close
Lactobacillus rhamnosus

What is Lactobacillus rhamnosus

Lactobacillus rhamnosus GG (named after the first initials of the last names of two Tufts University researchers, Barry Goldin and Sherwood Gorbach) was the first strain lactic acid producing bacteria belonging to the genus Lactobacillus to be patented in 1989 thanks to its ability to survive and to proliferate at gastric acid pH and in medium containing bile, and to adhere to gut cells 1. Lactobacillus rhamnosus GG (ATCC 53103) was originally isolated from fecal samples of a healthy human adult 2. Lactobacillus rhamnosus is a gram positive, anaerobic bacterium that exhibits the capacity to transport and metabolize carbohydrates and thereby helps in maintaining the epithelial-layer gut integrity 3. Recently, a study demonstrated that Lactobacillus rhamnosus GG-administration alleviates gut inflammation and improved barrier function of intestine 4. Lactobacillus rhamnosus GG was identified as a potential probiotic strain because of its resistance to acid and bile, good growth characteristics and adhesion capacity to the intestinal epithelial layer 5. Probiotics are organisms (bacteria and yeasts) that are taken to improve health. Probiotics are defined as “live microorganisms which, when administered in adequate amounts, confer a health benefit on the host” 6. According to the Dietary Supplement Health and Education Act (DSHEA) of 1994, nutritional supplements, including probiotics, are considered food rather than drugs 7. They are not regulated for clinical application, which results in so-called health claims rather than disease-based claims. The DSHEA requires that dietary supplements meet current good manufacturing practices, but unfortunately, there are substandard products that meet this threshold. Ever since, Lactobacillus rhamnosus GG has been one of the most widely studied probiotic strains, used in a variety of commercially available probiotic products. The beneficial effects of Lactobacillus rhamnosus GG strain have been studied extensively in clinical trials and human intervention studies 2. Furthermore Lactobacillus rhamnosus GG is able to produces both a biofilm that can mechanically protect the mucosa, and different soluble factors beneficial to the gut by enhancing intestinal crypt survival, diminishing apoptosis of the intestinal epithelium, and preserving cytoskeletal integrity.

Lactobacillus rhamnosus probiotic

The microbiome is the collection of all microbes, such as bacteria, fungi, viruses, and their genes, that naturally live on our bodies and inside us. Although microbes require a microscope to see them, they contribute to human health and wellness in many ways. They protect us against pathogens, help our immune system develop, and enable us to digest food to produce energy. Some microbes alter environmental chemicals in ways that make them more toxic, while others act as a buffer and make environmental chemicals less toxic. The intestinal microbiome is composed of microbes that reside in the gut and may be altered by diet, lifestyle, exposure to toxins, and antibiotic use 8. There is a relationship between disease, health, the immune system, and changes in the microbiota 8. Probiotics have an important role in the maintenance of immunologic equilibrium in the gastrointestinal tract through direct interaction with immune cells 9. The microbiome diversity is likely important in health maintenance, and it is likely that broad-spectrum probiotics may increase the effectiveness of treatment. The mechanisms of action of probiotics are complex and likely differ by species 10. Government regulation of probiotics in the United States is complex. Depending on a probiotic product’s intended use, the U.S. Food and Drug Administration (FDA) might regulate it as a dietary supplement, a food ingredient, or a drug. Dietary supplements are regulated by the U.S. Food and Drug Administration’s Center for Food Safety and Applied Nutrition 11. If the probiotics are considered to be drugs for therapeutic purposes, then the product is regulated by the U.S. Food and Drug Administration using Current Good Manufacturing Practices and Investigational New Drug approval processes 12.  Many probiotics are sold as dietary supplements, which don’t require FDA approval before they are marketed. Dietary supplement labels may make claims about how the product affects the structure or function of the body without FDA approval, but they aren’t allowed to make health claims, such as saying the supplement lowers your risk of getting a disease, without the FDA’s consent. If a probiotic is going to be marketed as a drug for treatment of a disease or disorder, it has to meet stricter requirements. It must be proven safe and effective for its intended use through clinical trials and be approved by the FDA before it can be sold.

Probiotic bacteria are proposed to benefit human health mainly by three general mechanisms of action 13. Firstly, certain probiotics can clearly exclude or inhibit pathogens, either through direct action or through influence on the commensal microbiota 14. A second mechanism is the capacity of certain probiotic strains to enhance the epithelial barrier function by modulating signaling pathways, such as nuclear factor-κB (NF-kB), Akt and mitogen- activated protein kinase (MAPK)-dependent pathways, which lead to for example the induction of mucus 15 or increased tight junction functioning 16. Thirdly, most probiotic strains can also modulate host immune responses, exerting strain-specific local and systemic effects 17. Many of the interactions between probiotic bacteria and intestinal epithelial and immune cells are thought to be mediated by molecular structures, known as microbe- associated molecular patterns, which can be recognized through specific pattern recognition receptors such as Toll-like receptors 18. Even though many experimental in vitro data and experiments in animal models validate these mechanisms for probiotic strains in general and for Lactobacillus rhamnosus GG in specific, most published in vivo data in humans pay less attention to mechanisms of action 2.

Lactobacillus rhamnosus benefits

Lactobacillus rhamnosus GG is one of the most widely used probiotic strains. Various health effects are well documented including the prevention and treatment of gastrointestinal infections and diarrhea, and stimulation of immune responses that promote vaccination or even prevent certain allergic symptoms. However, not all intervention studies could show a clinical benefit and even for the same conditions.

Lactobacillus rhamnosus GG has been shown to colonize the gut of newborns significantly better than adults 19. Interestingly, prenatal supplementation with Lactobacillus rhamnosus GG (1.8×1010 colony forming units [CFUs] in capsules, daily from 36th week of gestation) has been reported to change the composition of the neonates microbiota, promoting a beneficial profile dominated by bifidobacteria 20, although the overall microbial diversity did not seem to have changed 21. Others showed that postnatal application of Lactobacillus rhamnosus GG (109 CFU daily, lyophilized powder mixed with breast milk) appears to affect neonatal intestinal colonization patterns causing a higher species diversity compared to placebo 22, although the analysis was not done at detailed level. How exactly Lactobacillus rhamnosus GG could promote the colonization of bifidobacteria in vivo remains to be further explored.

