of a particulate antigen can induce the appearance of IgA antibodies producing cells in both the major and minor salivary glands of the rabbit. [22]

4. Minor salivary gland

The minor salivary glands populate the lips, cheeks, and soft palate. These glands have been suggested as potential routes for mucosal induction of salivary immune responses, given their short, broad secretory ducts that facilitate retrograde access of bacteria and their products and give the lymphatic tissue aggregates that are often found to be associated with these ducts. Experiments in which Streptococcus sobrinus GTF was topically administered onto the lower lips of young adults have suggested that this route may have potential for dental caries vaccine delivery. In these experiments, those who received labial application of GTF had a significantly lower proportion of indigenous S. mutans/total Streptococcal flora in their whole saliva during a 6-week period following a dental prophylaxis, compared with a placebo group. [22]

5. Rectal

More remote mucosal sites have also been investigated for their inductive potential. For example, rectal immunization with non oral bacterial antigens such as Helicobacter pylori or Streptococcus pneumoniae, presented in the context of toxin-based adjuvant, can result in the appearance of secretory IgA antibodies in distant salivary sites. The colo-rectal region as an inductive location for mucosal immune responses in humans is suggested from the fact that this site has the highest concentration of lymphoid follicles in the lower intestinal tract. Preliminary studies have indicated that this route could also be used to induce salivary IgA responses to mutans streptococcal antigens such as GTF. One could, therefore, foresee the use of vaccine suppositories as one alternative for children in whom respiratory ailments preclude the intranasal application of the vaccine. [22]

Systemic route of immunization

Subcutaneous administration of S. mutans was used successfully in monkeys and elicited predominantly serum IgG, IgM, and IgA antibodies. The antibodies find their way into the oral cavity via gingival crevicular fluid and are protective against dental caries. Whole cells, cell walls, and the 185 KD Streptococcal antigen have been administered on 2 to 4 occasions. A subcutaneous injection of killed cells of S. mutans in Freund's incomplete adjuvant or aluminium hydroxide elicits IgG, IgM, and IgA classes of antibodies. Studies have shown that IgG antibodies are well maintained at a high titer, IgM antibodies progressively fall and IgA antibodies increase slowly in titer. The development of serum IgG antibodies takes place within months of immunization, reaching a tire of up to 1:1280 with no change in antibodies being found in the corresponding sham-immunized monkeys. Protection against caries was associated predominantly with increased serum IgG antibodies. [3]

Active gingivo-salivary route

There has been some concern expressed regarding the side effects of using these vaccines with the other routes. In order to limit these potential side effects, and to localize the immune response, gingival crevicular fluid has been used as the route of administration. Apart from the IgG, it is also associated with increased IgA levels. [29]

The various modalities tried were as follows:

  • Injecting lysozyme into rabbit gingival, which elicited local antibodies from cell response
  • Brushing live S. mutans onto the gingiva of rhesus monkeys, which failed to induce antibody formation
  • Using smaller molecular weight Streptococci antigen, which resulted in better performance probably due to better penetration. [29]


Passive immunization

As the name suggests, passive immunization involves passive or external supplementation of the antibodies. This carries the disadvantage of repeated applications, as the immunity conferred is temporary.

Several approaches tried were:

  • Monoclonal antibodies


Monoclonal antibodies to S. mutans cell surface antigen I/II have been investigated. The topical application in human subjects brought a marked reduction in the implanted S. mutans. Thus, by bypassing the system, less concern exists about the potential side effects. [3],[29]

  • Bovine milk and whey


Systemic immunization of cows with a vaccine using whole S. mutans led to the bovine milk and whey containing polyclonal IgG antibodies. This was then added to the diet of a rat model. The immune whey brought a reduction in the caries level. This whey was also used in a mouth rinse, which resulted in a lower percentage of S. mutans in the plaque. [3,29]

  • Egg-yolk antibodies


The novel concept of using hen egg-yolk antibodies against the cell-associated glucosyltransferase of S. mutans was introduced by Hamada. Vaccines used were formalin killed whole cells and cell associated GTFs. Caries reduction has been found with both these treatments. [3]

  • Transgenic plants


The latest in these developments in passive immunization is the use of transgenic plants to give the antibodies. The researchers have developed a caries vaccine from a genetically modified (GM) tobacco plant. The vaccine, which is colorless and tasteless, can be painted onto the teeth rather than injected and is the first plant derived vaccine from GM plants. [30]

The advantages are listed below:

  • The genetic material can be easily exchanged.
  • It is possible to manipulate the antibody structure so that while the specificity of the antibody is maintained, the constant region can be modified to adapt to human conditions, thus avoiding cross reactivity.
  • Large scale production is possible as it would be quite inexpensive. [29]



   Adjuvants and Delivery Systems for Dental Caries Vaccines   Top


Various new approaches have been tried out to potentiate aspects of the immune response to induce sufficient antibodies to achieve a protective effect to overcome the existing disadvantages.

