Skip to main content

MINI REVIEW article

Front. Oral. Health, 14 April 2022
Sec. Oral Infections and Microbes
Volume 3 - 2022 | https://doi.org/10.3389/froh.2022.880746

Probiotics for Oral Candidiasis: Critical Appraisal of the Evidence and a Path Forward

  • 1Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
  • 2Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT, United States

Oropharyngeal Candidiasis (OPC) is a mucosal fungal infection that is prevalent among patients with compromised immunity. The success of probiotics in treating chronic diseases with a microbial etiology component at other mucosal sites (i.e., gastro-intestinal, genitourinary and alveolar mucosae) has inspired research into the use of probiotics in the treatment of OPC. A growing body of research in vitro and in animal models indicates that some probiotic species and strains have inhibitory activities against Candida albicans growth, morphological switching, and biofilm formation. However, recent review and meta-analysis studies reveal a dearth of human randomized, controlled clinical trials on the efficacy of probiotics to treat or prevent OPC, while the majority of these have not based their selection of probiotic strains or the type of administration on sound pre-clinical evidence. In this mini-review, we assess the state of the field, outline some of the difficulties in translating lab results to clinical efficacy, and make recommendations for future research needed in order to move the field forward.

Introduction

Oropharyngeal Candidiasis (OPC) is a common fungal infection affecting populations with immature or weakened immune systems including neonates, the elderly, AIDS patients, and those taking immunosuppressive drugs [14]. Like other diseases in which commensal microbes become pathogenic, OPC is associated with a dysbiotic state, an alteration in the composition or abundance of the oral health-associated host microbiota, contributing to a permissive environment for Candida spp. infection [5, 6]. An aging population, the rise in the use of immune-compromising treatments and the increasing prevalence of drug-resistant Candida strains make it necessary to find alternative treatments for OPC.

Probiotics are defined by the FAO/WHO as “live microorganisms which, when consumed in appropriate amounts in food, confer a health benefit on the host” [7]. Their use is believed to discourage pathogenic microbes and help the host microbiota to return to a health-associated, balanced state after disruptions [8]. Research has provided evidence supporting a positive role of some probiotic organisms in promoting gut, oral and vaginal health, but clinical studies showing no effect of probiotics also exist [911].

In this mini-review we explore the current evidence for the role of probiotics in the prevention and treatment of OPC. We will not summarize all work in this area, a task well-accomplished by recent systematic reviews [9, 1214]. Rather, our intention is to critically appraise the state of the science on this topic by integrating the information gleaned from studies in vitro and in animal models with the results of human clinical trials, where possible. We discuss the pitfalls of each type of research and the difficulties encountered in translating results from laboratory research to benefits for patients, then suggest areas that need more research attention in the future.

What Can We Learn from Clinical Trials?

Three recent systematic review and meta-analysis studies explored probiotics as treatment or prevention for OPC [1517]. These studies restricted their analyses to randomized, controlled clinical trials (RCTs) which measured Candida burdens in colony-forming units (CFUs) from saliva samples or swabs of oral mucosal tissues. The meta-analyses included between 3 [1820] and 8 clinical trials [2125] that compared a probiotic to a control or placebo. All three meta-analyses found significant reduction in Candida counts in the oral environment as a result of probiotic treatment, although this did not always lead to significant clinical improvements. This suggests that the development of probiotic approaches holds promise in changing current clinical guidelines of OPC prevention or treatment, especially for immunocompromised patients and for patients with refractory or recurring infections.

Bacterial Species Tested as Probiotics in Clinical Trials and Basis for Their Selection

Ideally, when probiotic strains are chosen for clinical trials involving OPC, it would be on the basis of experimental results, either in vitro, or in animal models, or both, that demonstrate their activity against Candida species. Typically, research begins with in vitro studies on the interactions between probiotic species and Candida species, proceeds to in vivo animal model studies to examine aspects of the interaction between probiotics, Candida, and the host, and then moves on to human clinical trials of the probiotic species that show promise. However, probiotics in fermented foods have been part of the human diet since ancient times and scientific study into the clinical benefits of probiotic bacteria for human health began in the late 19th century when scientific methods of inquiry were not well-developed [26]. In addition, much of the early work in the study of probiotics to treat or prevent disease has focused on gastro-intestinal conditions [27]. Because of their presumed safety and benefits in intestinal mucosal diseases, some probiotic species have been tested in humans for their ability to prevent OPC without any prior experimental evidence supporting their anti-Candida activities [18, 22].