Given its excellent intestinal mucus adherence capacities, Lactobacillus rhamnosus GG has also been often selected as candidate probiotic for the prevention and treatment of gastrointestinal infections and diarrhea, although the efficacy is not uniformly proven. Three subsequent meta-analysis studies have discussed the use of Lactobacillus rhamnosus GG for the treatment of acute diarrhea in children 23, 24, 25. Overall, the current data suggest that Lactobacillus rhamnosus GG can reduce the duration of diarrhea with 1.05 days, particularly in children from geographical Europe, treated with a high dose of Lactobacillus rhamnosus GG (≥1010 CFU/day). Lactobacillus rhamnosus GG (109 CFU daily in fermented milk product) was also shown to reduce the risk of acquiring nosocomial gastrointestinal infections when administered daily in hospitalized children 26. Undernourished Peruvian children showed a lower incidence of diarrhea when treated with Lactobacillus rhamnosus GG (>1010 CFU daily, lyophilized powder mixed with liquid cherry gelatin). Effects were the largest in non-breastfed children 27. However, long-term consumption of milk containing Lactobacillus rhamnosus GG (108 CFU daily) in children attending day care centers in Finland could not show an effect on the incidence of gastro-intestinal symptoms 28. It is important to note that the unsuccessful trial tested a 100-fold lower daily concentration, although other factors such as probiotic formulation and study subject heterogeneity could of course also play a role 2. A recent meta-analysis concluded that Lactobacillus rhamnosus GG treatment can also reduce pain frequency and intensity in children with abdominal pain-related disorders, particularly among irritable bowel syndrome (IBS) patients 29. However, it is important to mention that the clinical effects were significant but moderate 29, which is not unexpected if you consider the large subject heterogeneity in IBS patients.

In other conditions, Lactobacillus rhamnosus GG effects were better stratified. For instance, oral supplementation with Lactobacillus rhamnosus GG (6 × 109 CFU with human milk) has been shown to prevent enteric colonization by Candida species in preterm neonates in a randomized study 30, although the underlying mechanisms need to be explored. Two pilot studies also showed promising results for Lactobacillus rhamnosus GG treatment (respectively 1010 CFU in skim milk and 1.2 × 109 CFU in lyophilized powder daily) of recurrent Clostridium difficile induced colitis in children 31, 32, but this should be repeated in larger trials. Furthermore, application of a commercial yoghurt with Lactobacillus rhamnosus GG to renal patients during 8 weeks has been shown to succeed in clearing vancomycin-resistant enterococci in all patients in an double-blind, randomized, placebo-controlled trial 33. A larger single-blind, randomized, placebo-controlled trial focused on children (3 × 109 CFU daily, dissolved in water or milk) and could show a significant difference between the treated and the control groups only after three weeks 34. The mechanism of clearing of this vancomycin-resistant enterococci remains to be explored, but the SpaCBA pili of Lactobacillus rhamnosus GG share some sequence identity (30-40%) with the pili of Enterococcus faecalis and Enterococcus faecium 35. Clearly, experiments with non-GMO pili deficient variants of Lactobacillus rhamnosus GG would be highly interesting to study their role in the gastro-intestinal and pathogen exclusion effects of Lactobacillus rhamnosus GG.

Although many intervention studies with Lactobacillus rhamnosus GG are targeting the gastrointestinal, it is also interesting to investigate extra-intestinal effects of Lactobacillus rhamnosus GG. For instance, Lactobacillus rhamnosus GG has been shown to reduce oral counts of Streptococcus mutans, a bacterium correlated with caries formation, respectively in yogurt, milk and lozenges 36. Especially long-term consumption of Lactobacillus rhamnosus GG containing milk (5-10 × 105 CFU) appears to be able to reduce tooth decay development in children 37. Of note, there was no effect of short-term consumption of Lactobacillus rhamnosus GG (4×108 CFU daily) on the acidogenicity of plaque nor on caries formation in adults, although it should be noted that Lactobacillus rhamnosus GG was administered in a tablet, which might not be the best formulation 38. Importantly, Lactobacillus rhamnosus GG appears not to ferment sucrose to a significant level 39, indicating that it is itself not cariogenic, a property which is sometimes attributed to lactobacilli due to lactic acid production.

Others have investigated the effect of Lactobacillus rhamnosus GG consumption on respiratory health. For instance, Hojsak and colleagues 26 showed that fermented milk containing Lactobacillus rhamnosus GG was efficient in reducing the risk on respiratory tract infections (RTIs) that lasted longer than three days in hospitalized children. Also, preterm infants treated daily with 109 CFU Lactobacillus rhamnosus GG in capsules starting within one week after birth, appear to have significantly lower incidence of respiratory tract infections and rhinovirus-induced episodes in the first 2 months 40. Furthermore, capsulated Lactobacillus rhamnosus GG (109 CFU) was shown to protect hospitalized patients against ventilator-associated pneumonia, mainly when caused by Gram-negative pathogens like Pseudomonas aeruginosa 41. Moreover, in cystic fibrosis patients colonized with Pseudomonas aeruginosa, long-term Lactobacillus rhamnosus GG treatment (6×109 CFU daily, in oral rehydration solution) significantly decreased the incidence of pulmonary exacerbations and increased body weight 42. Unfortunately, this study did not evaluate Pseudomonas aeruginosa colonization status after Lactobacillus rhamnosus GG treatment. Clearly this area requires further research, because probably a combination of Lactobacillus rhamnosus GG’s antipathogenic and immune modulating capacities determines its potential in respiratory tract infections 2.