Synthetic peptides: Any antigen derived from animals or humans has the potential for hypersensitivity reaction. The chemically synthesized peptides hold an advantage in that this reaction can be avoided. This has been found to enhance the immune response. In humans, synthetic peptides elicited both IgG and T-cell proliferative responses, and the antibodies were both anti-peptide and anti-native. The synthetic peptides give antibodies not only in the GCF but also in the saliva. The synthetic peptide used is derived from the Glucosyltransferase enzyme. [23],[29]

Coupling with Cholera Toxin Subunits: Cholera toxin (CT) is a powerful mucosal immunoadjuvant, which is frequently used to enhance the induction of mucosal immunity to a variety of bacterial and viral pathogens in animals systems. Mucosal application of a soluble protein or peptide antigen alone rarely results in elevated or sustained IgA responses. However, the addition of small amounts of CT or the closely related E. coli heat-labile enterotoxins (LT) can greatly enhance mucosal immune responses to intragastrically or intranasally applied mutans Streptococcal antigens or to peptides derived from these antigens. The coupling of the protein with the nontoxic unit of the cholera toxin was effective in suppressing the colonization of S. mutans. [22],[24],[29]

Fusing with  Salmonella More Details: The avirulent strains of salmonella are an effective vaccine vector; fusion using recombinant techniques have been used. [29]

Microcapsules and microparticles: Combinations of antigens in or various types of particles have been used in an attempt to enhance mucosal immune responses. The microcapsules and microparticles made of poly lactide-co-glycolide (PLGA) have been used as local delivery systems because of their ability to control the rate of release, evade preexistent antibody clearance mechanisms, and degrade slowly without eliciting an inflammatory response to the polymer. [29]

Liposomes: Liposomes, which are bilayered phospholipids membrane vesicles manufactured to contain and deliver drugs and antigens, have been used to enhance mucosal responses to mutans Streptococcal carbohydrate and GTF. Liposomes are thought to improve mucosal immune responses by facilitating M cell uptake and delivery of antigen to lymphoid elements of inductive tissue. [22],[29]


   Risks of Using Caries Vaccine   Top


All vaccines, even if properly manufactured and administered, seem to have risks. The most serious is that sera of some patients with rheumatic fever who show serological cross-reactivity between heart tissue antigens and certain antigens from hemolytic Streptococci. [31] Experiments from antisera from rabbits immunized with whole cells of S. mutans and with a high molecular weight protein antigen of S. mutans were reported to cross react with normal rabbit and human heart tissues. Polypeptides (62-67 KDA) immunologically cross-reactive with human heart tissue and rabbit skeleton muscles myosin are found in the cell membrane of S. mutans and Streptococcus ratti. [32] On the other hand, demonstrations showed that rabbit antiserum to high molecular weight, Todd-Hewitt broth components reacted with monkey cardiac muscle with S. mutans coated with medium components. Heart cross-reactive antibodies do not develop in rhesus monkeys or rabbits immunized with purified Ag I/II from S. mutans. It is possible that increased production of heart-reactive antibody in rabbits immunized with mutans streptococci results in injury of heart tissue as a consequence of binding of this low molecular weight Streptococcal polypeptide. Because of the potential of Streptococcal whole cells to induce heart reactive antibodies, the development of a sub-unit vaccine for controlling dental caries has been the focus of intense research interest. Glucosyltransferase was also tested for cross-reactivity with human heart tissue and the results were negative. [21,33] Further research showed that the C-terminal part of Ag I/II contains an epitope, which is cross-reactive with human IgG and, although the clinical significance of this observation is unknown, it appears that this potentially harmful epitope should be excluded from a caries vaccine. The human IgG cross-reactive region is also present in other mutans streptococci such as Streptococcus sobrinus as well as in non mutans streptococci. [33]


