ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Prebiotics and probiotics for prevention of allergic disease

Prebiotics and probiotics for prevention of allergic disease
Literature review current through: Jan 2024.
This topic last updated: Mar 25, 2023.

INTRODUCTION — There is increasing evidence that disturbances in gut microbial composition play a role in the pathophysiology of immune-mediated disorders such as allergic disease [1]. Gut microbiota are key players in the early development of both local immune maturation and systemic immune programming. Differences in perinatal colonization patterns with both the level of industrialization and subsequent allergic outcomes provided a strong foundation for intervention studies designed to modify postnatal colonization in the prevention of allergic disease [2,3]. Consequently, the effects of beneficial bacteria (probiotics) or resistant starches or fiber (prebiotics) that selectively stimulate a limited number of beneficial bacteria have been evaluated in allergy prevention studies.

This topic reviews the evidence for prebiotics and probiotics in the prevention of allergic disease [4-11]. The use of prebiotics and probiotics for the treatment of allergic disease is discussed in detail separately. (See "Prebiotics and probiotics for treatment of allergic disease".)

The role of microbiota in the pathogenesis of food allergy and the use of probiotics for gastrointestinal diseases are discussed elsewhere. A detailed review of intestinal microbiota is also presented separately. (See "Pathogenesis of food allergy", section on 'Gut and skin microbiota' and "Probiotics for gastrointestinal diseases" and "Spatial organization of intestinal microbiota in health and disease" and "Evidence-based approach to prevention".)

DEFINITIONS — The terms "prebiotics," "probiotics," and "synbiotics" are defined as follows:

"Prebiotics" generally refers to "a substrate that is selectively used by host microorganisms conferring a health benefit" [12]. Prebiotics are typically comprised of nondigestible carbohydrates but can also include noncarbohydrates such as fatty acids, phenolics, and phytochemicals (figure 1) [12]. The most widely accepted prebiotics are the fermentable oligosaccharides inulin, fructooligosaccharides (FOS or fructans), galactooligosaccharides (GOS), and lactulose (table 1).

"Probiotics" is a general term, analogous to the term "antibiotics," for different strains and species of microorganisms with a wide and varying range of clinical and immunologic capacities. The United Nations Food and Agriculture Organization (FAO) and World Health Organization (WHO) have defined probiotics as "live microorganisms, which, when administered in adequate amounts, confer a health benefit on the host" [13]. Guidelines from a FAO/WHO expert group are available regarding evaluation of probiotics, including data necessary to substantiate health claims [14]. The most commonly used probiotics are lactobacilli and bifidobacteria strains, but other microorganisms have been used as probiotics, including the yeast Saccharomyces boulardii.

"Synbiotics" refers to a combination of prebiotics and probiotics.

BACKGROUND — The prevalence of allergic rhinitis increased in the first half of the 20th century, followed by an increase in asthma prevalence and then food allergies. The environmental changes that are driving the allergy epidemic are not fully identified, although the hygiene hypothesis makes a strong case that the reduction in general microbial exposure during early childhood [15], with corresponding changes in commensal microbiota [16], plays an important role. Normal immune homeostasis is dependent upon gut microbiota and may be influenced by differences in colonization patterns. The hygiene hypothesis provides the conceptual framework for pre- and/or postnatal dietary supplementation to prevent allergic disease by modifying early gut microbial composition, even if this hypothesis does not decisively explain all observations or provide definitive guidance on how to limit the allergy epidemic. (See "Increasing prevalence of asthma and allergic rhinitis and the role of environmental factors", section on 'Epidemiologic history' and "Food allergy in children: Prevalence, natural history, and monitoring for resolution", section on 'Prevalence of childhood food allergy'.)

Rationale for prebiotics and probiotics in allergic disease — Epidemiologic associations between declining microbial exposure and allergic disease [15] are supported by animal models demonstrating that early exposure to pathogenic or nonpathogenic microbial products can either prevent or modify allergic responses [17]. Gut microbiota confer specific immune-protective effects that are probably mediated through complex pathways within (and potentially even beyond) the gut-associated lymphoid tissue (GALT), the largest immune "organ" in humans. These effects include altered local immunoglobulin A (IgA) production and induction of tolerogenic dendritic cells and regulatory T cell populations, with production of immunomodulatory cytokines, such as interleukin (IL) 10 and transforming growth factor (TGF) beta [18]. These mechanisms appear to collectively inhibit local inflammation, improve gut barrier mechanisms, and consequently reduce the risk of inappropriate systemic immune responses.

Over the first months of life, there is an enormous shift from the "sterile" milieu in utero to microbial colonization with more than 1014 organisms, outnumbering total human host cells by more than 10:1. During the same period, the functionally immature neonatal immune system must also develop to attain a complex and finely tuned balance between host defense and immune tolerance. Studies in germ-free animals demonstrate that oral tolerance cannot be induced in the absence of gut microbiota [19]. Although full immune function and oral tolerance can be restored by introducing normal intestinal microbiota, this is age dependent and cannot be achieved in mature animals [19]. These observations underline the essential role of gut microbiota in the development of the immune system and exemplify the importance of the timing of initial colonization. (See "Pathogenesis of food allergy", section on 'The gut immune system'.)

A number of human studies have shown differences in the early colonization patterns of infants who develop allergic disease [20]. Early studies reported that low levels of bifidobacteria and early colonization with potentially pathogenic bacteria, such as Clostridioides difficile [2,3] and Staphylococcus aureus [2], were more prevalent in children who subsequently developed allergy. Subsequent larger, prospective studies have tried to link certain bacterial genera or species to the development of allergy with conflicting results [21,22]. Consequently, it has been suggested that a high gut microbial diversity might be more important than the absence or presence of specific genera or species in the context of immune system maturation and subsequent development of immune-mediated disorders. This view is supported by prospective studies that demonstrated reduced gut microbial diversity early in life in infants who later developed allergic manifestations [23-25].