Acute infectious diarrhea

Probiotics are effective for acute infectious diarrhea caused by bacteria, but there are inconsistent results for the effectiveness of probiotics for diarrhea caused by viruses. A Cochrane review of 63 randomized controlled trials and quasi-randomized controlled trials included 8,014 infants, children, and adults with acute infectious diarrhea. The researchers found that probiotics significantly reduced the mean duration of diarrhea (25 fewer hours); decreased the risk of diarrhea lasting four or more days by 59%; and led to approximately one fewer stool on day 2 43. For patients with acute infectious diarrhea, probiotics should be started at the onset of symptoms and although there is no evidence to support length of treatment, researchers suggest continuing for one to two weeks following the resolution of symptoms. A meta-analysis of 12 randomized controlled trials with 5,171 participants found a 15% relative decrease in the risk of traveler’s diarrhea with probiotic use 44. For prevention of traveler’s diarrhea, probiotics should be started two days before travel and continued throughout the trip.

A meta-analysis of 17 randomized controlled trials in 2,102 children comparing Saccharomyces boulardii probiotic vs. control for the treatment of acute diarrhea showed a significant reduction in the duration of diarrhea with probiotic use (20 fewer hours) 45. Another meta-analysis of eight randomized controlled trials involving 1,229 children found that Lactobacillus reuteri administration reduced the duration of diarrhea (25 fewer hours) and increased the cure rate on days 1 and 2 46. However, an randomized controlled trial of 646 children with acute watery diarrhea caused predominantly by rotavirus found no significant difference between the group that received Lactobacillus rhamnosus GG probiotics and the control group in the daily frequency of stools, duration of diarrhea, vomiting, or length of hospital stay 47. A meta-analysis of two randomized controlled trials in 201 children with diarrhea from rotavirus found a significant reduction in diarrhea in those treated with Lactobacillus rhamnosus GG vs. placebo (two fewer days) 48.

Antibiotic-associated diarrhea

Probiotics are effective for the prevention and treatment of antibiotic-associated diarrhea in children and adults, and the prevention of Clostridium difficile–associated diarrhea in children and adults; however, there are conflicting results for Clostridium difficile infection. Patients should start probiotics on the first day of antibiotic treatment and continue for one to two weeks following completion of antibiotic therapy. To simplify the treatment regimen, patients may take probiotics at the same time as antibiotics. A Cochrane review of probiotics for the prevention of antibiotic-associated diarrhea in children (23 studies with 3,938 participants) reported that children treated with probiotics vs. control were less likely to have antibiotic-associated diarrhea (absolute risk reduction = 11%; number needed to treat = 10) 49. The Cochrane review review found that Lactobacillus rhamnosus GG or Saccharomyces boulardii at 5 to 40 billion colony-forming units (CFUs) per day was effective, with rare adverse events. In an randomized controlled trial of 333 hospitalized children receiving antibiotics, diarrhea prevalence was lower in children receiving Saccharomyces boulardii probiotics compared with oral rehydration (absolute risk reduction = 21%; number needed to treat = 5). There was also a reduced risk of antibiotic-associated diarrhea, including Clostridium difficile–associated diarrhea and culture-negative diarrhea (absolute risk reduction = 15%; number needed to treat = 7), as well as significantly lower stool frequency, higher recovery rate, and shorter mean duration of diarrhea (2.3 vs. 9.0 days) 50.

A meta-analysis of 63 randomized controlled trials with 11,811 children and adults that compared probiotics with placebo or no treatment reported a significant reduction in the risk of antibiotic-associated diarrhea (number needed to treat = 13) 51. A meta-analysis of adult inpatients showed a reduction in antibiotic-associated diarrhea (15 randomized controlled trials; 2,296 patients; number needed to treat = 11) and a reduction in Clostridium difficile infection (nine randomized controlled trials; 1,099 patients; number needed to treat = 14) among patients randomly assigned to probiotics vs. placebo 52. A Cochrane review of 23 randomized controlled trials reported that probiotics significantly reduced the risk of Clostridium difficile–associated diarrhea vs. placebo (absolute risk reduction = 3.5%; number needed to treat = 29) 53. This review did not find a significant difference in Clostridium difficile infection with probiotics compared with placebo 53. A meta-analysis of 20 randomized controlled trials with 3,818 adults and children demonstrated a significantly decreased risk of Clostridium difficile–associated diarrhea (number needed to treat = 30) 54. A meta-analysis of two low-quality randomized controlled trials including 495 children and adults found no significant effect of yogurt vs. placebo to prevent antibiotic-associated diarrhea 55.

Helicobacter pylori infection

There are inconsistent results on the effectiveness of probiotics as an adjunct to antibiotic therapy to improve Helicobacter pylori eradication rates. A meta-analysis of nine randomized controlled trials involving 1,163 children and adults found that using Lactobacillus-containing probiotics as an adjunct to antibiotics increased the H. pylori eradication rate compared with control (number needed to treat = 10) 56. However, a meta-analysis of 21 randomized controlled trials with 3,452 adults found that probiotics as an adjunct to antibiotics did not improve the eradication of H. pylori infection (odds ratio = 1.44) compared with placebo 57.

Allergic diseases

The potential immunomodulatory effects of Lactobacillus rhamnosus GG that have yet received most attention include its widely discussed effects against allergic disease. In a study published in The Lancet, Kalliomäki and colleagues 58, 59, 60 showed that the combination of prenatal maternal (2-4 weeks) and postnatal pediatric (6 months) Lactobacillus rhamnosus GG treatment (1010 CFU daily, capsules or in water) in families with a history of atopic disease, significantly lowered the risk of eczema at the age of 2, 4 and 7. However, allergic rhinitis and asthma tended to be more common in the Lactobacillus rhamnosus GG treated group and no significant differences were found in incidence of cow milk allergy. Moreover, Kopp and colleagues 61 could not repeat the beneficial results against eczema using a similar protocol and concentration. The reason for these different outcomes is unknown, however it is thought that the different genetic background of the tested populations (Finnish versus German) might play a role. Also, the German trial had more infants with older siblings, which could be a potential cofounder 61. In addition, it seems that different probiotic products have been used for these studies, so that also differences in probiotic formulation, and for instance pili presence, cannot be ruled out.