   Public Health Aspects   Top


Although the prevalence of tooth decay has declined according to a national epidemiologic survey by the United States national institute of dental research, this oral disease is still a significant health problem that affects approximately 50% of the 5 to 17 year old children in the U.S. In fact, by the age of 17, only 16% remain free of caries. Interestingly, 25% of children and adolescents aged 5 to 17 years old account for 80% of caries in permanent teeth, indicating the existence of high-risk groups. Meanwhile, in many developing countries in Central and South America, Africa, Asia, as well as in certain European countries, dental caries is on the rise. Despite the wide spread use of fluoride (e.g., in toothpaste and drinking water) to which the decline of caries between the 1970s and 1980s is mainly attributed, this disease still remains among the most prevalent and costly in industrialized and in developing countries. Indeed, developing countries without a water fluoridation system and where access to dental health education and treatment may not be available to everyone are in great need of a vaccine. An effective, safe, and readily deliverable vaccine may not only help against pain and health issues associated with caries but also save the billions of dollars that are currently spent in the U.S. for restorative treatment. [33] An important question is whether the search for a caries vaccine is justified from a public health point of view. This question is especially critical as we already have effective means to control the disease. Given that S. mutans is not the only cariogenic microorganism and that a series of factors influence the development of the disease, the question arises as to what extent successful vaccination against S. mutans could reduce the incidence of dental caries. [31] Considerable caries reduction could be attained if colonization of S. mutans could be prevented or reduced at the time of eruption of both deciduous and permanent teeth. Thus, a successful vaccination directed against S. mutans could be a valuable adjunct to other caries-preventive measures. Some other studies also suggest that vaccination could be a supplement to antimicrobial treatment in individuals with high levels of S. mutans. In third-world countries, a rapid increase in caries has been observed both in children and adolescents. The low dentist to population ratio and the lack of organized dental health care limit the possibilities of utilizing conventional caries-preventive methods. Therefore, it was thought that vaccination against dental caries could be of great value as a preventive adjunct in some societies and as a major public health measure in others. It ought to be stressed, however, that a thorough analysis of the need, cost benefits, and risk-benefits of a vaccine against dental caries in various societies and subgroups has to be performed. [31]


   Discussion   Top


Loesche [34] stated that dental caries is one of the most widespread diseases of mankind. S. mutans is the primary etiologic agent of dental caries that are transmissible. It is also said that a strong association exists between the level of colonization with S. mutans and dental caries, although other organisms such as Lactobacilli have also been implicated in this disease. Adding to this, the studies conducted by Caufield, et al.[35] stated that under normal circumstances of diet, children become permanently colonized with S. mutans between the middle of the 2 nd year and the end of the 3 rd year of life. This period is called the window of infectivity. Many studies were conducted on active immunization. Childers, et al. [36] immunized adults orally by using enteric coated capsules filled with crude Streptoccoccus mutans Gs-5 GTF antigen preparation contained in liposome, resulting in the production of parotid salivary IgA antibodies. Similarly, the studies by Childers, et al.[37] have shown that nasal immunization with dehydrated liposome containing GTRF preparation induced significant IgA antibody response in nasal washes. However, the studies by Smith, et al.[38] reported that mucosal immunization with GTF could influence the re-emergence of mutans Streptococci in adults after a dental prophylaxis. Although the oral route was not ideal for reasons such as the detrimental effects of stomach acidity on antigens or because inductive sites were relatively distant, experiments with this route established that induction of mucosal immunity alone was sufficient to change the course of mutans Streptococci infection and disease in animal models by Michalek, et al.[39] and humans by Smith, et al.[38] Most recently, attempts have been made to induce protective immunity in mucosal inductive sites closes to the oral cavity. Studies conducted by Katz, et al.[40] have demonstrated that intranasal immunization of rats with AgI/II - CTB induced a protective salivary immune response, which was associated with a reduction in Streptococci mutans colonization and Streptococci mutans induced caries. The studies by Brandtzaed [41] have shown that tonsillar application of antigens to the palatine tonsils i.e., nasopharyngeal tonsils may contribute precursor cells to mucosal effectors sites such as salivary glands. The experiments performed by Schroeder, et al.[42] have shown that Streptococcus sobrinus GTF was topically administered on the lower lips of young adults and suggested that this route may have potential for dental caries vaccine delivery. Kleanthous, et al.[43] conducted experiments through rectal immunization with non oral bacteria antigens such as H. pylori resulting in secretory IgA antibodies in distant salivary sites. Filler, et al.[44] conducted experiments on passive immunization and showed that passive immune protection can be achieved with antibodies to GTF or GBPs mouth rinses containing bovine milk or hen egg yolk . Childers, et al. [45] reported that the antibody to S. mutans cells lead to a modest, short-term decrease in the number of Streptococci mutans in saliva or dental plaque.