The justification behind the choice of probiotic species and strains was specified in 6 of the 8 RCTs covered by the 3 meta-analyses. Commercial probiotic preparations usually contain several species of bacteria, thus clinical trials often use a mixture of species or strains. Lactobacillus species were included in all but one of the RCTs. L. rhamnosus was the most commonly included species (4 studies) and L. reuteri, L. acidophilus, and L. bulgaricus were each used in 2 studies. Additional species in probiotic mixtures included Bifidobacterium longum, Bifidobacterium bifidum, Propionibacterium freadenreichii, and Streptococcus thermophilus. In two of the studies, no reason was provided for the choice of probiotic species or strain [22, 24]. Two of the RCTs used a mixture of 2 strains of L. reuteri [20, 25] citing this species' production of the antibacterial toxin reuterin and its ability to inhibit the in vitro growth of many Candida spp. [28]. L. rhamnosus Lr32 and L. acidophilus NCFM, which were compared in Miyazima et al. [23], had been previously found to reduce oral C. albicans numbers in an immunocompromised mouse model [29] and to disrupt C. albicans biofilms in vitro [30]. This same group conducted a RCT of a commercial, three-species mixture of L. rhamnosus HS111, L. acidophilus HS 101 and Bifidobacterium bifidum [19], citing studies that tested these and other Lactobacillus species against Candida growth in vitro [31, 32]. In one study intended to improve dental caries incidence in children, Streptococcus salivarius was chosen for its antagonism with S. mutans [21]; changes in both Candida and S. mutans numbers were recorded because of the known synergistic roles of these 2 organisms in dental caries [33, 34]. However, in this study, there was no significant difference in Candida oral burdens between probiotic treatment and placebo. To explain their choice of probiotic species, Hatakka and colleagues [18] cited a successful clinical trial which measured gastrointestinal colonization of Candida in neonates given L. rhamnosus GG [35] and a mouse study which found that probiotic treatment with either Propionibacterium JS or L. rhamnosus GG increased the proliferation of T- and B-cells [36]. The third strain in their study, L. rhamnosus LC705, was used in combination with Propionibacterium JS, which is a preservative against yeast in food manufacture [18]. Thus, it appears that justification for the use of specific probiotic strains in existing RCTs is variable, and the majority of these studies were not based on strong experimental in vitro evidence or on results with preclinical animal models of oral infection.

Few research groups have progressively investigated the same probiotic species in vitro, in an animal model, and in human clinical trials. One such group first found each of 2 Lactobacillus strains, L. rhamnosus LR32 and L. acidophilus NCFM, to be effective against OPC in an immunocompromised mouse model [29]. They then showed the ability of the same strains to interfere with adhesion and hyphal growth in the early stages of C. albicans biofilm formation in vitro [30]. In the ensuing human clinical trial, both strains reduced the oral Candida levels in denture wearers, one (LR32) performing even better than the positive control drug Nystatin [23]. The same group tested a commercial product containing different strains of the same two species, L. rhamnosus HS111 and L. acidophilus HS 101, in a successful clinical trial in denture-wearers [19]. These results however were not reproduced independently by a different group and have not led to widespread implementation of these probiotics in clinical practice.

Lactobacillus rhamnosus GG (LrGG) was patented for use as a probiotic in 1989 and there is an extensive body of clinical research justifying claims of its health benefits, mostly in the treatment of gastro-intestinal disorders [37, 38]. Many of the characteristics that make it promising as a probiotic for gut ailments could apply to the oral environment–it adheres to epithelial cells, promotes type 1 immune response, reduces inflammation by inhibiting macrophage activation and increases production of IL-10, IL-12 and TNFα in innate immune cells [37]. A great deal of in vitro and in vivo laboratory work has delved into the mechanisms behind this strain's ability to inhibit potentially pathogenic organisms directly and through its influence on host immune responses [37]. LrGG inhibits C. albicans adhesion and morphological changes [39], induces metabolic reprogramming by nutrient limitation [40], and may break down cell-wall components [41]. LrGG also reduces oral Candida counts in immunocompromised mice [42]. Although there is strong experimental and preclinical evidence supporting the potential efficacy of this strain in limiting OPC, it was used in only one of the above listed RCTs and then, only in combination with other species [18]. More RCTs should provide independent support for the benefits of this probiotic strain in the context of OPC treatment or prevention in humans.