Atopic dermatitis in children could not be treated by Lactobacillus rhamnosus GG in three independent trials, using a daily concentration of 5×109 CFU/100 ml formula 62, 5×109 CFU in milk 63 or 1010 CFU in milk 64. However, in these trails there was a consistent but not significant effect of Lactobacillus rhamnosus GG in the IgE-sensitized subgroup. Two other trials also reported that treatment (5×109 CFU daily in milk) was efficient in IgE-sensitized infants, but not in non-IgE-sensitized infants 65, 66. This is probably a good example that patient stratification is important to identify potential responders, but more research is necessary to determine the effect of Lactobacillus rhamnosus GG in IgE-sensitized infants.

Related to food allergy, it was reported that administration of capsulated Lactobacillus rhamnosus GG (5×109 CFU) in infants with cow’s milk allergy augments IFN-γ production in stimulated peripheral blood mononuclear cells, thus possibly providing beneficial TH1 immunomodulatory signals 66. Infants acquire more oral tolerance when hydrolyzed casein formula was administered in combination with Lactobacillus rhamnosus GG (107 CFU/ 100 mL) than with the formula alone 67. In milk-hypersensitive adults, Lactobacillus rhamnosus GG (2.6×108 CFU daily in milk) has been shown to reduce the immunoinflammatory response by reducing the expression of specific receptors such as the complement receptors CR1 and CR3 68.

Lactobacillus rhamnosus administration in cancer patients

The most important effect of Lactobacillus rhamnosus GG, observed in cancers patients is linked to the reduction of acute or chronic diarrhea associated with chemotherapy and radiotherapy in cancer patients 69. Diarrhea affects the quality of life of patients as well as the anti-cancer treatment efficacy. Such patients experiencing diarrhea cannot tolerate the standard dose of therapy and need many adjustments in its dose and frequency. This side effect may also require hospitalization caused by the electrolytes loss. Acute or chronic diarrhea is observed among cancer patients treated with pelvic radiation therapy and/or with common antineoplastic agents. In detail, microvascular apoptosis and sclerosis of the vessels may occur during radiation therapy as results radiation-induced gut toxicity 70; while direct epithelial damage may be produced by chemotherapy 71. Drugs used to stop diarrhea, such as loperamide, were not useful to restore the gut microflora. While the appropriate use of probiotics has been shown to be active and safe 69. For instance, Osterlund et al. 72 reported interesting results in the phase 3 randomized study on cancer patients. The authors considered the combination of radio- and chemo-therapy and radiation therapy alone. The Lactobacillus rhamnosus GG was administered as 1–2 capsules/die (1010 CFU) for 24 weeks during any anti-cancer treatment. The reports for diarrhea episode of grades 3 and 4 were 15% lower for those receiving Lactobacillus rhamnosus GG than the control group, suggesting that Lactobacillus rhamnosus GG supplementation may be a well-tolerated product able to ameliorate the compliance to the common cancer therapies 72.

Recently a meta-analysis of 11 randomized and placebo controlled studies evaluating the efficacy and safety of probiotics for prevention of chemo- and radiotherapy-induced diarrhea in people with abdominal and pelvic cancer was published. The study reported a potential beneficial effect of probiotics in the prevention of chemoradiotherapy-induced diarrhea with rare adverse events 69.

Lactobacillus rhamnosus GG as a vaccine adjuvant

Another perhaps more elegant way to investigate the immunomodulatory effects of Lactobacillus rhamnosus GG is by studying its capacity to ameliorate humoral responses to vaccines when applied as an adjuvant. One study showed that the immunogenicity of an oral rotavirus vaccine was significantly ameliorated when mixed with 5×1010 CFU of Lactobacillus rhamnosus GG 73. Lactobacillus rhamnosus GG in milk (1010 CFU daily, 1 week before vaccination, 4 weeks after) was also shown to increase the poliovirus neutralizing antibody titer with a fourfold increase in poliovirus-specific IgA in adults receiving an oral vaccine against polio 1, 2 and 3 74. Moreover, Lactobacillus rhamnosus GG treatment (1010 CFU in a capsule, daily, 28 days starting at vaccination) increased protection rates after an oral life attenuated influenza vaccine. The effect was viral strain-dependent as antibody titers against H1N1 and B strains were low for placebo and Lactobacillus rhamnosus GG-treated groups. For the H3N2 strain, Lactobacillus rhamnosus GG increased protection significantly 75. However, there was no influence on the effect of an oral Salmonella Typhi Ty21a oral vaccine (4×1010 CFU daily, 7 days) 76. In addition, a recent study even showed that maternal supplementation with Lactobacillus rhamnosus GG (1.8 × 1010 CFU daily) from 36 weeks gestation until delivery reduces vaccine-specific immune responses for tetanus, Haemophilus influenzae type b (Hib) and pneumococcal conjugate (PCV7) vaccines in infants at high risk of developing allergic disease 77, indicating that the timing of administration is important if one desires an adjuvant effect and that Lactobacillus rhamnosus GG might not always be the best choice for these purposes. Moreover, van Baarlen et al. 78 showed other lactobacilli such as Lactobacillus plantarum WCFS1 show a more clear modulating of the NF-κB pathway.

Lactobacillus rhamnosus dosage

A Cochrane review found that a dosage of 5 billion colony-forming units or greater per day was significantly more effective than a lower dosage 49. However for disease specific dosage see Lactobacillus rhamnosus benefits section above.

  • Usual adult dose for general use as dietary supplement is 1 capsule per day
  • Usual adult dose for general use concurrently with antibiotics is 1 capsule twice a day throughout antibiotic therapy and for one week after antibiotic therapy
  • Usual adult dose for general use for traveling is 1 capsule twice daily throughout the trip. It is best to start 2 to 3 days prior to travel.
  • Usual Pediatric Dose for Dietary Supplement is one-half to one capsule per day. Open the capsule and stir the contents into a cool drink or mix into baby food or applesauce.

Lactobacillus rhamnosus side effects

The use of Lactobacillus rhamnosus GG in a wide variety of clinical trials without serious adverse events confirmed its safety. Common side effects of Lactobacillus rhamnosus may include stomach bloating or gas. Lactobacillus rhamnosus GG has been administered to, among others, low birth weight infants 79, pregnant women 80, HIV- infected patients 81 and patients with a mechanical ventilator 41. To support the safety of Lactobacillus rhamnosus GG, it was shown that despite increasing Lactobacillus rhamnosus GG consumption in Finland and Sweden respectively, the rate of Lactobacillus bacteremia remained constant 82.