   Conclusion   Top


Clearly, there is strong evidence that S. mutans and Streptococcus sobrinus are closely associated with dental caries. Fluoride treatment used abroad has successfully limited caries progression, but was not sufficient to control this infectious disease even when used together with professional tooth cleaning and dietary counseling in populations highly exposed to these cariogenic microbiota. Active and passive immunization strategies, which target key elements in the molecular pathogenesis of mutans Streptococci, hold promise. Integrating these approaches into broad-based public health programs may yet forestall dental caries disease in many of the world's children, among whom those of high risk might derive the greatest benefit. Despite the encouraging decline in dental caries observed in recent years in many populations, millions of children remain at risk of experiencing extensive tooth decay and it is particularly distressing that many of those suffering will be among the least likely to obtain satisfactory treatment. Along with established methods of caries prevention, caries vaccines have the potential of making a highly valuable contribution to disease control. In the meantime, basic research on the mode of action of caries vaccine and the search for new, more effective, and possibly polyvalent vaccines must continue if we are to fully explore their potential for helping us in the struggle against dental caries. Regardless of the mechanism by which immune protection against dental caries is achieved, further advances to make immunization against caries practical will depend upon clinical trials aimed at establishing whether the findings from animal experiments can be transferred to humans. Particular goals for such studies include determining whether appropriate immune responses can be safely generated in humans, especially in susceptible age groups and whether such responses will afford desirable levels of protection.

Although several methods such as topical or systemic use of fluorides, fissure sealants, and dietary control have been developed to prevent dental caries, the efficacy of these methods is not enough to eradicate dental caries in humans; however, there are a few studies on the efficacy of caries vaccines in humans.