Choosing the Best Probiotics for the Oral Environment

While these clinical and laboratory studies are good first steps, it is evident that in order to make progress in the development of probiotics-based new therapies or prevention strategies for patients who are most susceptible to OPC, we will need to approach scientific inquiry in a more systematic way. This process should include more in vitro work to screen potential probiotics for their activity against Candida species and examine mechanisms of anti-fungal activity. Some criteria for a good probiotic for the treatment of intestinal illness (i.e., the ability to withstand the acid environment of the stomach, be metabolically active under the low oxygen tension in the gut, or to attach to intestinal epithelial cells) may not apply to the oral environment. In choosing the best probiotics for the oral environment, in vitro experiments are needed to explore attachment to oral epithelial cells and the ability to colonize the oral mucosa. It is also important to isolate, identify and test metabolites produced by effective species as these postbiotics could prevent OPC without the risks associated with introducing live organisms in immunocompromised patients who make up most of the susceptible populations [43]. Animal models that recapitulate the different host susceptibility states (i.e., immunocompromised, elderly, cancer patients) will be needed to discover the best probiotic species for each condition and also to test a variety of delivery methods.

Pitfalls in Translating Laboratory Findings to Clinical Trials

Experimental work has provided evidence of several anti-Candida activities of Lactobacillus spp. that negatively affect C. albicans virulence characteristics including growth, yeast to hyphae transition, adhesion, and biofilm development [44, 45]. Antifungal mechanisms include production of acids, hydrogen peroxide, bacteriocins, and biosurfactants, as well as competition for adhesion sites. Probiotics can also activate protective functions of the immune system, affecting cytokine profiles, innate and adaptive immune responses, and Candida recognition by epithelial and immune cells [44, 45]. However, which of these protective mechanisms are active in the human oral environment is not fully understood. In the oral environment some of the probiotic protective functions may be compromised by dietary habits and nutrient availability, antagonistic interactions with a complex resident microbiota, and the salivary flow which may limit colonization and persistence.

With respect to persistence in the oral cavity, the impact of probiotic consumption on the oral microbiome is relatively unexplored. The question of whether probiotic organisms become stably incorporated into the human host oral microbiome is not often addressed in clinical trials. However, there is some evidence that while ingested organisms are often detected in the oral cavity of participants during probiotic treatment, they do not persist once treatment ceases [24, 46]. Nevertheless, because OPC is precipitated by an underlying dysbiosis in the oral microbiome [5, 6], it would be important to demonstrate that probiotics can encourage the recovery of a “healthy” microbiome in the oral environment after disruptions caused by antibiotics, cancer treatment or chronic disease, as they appear to do in the gut [8]. These important questions are beginning to be explored using next generation sequencing technology [47, 48] and require more investigation using preclinical models of infection.

It is clear that complex interactions between the fungus, the resident microbiota and the host immune system determine whether C. albicans remains commensal or becomes pathogenic in the oral environment [49, 50]. T helper-17 (Th17) cell-mediated immunity is crucial in maintaining commensal colonization of the oral mucosa by C. albicans [51, 52]. The host aryl hydrocarbon receptor (AhR) binds ligands that are products of tryptophan catabolism and activates IL-17/IL-22 production by Th17 cells. Lactobacilli metabolize tryptophan to produce an AhR ligand, indole-3-aldehyde, which triggers IL-22 production to protect from C. albicans infection and dampen inflammation [53]. Further exploration in this area is needed to reveal how probiotic organisms contribute to metabolic control of protective immune responses in the oral environment.

The effects of probiotic organisms on epithelial barrier function and host immunity have been extensively studied in the context of gastro-intestinal diseases [11, 54]. Probiotics affect host immunity at both the local and systemic levels. Systemic immune effects of probiotic consumption could influence the host's ability to control Candida in the oral environment without the requirement for probiotic organisms to take up residence there. For example, consumption of Lactobacillus strains in mice was tied to improvement in the function of both peritoneal and alveolar macrophages and protection against both lung and peritoneal infection by C. albicans [55]. These distant protective effects may extend to the oral environment.