However, case reports show that probiotic therapy is to be discouraged in certain groups of patients. In a review concerning the safety of probiotics, it was concluded that adverse effects of probiotics were correlated with (i) impaired intestinal barrier function, (ii) immune compromised state and (iii) central venous catheter 83. Indeed, cases specifically reported for Lactobacillus rhamnosus GG show that these risk factors might play a role. For instance, a number of infants treated with Lactobacillus rhamnosus GG for short gut syndrome associated with intestinal friability seemed to manifest sepsis with Lactobacillus rhamnosus GG-like bacteria 84 and an ulcerative colitis patient was diagnosed with Lactobacillus rhamnosus GG bacteremia 85, although these identifications were not done at a detailed genomic level. Nevertheless, in patients with a seriously compromised integrity of the barrier function of the intestine, the administration of specific Lactobacillus rhamnosus GG molecules is probably a better strategy than living Lactobacillus rhamnosus GG cells, because of the risk for translocation of the bacteria from the intestines into the blood 2. The long-term effects of probiotics are largely unknown, and additional randomized trials are needed to address this question 86.

References
  1. Capurso L. Thirty Years of Lactobacillus rhamnosus GG: A Review. J Clin Gastroenterol. 2019 Mar;53 Suppl 1:S1-S41. doi: 10.1097/MCG.0000000000001170
  2. Segers, M. E., & Lebeer, S. (2014). Towards a better understanding of Lactobacillus rhamnosus GG–host interactions. Microbial cell factories, 13 Suppl 1(Suppl 1), S7. https://doi.org/10.1186/1475-2859-13-S1-S7
  3. Westerik N, Kort R, Sybesma W, Reid G. Lactobacillus rhamnosus Probiotic Food as a Tool for Empowerment Across the Value Chain in Africa. Front Microbiol. 2018 Jul 10;9:1501. doi: 10.3389/fmicb.2018.01501
  4. Mao J, Qi S, Cui Y, Dou X, Luo XM, Liu J, Zhu T, Ma Y, Wang H. Lactobacillus rhamnosus GG Attenuates Lipopolysaccharide-Induced Inflammation and Barrier Dysfunction by Regulating MAPK/NF-κB Signaling and Modulating Metabolome in the Piglet Intestine. J Nutr. 2020 May 1;150(5):1313-1323. doi: 10.1093/jn/nxaa009
  5. Doron S, Snydman DR, Gorbach SL. Lactobacillus GG: bacteriology and clinical applications. Gastroenterol Clin North Am. 2005 Sep;34(3):483-98, ix. https://doi.org/10.1016/j.gtc.2005.05.011
  6. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, Calder PC, Sanders ME. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014 Aug;11(8):506-14. doi: 10.1038/nrgastro.2014.66
  7. Dietary Supplement Health and Education Act of 1994. Public Law 103-417, 103rd Congress. https://ods.od.nih.gov/About/DSHEA_Wording.aspx
  8. Logan AC, Jacka FN, Prescott SL. Immune-microbiota interactions: dysbiosis as a global health issue. Curr Allergy Asthma Rep. 2016;16(2):13.
  9. Ricci A, Tagliacarne SC, Valsecchi C, et al. Probiotics and inflammatory bowel diseases. J Biol Regul Homeost Agents. 2015;29(2 suppl 1):96–113.
  10. Probiotics for Gastrointestinal Conditions: A Summary of the Evidence. Am Fam Physician. 2017 Aug 1;96(3):170-178. https://www.aafp.org/afp/2017/0801/p170.html
  11. Dietary Supplement Products & Ingredients. https://www.fda.gov/food/dietary-supplements/dietary-supplement-products-ingredients
  12. Development & Approval Process | Drugs. https://www.fda.gov/drugs/development-approval-process-drugs
  13. Lebeer S, Vanderleyden J, De Keersmaecker SC. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol. 2010;8:171–184. doi: 10.1038/nrmicro2297
  14. Corr SC, Hill C, Gahan CG. Understanding the mechanisms by which probiotics inhibit gastrointestinal pathogens. Adv Food Nutr Res. 2009;56:1–15.
  15. Mack DR, Ahrne S, Hyde L, Wei S, Hollingsworth MA. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut. 2003;52:827–833. doi: 10.1136/gut.52.6.827
  16. Rao RK, Samak G. Protection and Restitution of Gut Barrier by Probiotics: Nutritional and Clinical Implications. Curr Nutr Food Sci. 2013;9:99–107. doi: 10.2174/1573401311309020004
  17. Wells JM. Immunomodulatory mechanisms of lactobacilli. Microb Cell Fact. 2011;10(Suppl 1):S17. doi: 10.1186/1475-2859-10-S1-S17
  18. Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nature Reviews Immunology. 2010;10:131–144. doi: 10.1038/nri2707
  19. Sepp E, Mikelsaar M, Salminen S. Effect of Administration of Lactobacillus-Casei Strain Gg on the Gastrointestinal Microbiota of Newborns. Microbial Ecology in Health and Disease. 1993;6:309–314. doi: 10.3109/08910609309141340
  20. Lahtinen SJ, Boyle RJ, Kivivuori S, Oppedisano F, Smith KR, Robins-Browne R, Salminen SJ, Tang ML. Prenatal probiotic administration can influence Bifidobacterium microbiota development in infants at high risk of allergy. J Allergy Clin Immunol. 2009;123:499–501. doi: 10.1016/j.jaci.2008.11.034
  21. Ismail IH, Oppedisano F, Joseph SJ, Boyle RJ, Robins-Browne RM, Tang ML. Prenatal administration of Lactobacillus rhamnosus has no effect on the diversity of the early infant gut microbiota. Pediatr Allergy Immunol. 2012;23:255–258. doi: 10.1111/j.1399-3038.2011.01239.x