 
   References   Top

1. Clifford MS. The art and science of Operative dentistry. 4 th ed. Mosby Publications; 1995.  Back to cited text no. 1    
2. Hajishengallis G, Russel MW, Michalek SM. Comparison of an adherence domain and a structural region of S. mutans antigen I/II in protective immunity against dental caries in rats after intranasal immunization. Infect Immun 1998;66:1740-3.  Back to cited text no. 2    
3. Lehner T. Immunology of dental caries. Immunology of oral diseases. 3 rd ed. Blackwell scientific publications; 1992.  Back to cited text no. 3    
4. Smith DJ, Godiska R. Passive immunization approaches for dental caries prevention. Conference Proceedings. Egg Symposium; 2004. p. 1-6.  Back to cited text no. 4    
5. Winston AE, Bhaskar SN. Caries Prevention in the 21 st century. J Am Dent Assoc 1998;129:1579-87.  Back to cited text no. 5    
6. Kruger C, Pearson SK, Kodama Y, Vacca Smith A, Bowen WH, Hammarstrom L. The effects of Egg-derived antibodies to Glucosyltransferase on dental caries in rats. Caries Res 2004;38:9-14.  Back to cited text no. 6    
7. Krithika AC, Kandaswamy D, Gopikrishna V. Caries vaccine-I today's myth. J Indian Assoc Public Health Dent 2004;4:21-5.  Back to cited text no. 7    
8. Russel MW, Childers NK, Michalek SM, Smith DJ, Taubman MA. A caries vaccine? The state of science of immunization against dental caries. Caries Res 2004;38:230-5.  Back to cited text no. 8    
9. Guo JH, Jia R, Fan MW, Bian Z, Chen Z, Peng B. Construction and Immunologic characterization of a fusion anti-caries DNA vaccine against Pac and Glucosyltransferase - I of S. mutans . J Dent Res 2004;83:266-70.   Back to cited text no. 9    
10. Mattos-Graner RO, Smith DJ. The vaccination approach to control infections leading to dental caries. Braz J Oral Sci 2004;3:595-608.  Back to cited text no. 10    
11. Park K. Text book of preventive and social medicine. 17 th ed. Bhanotidas Publication; 2004.  Back to cited text no. 11    
12. Warren L, Ernest J. Medical microbiology and immunology. 6 th ed. Lange Medical Publishing Division; 2000.  Back to cited text no. 12    
13. Newman, Nisengard. Oral microbiology and immunology. W.B. Saunder's Company; 1988.   Back to cited text no. 13    
14. Bowen WH. Vaccine against dental caries: A personal view. J Dent Res 1996;75:1530-9.  Back to cited text no. 14    
15. Wilton JM. Future control of dental disease by immunization: Vaccines and oral health. Int Dent J 1984;34:177-83.  Back to cited text no. 15    
16. Lehner T, Challacombe SJ, Caldwell J. Immuologic basis for vaccination against dental caries in Rhesus monkeys. J Dent Res 1976;55:C166-80.  Back to cited text no. 16    
17. Russel RR, Johnson NW. The prospects for vaccination against dental caries. Br Dent J 1987;10:29-33.  Back to cited text no. 17    
18. Lehner T. Future possibilities for the prevention of caries and periodontal disease. Br Dent J 1980;149:318-25.  Back to cited text no. 18    
19. Bowen WH. Do we need to be concerned about dental caries in the coming millennium? Crit Rev Oral Biol Med 2002;13:126-31.  Back to cited text no. 19    
20. Russel RR, Beighton D, Cohen B. Immunization of monkeys (Macaca fascicularis) with Antigens purified from S. mutans . Br Dent J 1982;152:81-4.  Back to cited text no. 20    
21. Koga T, Oho T, Shimazaki Y , Nakano Y. Immunization against dental caries. Vaccine 2002;20:2027-44.   Back to cited text no. 21    
22. Smith DJ. Dental caries vaccines: Prospects and concerns. Crit Rev Oral Biol Med 2002;13:335-49.  Back to cited text no. 22    
23. Moro I, Lehner T. Symposium report: Sixth International congress of mucosal immunology; Dental caries vaccine. J Dent Res July 23 rd 1990. p. 1863-4.  Back to cited text no. 23    
24. Russell RR. The application of molecular genetics to the microbiology of dental caries. Caries Res 1994;28:69-82.  Back to cited text no. 24    