Experimental evidence suggests that probiotic organisms prime epithelial and immune cells to respond differently to Candida challenge. Pretreatment with Lactobacillus strains before exposure to C. albicans causes changes in cytokine production and pattern recognition receptor expression in macrophages [56]. In epithelial (HeLa) cell lines, pretreatment with L. crispatus altered β-defensin and cytokine production and toll-like receptor expression in response to C. albicans [57]. To shed light on the role of probiotics in the oral environment, more work must be done using oral epithelial cell lines, primary epithelial cells and mucosal organotypic cultures [58]. It will also be important to differentiate between direct effects of the probiotic organisms on epithelial tissues vs. effects secondary to Candida growth and morphology, which can also alter immune responses [59].

The Path Forward: Developing Better-Targeted Probiotics-Based Approaches

Evidence from RCT meta-analyses suggests that probiotic treatments may work better in preventing OPC, compared to treating established infection in susceptible patient categories [1517]. This may indicate that short-term administration of probiotics cannot lead to changes in the oral microbiota that are sufficient in magnitude or duration to treat this infection. Interestingly, in these meta-analyses when studies that involved elderly patients or denture wearers were considered separately [1820, 23], a stronger beneficial effect was found compared to including all studies and all susceptible patient groups together [15, 16]. A recent RCT, not included in the meta-analyses, confirmed this trend [60]. In other patient populations (children and adults), oral Candida numbers showed low or no responsiveness to probiotic treatments [21, 24, 25]. This evidence suggests that probiotics must be better targeted to specific vulnerable populations such as patients who are HIV positive, undergoing cancer treatment, or taking immunosuppressive drugs. In vivo animal models mimicking these conditions may be useful in developing a better-targeted, host immune susceptibility type-combined with a specific probiotic strain approach in treatment or prevention.

In the 8 RCTs mentioned above, the dose, timing, duration and delivery methods for probiotics were varied. Dosages in adult populations ranged from 7.2 x 107 to 2 x 1010 CFUs per day. To put these numbers in perspective, a dosage of 1010 CFU per day was required to colonize the digestive tract and to treat acute gastroenteritis in children [61, 62], suggesting that better outcomes might have been achieved with higher doses in OPC studies with adults. Delivery methods included lozenges, cheese, yogurt, milk and the application of probiotic organisms directly onto denture surfaces. The timing of delivery ranged from once per day to 3 x per day and the duration of treatment ranged from 4 weeks to 3 months. No pilot studies were conducted to compare the efficacy of particular dosages, timing, delivery or duration and there is no indication of whether any of these variables are essential for success. Again, preclinical animal models could provide useful insights on these variables.

Discussion

Based on recent studies, it is clear that relatively few clinical trials of probiotics for OPC were based on sound in vitro experimental data and/or appropriate preclinical animal models of infection. Because of the beneficial immunomodulatory effects of several probiotic strains, we suggest that further research is needed in animal models that recapitulate the specific host immune deficiency, to better target the probiotics to the specific underlying immune-compromising condition of susceptible patients.

It will also be important to further study the mechanisms of inhibition of Candida spp. by probiotic organisms and particularly to determine whether these mechanisms are active in the oral environment. Animal models should be used to determine the best methods for delivery of probiotics to the oral cavity before being applied in humans. Molecules with anti-Candida activity that are produced by probiotic species must be identified and their activity tested in appropriate animal models. Further, it will be essential to explore immune effects of probiotics both at the local level of the oral mucosa and at the systemic level and to determine how these changes in immune responses impact the susceptibility or severity of OPC.

Author Contributions

LA researched the literature and wrote and edited the article. AD-B provided guidance and focus and edited the article. Both authors contributed to the article and approved the submitted version.