  22. Agarwal R, Sharma N, Chaudhry R, Deorari A, Paul VK, Gewolb IH, Panigrahi P. Effects of oral Lactobacillus GG on enteric microflora in low-birth-weight neonates. J Pediatr Gastroenterol Nutr. 2003;36:397–402. doi: 10.1097/00005176-200303000-00019
  23. Szajewska H, Skorka A, Ruszczynski M, Gieruszczak-Bialek D. Meta-analysis: Lactobacillus GG for treating acute diarrhoea in children. Aliment Pharmacol Ther. 2007;25:871–881. doi: 10.1111/j.1365-2036.2007.03282.x
  24. Szajewska H, Wanke M, Patro B. Meta-analysis: the effects of Lactobacillus rhamnosus GG supplementation for the prevention of healthcare-associated diarrhoea in children. Aliment Pharmacol Ther. 2011;34:1079–1087. doi: 10.1111/j.1365-2036.2011.04837.x
  25. Szajewska H, Skorka A, Ruszczynski M, Gieruszczak-Bialek D. Meta-analysis: Lactobacillus GG for treating acute gastroenteritis in children – updated analysis of randomised controlled trials. Aliment Pharmacol Ther. 2013;38:467–476. doi: 10.1111/apt.12403
  26. Hojsak I, Abdovic S, Szajewska H, Milosevic M, Krznaric Z, Kolacek S. Lactobacillus GG in the Prevention of Nosocomial Gastrointestinal and Respiratory Tract Infections. Pediatrics. 2010;125:E1171–E1177. doi: 10.1542/peds.2009-2568
  27. Oberhelman RA, Gilman RH, Sheen P, Taylor DN, Black RE, Cabrera L, Lescano AG, Meza R, Madico G. A placebo-controlled trial of Lactobacillus GG to prevent diarrhea in undernourished Peruvian children. J Pediatr. 1999;134:15–20. doi: 10.1016/S0022-3476(99)70366-5
  28. Kumpu M, Kekkonen RA, Kautiainen H, Jarvenpaa S, Kristo A, Huovinen P, Pitkaranta A, Korpela R, Hatakka K. Milk containing probiotic Lactobacillus rhamnosus GG and respiratory illness in children: a randomized, double-blind, placebo-controlled trial. Eur J Clin Nutr. 2012;66:1020–1023. doi: 10.1038/ejcn.2012.62
  29. Horvath A, Dziechciarz P, Szajewska H. Meta-analysis: Lactobacillus rhamnosus GG for abdominal pain-related functional gastrointestinal disorders in childhood. Aliment Pharmacol Ther. 2011;33:1302–1310. doi: 10.1111/j.1365-2036.2011.04665.x
  30. Manzoni P, Mostert M, Leonessa ML, Priolo C, Farina D, Monetti C, Latino MA, Gomirato G. Oral supplementation with Lactobacillus casei subspecies rhamnosus prevents enteric colonization by Candida species in preterm neonates: a randomized study. Clin Infect Dis. 2006;42:1735–1742. doi: 10.1086/504324
  31. Gorbach SL, Chang TW, Goldin B. Successful Treatment of Relapsing Clostridium Difficile Colitis with Lactobacillus Gg. Lancet. 1987;2:1519–1519.
  32. Biller JA, Katz AJ, Flores AF, Buie TM, Gorbach SL. Treatment of Recurrent Clostridium-Difficile Colitis with Lactobacillus Gg. J Pediatr Gastroenterol Nutr. 1995;21:224–226. doi: 10.1097/00005176-199508000-00016
  33. Manley KJ, Fraenkel MB, Mayall BC, Power DA. Probiotic treatment of vancomycin-resistant enterococci: A randomised controlled trial. Medical Journal of Australia. 2007;186:454–457.
  34. Szachta P, Ignys I, Cichy W. An Evaluation of the Ability of the Probiotic Strain Lactobacillus rhamnosus GG to Eliminate the Gastrointestinal Carrier State of Vancomycin-resistant Enterococci in Colonized Children. J Clin Gastroenterol. 2011;45:872–877. doi: 10.1097/MCG.0b013e318227439f
  35. Lebeer S, De Keersmaecker SC, Verhoeven TL, Fadda AA, Marchal K, Vanderleyden J. Functional analysis of luxS in the probiotic strain Lactobacillus rhamnosus GG reveals a central metabolic role important for growth and biofilm formation. J Bacteriol. 2007;189:860–871. doi: 10.1128/JB.01394-06
  36. Toiviainen A, Jalasvuori H, Lahti E, Gursoy U, Salminen S, Fontana M, Flannagan S, Eckert G, Kokaras A, Paster B, Söderling E. Impact of orally administered lozenges with Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 on the number of salivary mutans streptococci, amount of plaque, gingival inflammation and the oral microbiome in healthy adults. Clin Oral Investig. 2014.
  37. Nase L, Hatakka K, Savilahti E, Saxelin M, Ponka A, Poussa T, Korpela R, Meurman JH. Effect of long-term consumption of a probiotic bacterium, Lactobacillus rhamnosus GG, in milk on dental caries and caries risk in children. Caries Res. 2001;35:412–420. doi: 10.1159/000047484
  38. Marttinen A, Haukioja A, Karjalainen S, Nylund L, Satokari R, Ohman C, Holgerson P, Twetman S, Soderling E. Short-term consumption of probiotic lactobacilli has no effect on acid production of supragingival plaque. Clin Oral Investig. 2012;16:797–803. doi: 10.1007/s00784-011-0584-1
  39. Kankainen M, Paulin L, Tynkkynen S, von Ossowski I, Reunanen J, Partanen P, Satokari R, Vesterlund S, Hendrickx APA, Lebeer S. et al.Comparative genomic analysis of Lactobacillus rhamnosus GG reveals pili containing a human- mucus binding protein. Proceedings of the National Academy of Sciences. 2009;106:17193–17198. doi: 10.1073/pnas.0908876106
  40. Luoto R, Ruuskanen O, Waris M, Kalliomaki M, Salminen S, Isolauri E. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: A randomized, placebo-controlled trial. J Allergy Clin Immunol. 2013.