25. Luo Z, Smith DJ, Taubman MA, King WF. Cross sectional analysis of serum antibody to oral streptococcal antigens in children. J Dent Res 1998;67:554-60.  Back to cited text no. 25    
26. Russell MW, Hajishengallis G, Childers NK, Michalek SM. Secretory Immunity in defense against cariogenic Mutans streptococci. Caries Res 1999;33:4-15.  Back to cited text no. 26    
27. Curtis R 3rd. 1984 Kreshover lecture: Genetic analysis of S. mutans virulence and prospects for an anticaries vaccine. J Dent Res 1986;65:1034-45.  Back to cited text no. 27    
28. Bowen WH, Cohen B, Cole M, Colman G. Immunization against dental caries: Summary. J Dent Res 1976;55:C164-5.   Back to cited text no. 28    
29. Tandon S. Textbook of Pedodontics. 1 st ed. Paras Publishing; 2001.  Back to cited text no. 29    
30. News. Genes 'n' Greens: The future of oral medicine? Br Dent J 2002;192:674.  Back to cited text no. 30    
31. Krasse B, Emilson CG, Gahnberg L. An anticaries vaccine: Report on the status of research. Caries Res 1987;21:255-76.  Back to cited text no. 31    
32. Harris R. Vaccines for dental caries. Aust Dent J 1983;28:115-6.  Back to cited text no. 32    
33. Hajishengallis G, Michalek SM. Current status of a mucosal vaccine against dental caries. Oral Microbiol Immunol 1999;14:1-20.  Back to cited text no. 33    
34. Loesche WJ. Role of S. mutans in human dental decay. Microbiol Rev 1986;50:353-80.  Back to cited text no. 34    
35. Caufield PW, Cutter GR, Dasanayake AP. Initial acquisition of Mutans Streptococci by infants: Evidence for a discrete window of infectivity. J Dent Res 1993;72:37-45.  Back to cited text no. 35    
36. Childers NK, Zhang SS, Michalek SM. Oral immunization of humans with dehydrated liposomes conataining S. mutans Glycosyltransferase induces salivary immunoglobulin A2 antibody responses. Oral Microbiol Immunol 1994;9:146-53.  Back to cited text no. 36    
37. Childers NK, Tong G, Mitchell S, Kirk K, Russell MW, Michalek SM. A controlled clinical study of the effect of nasal immunization with a S. mutans antigen alone or incorporated into liposomes on induction of immune responses. Infect Immun 1999;67:618-23.  Back to cited text no. 37    
38. Smith DJ, Taubman MA. Oral immunizaqtion of humans with Streptococcus sobrinus glucosyltransferase. Infect Immun 1987;55:2562-9.  Back to cited text no. 38    
39. Michalek SM, McGhee JR, Mestecky J, Arnold RR, Bozzo L. Ingestion of S. mutans induces secretory IgA and caries immunity. Science 1976;192:1238-40.  Back to cited text no. 39    
40. Katz J, Harmon CC, Buckner GP, Richardson GJ, Russsell MW, Michalek SM. Proctective salivary immunoglobulin A responses against S. mutans infection after intranasal immunization with S. mutans antigen I/II coupled to the B Subunit of Cholera toxin. Infect Immun 1993;61:1964-71.  Back to cited text no. 40    
41. Brandtzaeg P. The B-cell development in tonsillary lymphoid follicles. Acta Otolaryngol Suppl 1996;523:55-9.  Back to cited text no. 41    
42. Schroeder HE, Moreillon MC, Nair PN. Architecture of minor salivary gland duct/lymphoid follicle associations and possible antigen recognition sites in the monkey Macaca fascicularis. Arch Oral Biol 1983;28:133-43.  Back to cited text no. 42    
43. Kleanthous H, Myers GA, Geoorgakopoulos KM, Tibbitts TJ, Ingrassia JW, Gray HL, et al . Rectal and intranasal immunizations with recombinant urease induce distinct local and serum immune responses in mice and protect against Helicobacter pylori infection. Infect Immun 1998;66:2879-86.  Back to cited text no. 43    
44. Filler SJ, Gregory RL, Michalek SM, Katz J, McGhee JR. Effect of immune bovine milk on S. mutans in human dental plaque. Arch Oral Biol 1991;36:41-7.  Back to cited text no. 44    
45. Childers NK, Tong G, Li F, Dasanayake AP, Kirk K, Michalek SM. Humans Immunized with S. mutans antigens by Mucosal Routes. J Dent Res 2001;81:48-52.  Back to cited text no. 45    