Funding

This study was funded by NIH/NIDCR Grant RO1 DE013986 and NIH/NIGMS Grant RO1 GM127909.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Lombardi A, Ouanounou A. Fungal infections in dentistry : clinical presentations, diagnosis, and treatment alternatives. Oral Surg Oral Med Oral Pathol Oral Radiol. (2020) 130:533–46. doi: 10.1016/j.oooo.2020.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Fidel PL. Candida-host interactions in HIV disease: implications for oropharyngeal candidiasis. Adv Dent Res. (2011) 23:45–9. doi: 10.1177/0022034511399284

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Brent NB. Thrush in the breastfeeding dyad: results of a survey on diagnosis and treatment. Clin Pediatr. (2001) 40:503–6. doi: 10.1177/000992280104000905

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Gendreau L, Loewy ZG. Epidemiology and etiology of denture stomatitis. J Prosthodont. (2011) 20:251–60. doi: 10.1111/j.1532-849X.2011.00698.x

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Bertolini M, Ranjan A, Thompson A, Diaz PI, Sobue T, Maas K, et al. Candida albicans induces mucosal bacterial dysbiosis that promotes invasive infection. PLOS Pathog. (2019) 15:e1007717. doi: 10.1371/journal.ppat.1007717

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Abusleme L, Diaz PI, Freeman AF, Greenwell-Wild T, Brenchley L, Desai JV, et al. Human defects in STAT3 promote oral mucosal fungal and bacterial dysbiosis. JCI Insight. (2018) 3:e122061. doi: 10.1172/jci.insight.122061

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Food Argricultural Organization/World Health Organization. Joint FAO/WHO Expert Consultation on Evaluation of Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria. Cordoba, Argentina (2001). Available online at: https://scholar.google.com/scholar?hl=en&as_sdt=0%2C7&q=Joint+FAO%2FWHO+exper+consultation+on+evaluation+of+health+and+nutritional+properties+of+probiotics+in+food&btnG= (accessed January 16, 2022).

Google Scholar

8. McFarland L. Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review. BMJ Open. (2014) 4:e005047. doi: 10.1136/bmjopen-2014-005047

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Shenoy A, Gottlieb A. Probiotics for oral and vulvovaginal candidiasis: a review. Dermatol Ther. (2019) 32:e12970. doi: 10.1111/dth.12970

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Gerritsen J, Smidt H, Rijkers GT, de Vos WM. Intestinal microbiota in human health and disease: the impact of probiotics. Genes Nutr. (2011) 6:209–40. doi: 10.1007/s12263-011-0229-7

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Wilkins T, Sequoia J. Probiotics for gastrointestinal conditions: a summary of the evidence. Am Fam Physician. (2017) 96:170–8.

PubMed Abstract | Google Scholar

12. Matsubara VH, Bandara HMHN, Mayer MPA, Samaranayake LP. Probiotics as Antifungals in Mucosal Candidiasis. Clin Infect Dis. (2016) 62:1143–53. doi: 10.1093/cid/ciw038

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Andrade JC, Kumar S, Kumar A, Cernáková L, Rodrigues CF. Application of probiotics in candidiasis management. Crit Rev Food Sci Nutr. (2021) 1–16. doi: 10.1080/10408398.2021.1926905

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Chugh P, Dutt R, Sharma A, Bhagat N, Dhar MS. A critical appraisal of the effects of probiotics on oral health. J Funct Foods. (2020) 70:103985. doi: 10.1016/j.jff.2020.103985

CrossRef Full Text | Google Scholar

15. Mundula T, Ricci F, Barbetta B, Baccini M, Amedei A. Effect of probiotics on oral candidiasis: a systematic review and meta-analysis. Nutrients. (2019) 11:2449. doi: 10.3390/nu11102449

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Hu L, Zhou M, Young A, Zhao W, Yan Z. In vivo effectiveness and safety of probiotics on prophylaxis and treatment of oral candidiasis: a systematic review and meta-analysis. BMC Oral Health. (2019) 19:140. doi: 10.1186/s12903-019-0841-2

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Ai R, Wei J, Ma D, Jiang L, Dan H, Zhou Y, et al. A meta -analysis of randomized trials assessing the effects of probiotic preparations on oral candidiasis in the elderly. Arch Oral Biol. (2017) 83:187–92. doi: 10.1016/j.archoralbio.2017.04.030

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Hatakka K, Ahola AJ, Yli-Knuuttila H, Richardson M, Poussa T, Meurman JH, et al. Probiotics reduce the prevalence of oral Candida in the elderly a randomized controlled trial. J Dent Res. (2007) 86:125–30. doi: 10.1177/154405910708600204