  41. Morrow LE, Kollef MH, Casale TB. Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med. 2010;182:1058–1064. doi: 10.1164/rccm.200912-1853OC
  42. Bruzzese E, Raia V, Spagnuolo MI, Volpicelli M, De Marco G, Maiuri L, Guarino A. Effect of Lactobacillus GG supplementation on pulmonary exacerbations in patients with cystic fibrosis: A pilot study. Clinical Nutrition. 2007;26:322–328. doi: 10.1016/j.clnu.2007.01.004
  43. Allen SJ, et al. Probiotics for treating acute infectious diarrhoea. Cochrane Database Syst Rev. 2010;(11):CD003048.
  44. McFarland LV. Meta-analysis of probiotics for the prevention of traveler’s diarrhea. Travel Med Infect Dis. 2007;5(2):97–105.
  45. Feizizadeh S, Salehi-Abargouei A, Akbari V. Efficacy and safety of Saccharomyces boulardii for acute diarrhea. Pediatrics. 2014;134(1):e176–e191.
  46. Urbańska M, Gieruszczak-Białek D, Szajewska H. Systematic review with meta-analysis: Lactobacillus reuteri DSM 17938 for diarrhoeal diseases in children. Aliment Pharmacol Ther. 2016;43(10):1025–1034.
  47. Basu S, Chatterjee M, Ganguly S, Chandra PK. Efficacy of Lactobacillus rhamnosus GG in acute watery diarrhoea of Indian children: a randomised controlled trial. J Paediatr Child Health. 2007;43(12):837–842.
  48. Szajewska H, et al. Meta-analysis: Lactobacillus GG for treating acute gastroenteritis in children—updated analysis of randomised controlled trials. Aliment Pharmacol Ther. 2013;38(5):467–476.
  49. Goldenberg JZ, et al. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev. 2015;(12):CD004827.
  50. Shan LS, Hou P, Wang ZJ, et al. Prevention and treatment of diarrhoea with Saccharomyces boulardii in children with acute lower respiratory tract infections. Benef Microbes. 2013;4(4):329–334.
  51. Hempel S, Newberry SJ, Maher AR, et al. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA. 2012;307(18):1959–1969.
  52. Pattani R, et al. Probiotics for the prevention of antibiotic-associated diarrhea and Clostridium difficile infection among hospitalized patients: systematic review and meta-analysis. Open Med. 2013;7(2):e56–e67.
  53. Goldenberg JZ, Ma SS, Saxton JD, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2013;5(5):CD006095.
  54. Johnston BC, Ma SS, Goldenberg JZ, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea: a systematic review and meta-analysis. Ann Intern Med. 2012;157(12):878–888.
  55. Patro-Golab B, et al. Yogurt for treating antibiotic-associated diarrhea: systematic review and meta-analysis [published correction appears in Nutrition. 2015;31(7–8):1060]. Nutrition. 2015;31(6):796–800.
  56. Zheng X, Lyu L, Mei Z. Lactobacillus-containing probiotic supplementation increases Helicobacter pylori eradication rate: evidence from a meta-analysis. Rev Esp Enferm Dig. 2013;105(8):445–453.
  57. Lu C, Sang J, He H, et al. Probiotic supplementation does not improve eradication rate of Helicobacter pylori infection compared to placebo based on standard therapy: a meta-analysis. Sci Rep. 2016;6:23522.
  58. Kalliomaki M, Salminen S, Poussa T, Isolauri E. Probiotics during the first 7 years of life: a cumulative risk reduction of eczema in a randomized, placebo- controlled trial. J Allergy Clin Immunol. 2007;119:1019–1021. doi: 10.1016/j.jaci.2006.12.608
  59. Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet. 2003;361:1869–1871. doi: 10.1016/S0140-6736(03)13490-3
  60. Kalliomaki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357:1076–1079. doi: 10.1016/S0140-6736(00)04259-8
  61. Kopp MV, Hennemuth I, Heinzmann A, Urbanek R. Randomized, double-blind, placebo-controlled trial of probiotics for primary prevention: no clinical effects of Lactobacillus GG supplementation. Pediatrics. 2008;121:e850–856. doi: 10.1542/peds.2007-1492
  62. Brouwer ML, Wolt-Plompen SA, Dubois AE, van der Heide S, Jansen DF, Hoijer MA, Kauffman HF, Duiverman EJ. No effects of probiotics on atopic dermatitis in infancy: a randomized placebo-controlled trial. Clin Exp Allergy. 2006;36:899–906. doi: 10.1111/j.1365-2222.2006.02513.x
  63. Gruber C, Wendt M, Sulser C, Lau S, Kulig M, Wahn U, Werfel T, Niggemann B. Randomized, placebo-controlled trial of Lactobacillus rhamnosus GG as treatment of atopic dermatitis in infancy. Allergy. 2007;62:1270–1276. doi: 10.1111/j.1398-9995.2007.01543.x
  64. Folster-Holst R, Muller F, Schnopp N, Abeck D, Kreiselmaier I, Lenz T, von Ruden U, Schrezenmeir J, Christophers E, Weichenthal M. Prospective, randomized controlled trial on Lactobacillus rhamnosus in infants with moderate to severe atopic dermatitis. Br J Dermatol. 2006;155:1256–1261. doi: 10.1111/j.1365-2133.2006.07558.x
  65. Viljanen M, Savilahti E, Haahtela T, Juntunen-Backman K, Korpela R, Poussa T, Tuure T, Kuitunen M. Probiotics in the treatment of atopic eczema/dermatitis syndrome in infants: a double-blind placebo-controlled trial. Allergy. 2005;60:494–500. doi: 10.1111/j.1398-9995.2004.00514.x
  66. Pohjavuori E, Viljanen M, Korpela R, Kuitunen M, Tiittanen M, Vaarala O, Savilahti E. Lactobacillus GG effect in increasing IFN-gamma production in infants with cow’s milk allergy. J Allergy Clin Immunol. 2004;114:131–136. doi: 10.1016/j.jaci.2004.03.036