Top
Correspondence Address:
K M Shivakumar
Department of Preventive and Community Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Medical Sciences (Deemed University), Sawangi (Meghe), Wardha Maharashtra, 442 004
India
Login to access the Email id


DOI: 10.4103/0970-9290.49066

PMID: 19336869

Get Permissions




This article has been cited by
1 Association between immunological status and caries experience among adults attending Hospital Universiti Sains Malaysia (HUSM)
Wan Mohamad, W.M. and Wan Aziz, W.N.A.B. and Ismail, N.M. and Basri, R. and Alam, M.K.
International Medical Journal. 2013; 20(2): 208-210
[Pubmed]
2 Oral manifestations and their treatment in Sjögren's syndrome
S González,H Sung,D Sepúlveda,MJ González,C Molina
Oral Diseases. 2013; : n/a
[Pubmed]
3 Antimicrobial effect of tea and tea with milk beverages on oral Streptococcus mutans and Lactobacilli
Abd Allah, A.A. and Ibrahium, M.I. and Abd Allah, S.M. and Amin, M.A.
World Applied Sciences Journal. 2012; 19(9): 1327-1334
[Pubmed]
4 Developing a system for antibacterial treatment of dental caries using culture based approach
Mishra, M. and Ghosh, S. and Alex, L.E. and Mukherjee, I. and Sinha, T.P. and Ray Chaudhuri, S.
OnLine Journal of Biological Sciences. 2012; 12(2): 44-53
[Pubmed]
5 Vaccine against dental caries- an urgent need
Gambhir, R.S. and Singh, S. and Singh, G. and Singh, R. and Nanda, T. and Kakar, H.
Journal of Vaccines and Vaccination. 2012; 3(2)
[Pubmed]
6 Bacteriology of deep carious lesions underneath amalgam restorations with different pulp-capping materials - An in vivo analysis
Neelakantan, P. and Rao, C.V.S. and Indramohan, J.
Journal of Applied Oral Science. 2012; 20(2): 139-145
[Pubmed]
7 Prevalence of dental caries bacterial pathogens and evaluation of inhibitory concentration effect on different tooth pastes against Streptococcus spp
Maripandi, A., Kumar, A.T., Al Salamah, A.A.
African Journal of Microbiology Research. 2011; 5(14): 1778-1783
[Pubmed]
8 Anti-cell-associated glucosyltransferase immunoglobulin Y suppression of salivary mutans streptococci in healthy young adults
Nguyen, S.V., Icatlo Jr., F.C., Nakano, T., Isogai, E., Hirose, K., Mizugai, H., Kobayashi-Sakamoto, M., (...), Chiba, I.
Journal of the American Dental Association. 2011; 142(8): 943-949
[Pubmed]
9 Saliva microbiomes distinguish caries-active from healthy human populations
The ISME Journal. 2011;
[VIEW]
10 Contemporary anticarious agents and procedures [Suvremena antikarijesna sredstva i postupci]
Verzak, Ž. and Čuković-Bagić, I. and Škrinjarić, T.
Paediatria Croatica, Supplement. 2010; 54(SUPPL. 1): 163-168
[Pubmed]
11 Preliminary X-ray crystallographic analysis of SMU.2055 protein from the caries pathogenStreptococcus mutans
Wang-Hong Zhao,Xiu-Rong Zhan,Xiong-Zhuo Gao,Xiang Liu,Yi-Fei Zhang,Jiuxiang Lin,Lan-Fen Li,Shi-Cheng Wei,Xio-Dong Su
Acta Crystallographica Section F Structural Biology and Crystallization Communications. 2010; 66(5): 530
[Pubmed]
12 Induction of neutralizing antibodies in mice immunized with an amino-terminal polypeptide of Streptococcus mutans P1 protein produced by a recombinant Bacillus subtilis strain
Milene B. Tavares,Bruno M. Silva,Rafael C.M. Cavalcante,Renata D. Souza,Wilson B. Luiz,Juliano D. Paccez,Paula J. Crowley,L. Jeannine Brady,Luís C.S. Ferreira,Rita C.C. Ferreira
FEMS Immunology & Medical Microbiology. 2010; 59(2): 131
[Pubmed]
13 Induction of neutralizing antibodies in mice immunized with an amino-terminal polypeptide of Streptococcus mutans P1 protein produced by a recombinant Bacillus subtilis strain
Tavares, M.B. and Silva, B.M. and Cavalcante, R.C.M. and Souza, R.D. and Luiz, W.B. and Paccez, J.D. and Crowley, P.J. and Brady, L.J. and Ferreira, L.C.S. and Ferreira, R.C.C.
FEMS Immunology and Medical Microbiology. 2010; 59(2): 131-142
[Pubmed]
14 Novel technologies for the prevention and treatment of dental caries: A patent survey
Chen, F. and Wang, D.
Expert Opinion on Therapeutic Patents. 2010; 20(5): 681-694
[Pubmed]
15 Preliminary X-ray crystallographic analysis of SMU.2055 protein from the caries pathogen Streptococcus mutans
Zhao, W.-H. and Zhan, X.-R. and Gao, X.-Z. and Liu, X. and Zhang, Y.-F. and Lin, J. and Li, L.-F. and Wei, S.-C. and Su, X.-D.
Acta Crystallographica Section F: Structural Biology and Crystallization Communications. 2010; 66(5): 530-533
[Pubmed]



 

Top
 
 
  Search
 
 
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Email Alert *
    Add to My List *
* Registration required (free)  
 


    Abstract
    Vaccines
    The Immune Response
    Mechanism of Act...
    Studies
    Antigenic Compon...
    Glucosyltransferase
    Routes of Immuni...
    Adjuvants and De...
    Risks of Using C...
    Public Health As...
    Discussion
    Conclusion
    References

 Article Access Statistics
    Viewed 19912    
    Printed 600    
    Emailed 57    
    PDF Downloaded 3181    
    Comments  [Add]    
    Cited by others  15    

Recommend this journal