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Ishikawa KH, Mayer MPA, Miyazima TY, Matsubara VH, Silva EG, Paula CR, et al. A multispecies probiotic reduces oral candida colonization in denture wearers. J Prosthodont. (2015) 24:194–9. doi: 10.1111/jopr.12198

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Kraft-Bodi E, Jørgensen MR, Keller MK, Kragelund C, Twetman S. Effect of probiotic bacteria on oral candida in frail elderly. J Dent Res. (2015) 94:181S−6. doi: 10.1177/0022034515595950

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Burton JP, Drummond BK, Chilcott CN, Tagg JR, Thomson WM, Hale JDF, et al. Influence of the probiotic Streptococcus salivarius strain M18 on indices of dental health in children: a randomized double-blind, placebo-controlled trial. J Med Microbiol. (2013) 62:875–84. doi: 10.1099/jmm.0.056663-0

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Li D, Li Q, Liu C, Lin M, Li X, Xiao X, et al. Efficacy and safety of probiotics in the treatment of Candida-associated stomatitis. Mycoses. (2014) 57:141–6. doi: 10.1111/myc.12116

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Miyazima TY, Ishikawa KH, Mayer MPA, Saad SMI, Nakamae AEM. Cheese supplemented with probiotics reduced the Candida levels in denture wearers—RCT. Oral Dis. (2017) 23:919–25. doi: 10.1111/odi.12669

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Petti S, Tarsitani G, D'Arca AS. A randomized clinical trial of the effect of yoghurt on the human salivary microflora. Arch Oral Biol. (2001) 46:705–12. doi: 10.1016/S0003-9969(01)00033-4

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Keller MK, Kragelund C. Randomized pilot study on probiotic effects on recurrent candidiasis in oral lichen planus patients. Oral Dis. (2018) 24:1107–14. doi: 10.1111/odi.12858

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Santacroce L, Charitos IA, Bottalico L. A successful history: probiotics and their potential as antimicrobials. Expert Rev Anti Infect Ther. (2019) 17:635–45. doi: 10.1080/14787210.2019.1645597

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Floch MH. The role of prebiotics and probiotics in gastrointestinal disease. Gastroenterol Clin North Am. (2018) 47:179–91. doi: 10.1016/j.gtc.2017.09.011

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Jørgensen MR, Kragelund C, Jensen PØ, Keller MK, Twetman S. Probiotic lactobacillus reuteri has antifungal effects on oral Candida species in vitro. J Oral Microbiol. (2017) 9:1274582. doi: 10.1080/20002297.2016.1274582

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Matsubara VH, Silva EG, Paula CR, Ishikawa KH, Nakamae AEM. Treatment with probiotics in experimental oral colonization by Candida albicans in murine model (DBA/2). Oral Dis. (2012) 18:260–4. doi: 10.1111/j.1601-0825.2011.01868.x

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Matsubara VH, Wang Y, Bandara HMHN, Mayer MPA, Samaranayake LP. Probiotic lactobacilli inhibit early stages of Candida albicans biofilm development by reducing their growth, cell adhesion, and filamentation. Appl Microbiol Biotechnol. (2016) 100:6415–26. doi: 10.1007/s00253-016-7527-3

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Hasslöf P, Hedberg M, Twetman S. Stecksén-blicks C. Growth inhibition of oral mutans streptococci and candida by commercial probiotic lactobacilli - an in vitro study. BMC Oral Health. (2010) 10:2–7. doi: 10.1186/1472-6831-10-18

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Sookkhee S, Chulasiri S, Prachyabrued W. Lactic acid bacteria from healthy oral cavity of Thai volunteers: inhibition of oral pathogens. J Appl Microbiol. (2001) 90:172–9. doi: 10.1046/j.1365-2672.2001.01229.x

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Raja M, Hannan A, Ali K. Association of oral candidal carriage with dental caries in children. Caries Res. (2010) 44:272–6. doi: 10.1159/000314675

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Falsetta ML, Klein MI, Colonne PM, Scott-Anne K, Gregoire S, Pai CH, et al. Symbiotic relationship between Streptococcus mutans and Candida albicans synergizes virulence of plaque biofilms in vivo. Infect Immun. (2014) 82:1968–81. doi: 10.1128/IAI.00087-14