  67. Berni Canani R, Nocerino R, Terrin G, Frediani T, Lucarelli S, Cosenza L, Passariello A, Leone L, Granata V, Di Costanzo M. et al.Formula selection for management of children with cow’s milk allergy influences the rate of acquisition of tolerance: a prospective multicenter study. J Pediatr. 2013;163:771–777 e771. doi: 10.1016/j.jpeds.2013.03.008
  68. Pelto L, Isolauri E, Lilius EM, Nuutila J, Salminen S. Probiotic bacteria down- regulate the milk-induced inflammatory response in milk-hypersensitive subjects but have an immunostimulatory effect in healthy subjects. Clin Exp Allergy. 1998;28:1474–1479. doi: 10.1046/j.1365-2222.1998.00449.x
  69. Wang, Y. H., Yao, N., Wei, K. K., Jiang, L., Hanif, S., Wang, Z. X., et al. (2016). The efficacy and safety of probiotics for prevention of chemoradiotherapy-induced diarrhea in people with abdominal and pelvic cancer: a systematic review and meta-analysis. Eur. J. Clin. Nutr. 70, 1246–1253. doi: 10.1038/ejcn.2016.102
  70. Stansborough, R. L., Bateman, E. H., Al-Dasooqi, N., Bowen, J. M., Keefe, D. M. K., Yeoh, A. S. J., et al. (2017). Fractionated abdominal irradiation induces intestinal microvascular changes in an in vivo model of radiotherapy-induced gut toxicity. Support. Care Cancer 25, 1973–1983. doi: 10.1007/s00520-017-3601-3
  71. Van Sebille, Y. Z., Stansborough, R., Wardill, H. R., Bateman, E., Gibson, R. J., and Keefe, D. M. (2015). Management of mucositis during chemotherapy: from pathophysiology to pragmatic therapeutics. Curr. Oncol. Rep. 17, 50. doi: 10.1007/s11912-015-0474-9
  72. Osterlund, P., Ruotsalainen, T., Korpela, R., Saxelin, M., Ollus, A., Valta, P., et al. (2007). Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: a randomised study. Br. J. Cancer 97, 1028–1034. doi: 10.1038/sj.bjc.6603990
  73. Isolauri E, Joensuu J, Suomalainen H, Luomala M, Vesikari T. Improved immunogenicity of oral D × RRV reassortant rotavirus vaccine by Lactobacillus casei GG. Vaccine. 1995;13:310–312. doi: 10.1016/0264-410X(95)93319-5
  74. de Vrese M, Rautenberg P, Laue C, Koopmans M, Herremans T, Schrezenmeir J. Probiotic bacteria stimulate virus-specific neutralizing antibodies following a booster polio vaccination. European Journal of Nutrition. 2005;44:406–413. doi: 10.1007/s00394-004-0541-8
  75. Davidson LE, Fiorino AM, Snydman DR, Hibberd PL. Lactobacillus GG as an immune adjuvant for live-attenuated influenza vaccine in healthy adults: a randomized double-blind placebo-controlled trial. Eur J Clin Nutr. 2011;65:501–507. doi: 10.1038/ejcn.2010.289
  76. Fang H, Elina T, Heikki A, Seppo S. Modulation of humoral immune response through probiotic intake. FEMS Immunol Med Microbiol. 2000;29:47–52. doi: 10.1111/j.1574-695X.2000.tb01504.x
  77. Boyle RJ, Ismail IH, Kivivuori S, Licciardi PV, Robins-Browne RM, Mah LJ, Axelrad C, Moore S, Donath S, Carlin JB. et al.Lactobacillus GG treatment during pregnancy for the prevention of eczema: a randomized controlled trial. Allergy. 2011;66:509–516. doi: 10.1111/j.1398-9995.2010.02507.x
  78. Van Baarlen P, Troost FJ, Van der Meer C, Hooiveld GJ, Boekschoten M, Brummer RJ, Kleerebezem M. Human mucosal in vivo transcriptome responses to three lactobacilli indicate how probiotics may modulate human cellular pathways. PNAS. 2011;108:4562–4569. doi: 10.1073/pnas.1000079107
  79. Manzoni P, Lista G, Gallo E, Marangione P, Priolo C, Fontana P, Guardione R, Farina D. Routine Lactobacillus rhamnosus GG administration in VLBW infants: a retrospective, 6-year cohort study. Early Human Development. 2011;87(Suppl 1):S35–38.
  80. Boyle RJ, Mah LJ, Chen A, Kivivuori S, Robins-Browne RM, Tang ML. Effects of Lactobacillus GG treatment during pregnancy on the development of fetal antigen-specific immune responses. Clin Exp Allergy. 2008;38:1882–1890. doi: 10.1111/j.1365-2222.2008.03100.x
  81. Salminen MK, Tynkkynen S, Rautelin H, Poussa T, Saxelin M, Ristola M, Valtonen V, Jarvinen A. The efficacy and safety of probiotic Lactobacillus rhamnosus GG on prolonged, noninfectious diarrhea in HIV Patients on antiretroviral therapy: a randomized, placebo-controlled, crossover study. HIV Clin Trials. 2004;5:183–191. doi: 10.1310/6F83-N39Q-9PPP-LMVV
  82. Sullivan A, Nord CE. Probiotic lactobacilli and bacteraemia in Stockholm. Scand J Infect Dis. 2006;38:327–331. doi: 10.1080/00365540500449826
  83. Sanders ME, Akkermans LMA, Haller D, Hammerman C, Heimbach J, Hörmannsperger G, Huys G, Levy DD, Lutgendorff F, Mack D. et al.Safety assessment of probiotics for human use. Gut Microbes. 2010;1:164–185. doi: 10.4161/gmic.1.3.12127
  84. De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. Pediatr Infect Dis J. 2005;24:278–280. doi: 10.1097/01.inf.0000154588.79356.e6
  85. Farina C, Arosio M, Mangia M, Moioli F. Lactobacillus casei subsp. rhamnosus sepsis in a patient with ulcerative colitis. J Clin Gastroenterol. 2001;33:251–252. doi: 10.1097/00004836-200109000-00019
  86. Hempel S, et al. Safety of probiotics used to reduce risk and prevent or treat disease. Evid Rep Technol Assess (Full Rep). 2011;(200):1–645.
Health Jade Team

The author Health Jade Team

Health Jade