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Manzoni P, Mostert M, Leonessa ML, Priolo C, Farina D, Monetti C, et al. Oral supplementation with lactobacillus casei subspecies rhamnosus prevents enteric colonization by candida species in preterm neonates: a randomized study. Clin Infect Dis. (2006) 42:1735–42. doi: 10.1086/504324

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kirjavainen PV, ElNezami HS, Salminen SJ, Ahokas JT, Wright PFA. Effects of orally administered viable lactobacillus rhamnosus gg and propionibacterium freudenreichii subsp. shermanii js on mouse lymphocyte proliferation. Clin Diagn Lab Immunol. (1999) 6:799–802. doi: 10.1128/CDLI.6.6.799-802.1999

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Capurso L. Thirty years of lactobacillus rhamnosus GG a review. J Clin Gastroenterol. (2019) 53:1–41. doi: 10.1097/MCG.0000000000001170

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Manzoni P. Use of Lactobacillus casei subspecies rhamnosus GG and gastrointestinal colonization by candida species in preterm neonates. J Pediatr Gastroenterol Nutr. (2007) 45(Suppl. 3):S190–4. doi: 10.1097/01.mpg.0000302971.06115.15

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Allonsius CN, van den Broek MFLL, de Boeck I, Kiekens S, Oerlemans EFMM, Kiekens F, et al. Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides. Microb. Biotechnol. (2017) 10:1753–63. doi: 10.1111/1751-7915.12799

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Mailänder-Sánchez D, Braunsdorf C, Grumaz C, Müller C, Lorenz S, Stevens P, et al. Antifungal defense of probiotic Lactobacillus rhamnosus GG is mediated by blocking adhesion and nutrient depletion. PLoS ONE. (2017) 12:e0184438. doi: 10.1371/journal.pone.0184438

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Allonsius CN, Vandenheuvel D, Oerlemans EFM, Petrova MI, Donders GGG, Cos P, et al. Inhibition of Candida albicans morphogenesis by chitinase from Lactobacillus rhamnosus GG. Sci Rep. (2019) 9:1–12. doi: 10.1038/s41598-019-39625-0

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Leão MVP, Tavares TAA, Gonçalves e Silva CR, dos Santos SSF, Junqueira JC, de Oliveira LD, et al. Lactobacillus rhamnosus intake can prevent the development of Candidiasis. Clin Oral Investig. (2018) 22:2511–8. doi: 10.1007/s00784-018-2347-8

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis. (2015) 60:S129–34. doi: 10.1093/cid/civ085

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Vazquez-Munoz R, Dongari-Bagtzoglou A. Anticandidal activities by lactobacillus species: an update on mechanisms of action. Front Oral Health. (2021) 16:2. doi: 10.3389/froh.2021.689382

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Ribeiro FC, Rossoni RD, de Barros PP, Santos JD, Fugisaki LRO, Leão MPV, et al. Action mechanisms of probiotics on Candida spp. and candidiasis prevention: an update. J Appl Microbiol. (2020) 129:175–85. doi: 10.1111/jam.14511

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Keller MK, Brandsborg E, Holmstrøm K, Twetman S. Effect of tablets containing probiotic candidate strains on gingival inflammation and composition of the salivary microbiome: a randomised controlled trial. Benef Microbes. (2018) 9:487–94. doi: 10.3920/BM2017.0104

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Dassi E, Ballarini A, Covello G., Abura'ed A, Anderle M, Asnicar F, et al. Enhanced microbial diversity in the saliva microbiome induced by short-term probiotic intake revealed by 16S rRNA sequencing on the IonTorrent PGM platform. J Biotechnol. (2014) 190:30–9. doi: 10.1016/j.jbiotec.2014.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Dassi E, Ferretti P, Covello G, Speccher A, Migazzi A, Bosco B, et al. The short-term impact of probiotic consumption on the oral cavity microbiome. Sci Rep. (2018) 8:2–9. doi: 10.1038/s41598-018-28491-x

PubMed Abstract | CrossRef Full Text | Google Scholar

49. D'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, et al. The impact of the fungus-host-microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev. (2021) 45:fuaa060. doi: 10.1093/femsre/fuaa060

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Romani L, Zelante T, Palmieri M, Napolioni V, Picciolini M, Velardi A, et al. The cross-talk between opportunistic fungi and the mammalian host via microbiota's metabolism. Semin Immunopathol. (2015) 37:163–71. doi: 10.1007/s00281-014-0464-2

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Sparber F, LeibundGut-Landmann S. Interleukin 17-mediated host defense against candida albicans. Pathogens. (2015) 4:606–19. doi: 10.3390/pathogens4030606

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Conti HR, Bruno VM, Childs EE, Daugherty S, Hunter JP, Mengesha BG, et al. IL-17 Receptor signaling in oral epithelial cells is critical for protection against oropharyngeal candidiasis. Cell Host Microbe. (2016) 20:606–17. doi: 10.1016/j.chom.2016.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Zelante T, Iannitti RG, Cunha C, DeLuca A, Giovannini G, Pieraccini G, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. (2013) 39:372–85. doi: 10.1016/j.immuni.2013.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Lebeer S, Vanderleyden J, de Keersmaecker SCJ. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol. (2010) 8:171–84. doi: 10.1038/nrmicro2297

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Marranzino G, Villena J, Salva S, Alvarez S. Stimulation of macrophages by immunobiotic Lactobacillus strains: influence beyond the intestinal tract. Microbiol Immunol. (2012) 56:771–81. doi: 10.1111/j.1348-0421.2012.00495.x

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Matsubara VH, Ishikawa KH, Ando-Suguimoto ES, Bueno-Silva B, Nakamae AEM, Mayer MPA. Probiotic bacteria alter pattern-recognition receptor expression and cytokine profile in a human macrophage model challenged with Candida albicans and lipopolysaccharide. Front Microbiol. (2017) 8:2280. doi: 10.3389/fmicb.2017.02280

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Rizzo A, Losacco A, Carratelli CR. Lactobacillus crispatus modulates epithelial cell defense against Candida albicans through Toll-like receptors 2 and 4, interleukin 8 and human β-defensins 2 and 3. Immunol Lett. (2013) 156:102–9. doi: 10.1016/j.imlet.2013.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Dongari-Bagtzoglou A, Kashleva H. Development of a highly reproducible three-dimensional organotypic model of the oral mucosa. Nat Protoc. (2006) 1:2012–8. doi: 10.1038/nprot.2006.323

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Tang S, Moyes D, Richardson J, Blagojevic M, Naglik J. Epithelial discrimination of commensal and pathogenic Candida albicans. Oral Dis. (2016) 22:114–9. doi: 10.1111/odi.12395

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Lee X, Vergara C, Lozano CP. Severity of Candida-associated denture stomatitis is improved in institutionalized elders who consume Lactobacillus rhamnosus SP1. Aust Dent J. (2019) 64:229–36. doi: 10.1111/adj.12692

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Saxelin M, Pessi T, Salminen S. Fecal recovery following oral administration of Lactobacilhs Strain GG (ATCC 53103) in gelatine capsules to healthy volunteers. Int J Food Microbiol. (1995) 25:199–203. doi: 10.1016/0168-1605(94)00091-J

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Szajewska H, Kołodziej M, Gieruszczak-Białek D, Skórka A, Ruszczyński M, Shamir R. Systematic review with meta-analysis: Lactobacillus rhamnosus GG for treating acute gastroenteritis in children – a 2019 update. Aliment Pharmacol Ther. (2019) 49:1376–84. doi: 10.1111/apt.15267

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Candida, oropharyngeal candidiasis, probiotics, Lactobacillus, clinical trials, preclinical models

Citation: Archambault LS and Dongari-Bagtzoglou A (2022) Probiotics for Oral Candidiasis: Critical Appraisal of the Evidence and a Path Forward. Front. Oral. Health 3:880746. doi: 10.3389/froh.2022.880746

Received: 21 February 2022; Accepted: 23 March 2022;
Published: 14 April 2022.

Edited by:

Georgios N. Belibasakis, Karolinska Institutet (KI), Sweden

Reviewed by:

Bastiaan P. Krom, VU Amsterdam, Netherlands
Christopher John Nile, Newcastle University, United Kingdom

Copyright © 2022 Archambault and Dongari-Bagtzoglou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Anna Dongari-Bagtzoglou, adongari@uchc.edu

Download