Proposed mechanisms — The exact mechanisms are not clear, but both prebiotics and probiotics probably influence immune development through a number of different pathways that can be modulated by host and environmental factors. In addition, differences in the probiotic supplement, such as the strain, dose, viability, timing, duration, adherence, and method of administration, may result in differences in clinical effect.

Prebiotics — Prebiotic carbohydrates are a major substrate for bacterial growth, selectively stimulating the growth and/or activity of beneficial members of the gut microbiota, particularly bifidobacteria [26,27]. Thus, prebiotics could, at least in theory, have more global effects on colonization than adding a single probiotic strain. A second, more direct immune effect appears to be mediated by the fermentation products of prebiotics. Gut microorganisms ferment prebiotics to produce short-chain fatty acids (SCFAs) that have direct antiinflammatory effects [28]. SCFAs also promote intestinal integrity through effects on epithelial cell proliferation and differentiation [29]. Animal and human studies suggest that prebiotics may directly affect both mucosal and systemic immunity [30-32]. However, more studies are needed to confirm that these are clinically relevant effects.

Probiotics — Numerous mechanisms have been proposed to explain the potential effects of probiotics based upon evidence from in vitro studies and animal models [18]. Probiotics may alter gut microbial composition directly, or they may act indirectly by effects on microbial products, host products, or food components or by modulation of the host's immune system. Commensal gut bacteria can decrease local inflammation [33] and may stabilize intestinal barrier integrity [34], potentially reducing systemic antigen load. Certain strains of lactobacilli and bifidobacteria may influence immune function through effects on enterocytes, antigen-presenting cells (including both circulating monocytes and local dendritic cells), regulatory T cells, and effector T and B cells [18]. However, it is uncertain how (or if) these observations translate into clinically relevant effects. In addition, it is also not known to what degree the effects of a specific bacterial strain are similar to other strains, even of the same species. Finally, the immune-stimulating effects of probiotics in in vitro studies can be divergent from those observed in vivo [35]. More studies are needed to clarify these issues.

Modulating factors — The effects of prebiotics and probiotics are probably influenced by a range of host and environmental factors [18]. Host factors which contribute to this variability include variation in genetic predisposition to allergic disease (across multiple loci) and specific polymorphisms in microbial recognition pathways that may influence effects and susceptibility to colonization. A range of environmental factors, such as maternal microbiota, general microbial burden, mode of delivery, feeding practices and weaning diet, antibiotics, and other immunomodulatory influences may have secondary effects on colonization.

RECOMMENDATIONS FROM GUIDELINES — Many experts bodies such as the European Academy of Allergy and Clinical Immunology (EAACI) Food Allergy and Anaphylaxis Guidelines do not recommend using prebiotics, probiotics, or synbiotics for the prevention of any allergic condition, because of to the great heterogeneity in the studies and variability in results [36,37]. Others, such as the US National Institute of Allergy and Infectious Diseases (NIAID) have not given any specific recommendations on these products in their guidelines [38].

In contrast, the World Allergy Organization (WAO) suggests using prebiotics only in infants who are not exclusively breastfed [39] and probiotics in pregnant and lactating women and in infants when there is high risk of allergy in the children (defined as presence of a biologic parent or sibling with asthma, allergic rhinitis, eczema, or food allergy) [40]. The probiotic recommendation was based upon their analysis of the data that found a net benefit of prevention of eczema, but not any other allergic outcome, with probiotic treatment. In addition, the risk of possible adverse events was felt to be low.

The timing, duration, and choice of probiotic (strain and dose) are not specified in the WAO guidelines. Probiotics were given in the last four to six weeks of pregnancy in most of the studies reviewed, but there was much greater variability in timing and duration of postnatal therapy in the infant and/or breastfeeding mother. In addition, the only probiotic strain with reproducible data is Lactobacillus rhamnosus GG (LGG). (See 'Probiotics' below.)

OUR APPROACH — Meta-analysis data suggest that there is a modest preventive effect of probiotics on the development of eczema, but not other atopic diseases, in at-risk infants (defined as presence of a biologic parent or sibling with asthma, allergic rhinitis, eczema, or food allergy) [40]. However, the great heterogeneity of the studies makes it difficult to advise on specifics regarding therapy (eg, strains, dose, timing, and duration). Thus, we suggest not giving probiotics during pregnancy, lactation, and infancy for the prevention of eczema. We do not discourage this approach, however, if parents/caregivers are strongly interested, given the low risk of adverse events. Parents/caregivers who chose this approach should first discuss it with the appropriate clinician (eg, obstetrician, pediatrician, other primary care provider).

PROBIOTICS — Several meta-analyses have generally concurred that probiotics reduce the risk of eczema but not other allergic outcomes (when examined) [5,41-48]. However, more specific conclusions have varied due in part to the wide heterogeneity of the studies included. Further clinical studies are needed to identify if specific dosing regimens, as well as specific strains or combinations, are more effective than others. In addition, more studies are necessary to evaluate if interventions should be tailored to specific subgroups [49].

Efficacy — A number of randomized trials have evaluated the effects of probiotics in allergy prevention [50-80]. Most of these studies looked primarily at early outcomes of allergic disease, such as eczema and immunoglobulin E (IgE) mediated food allergy. Over one-half of them showed no significant reduction in the development of eczema, while there was benefit in the remaining studies, even when the same probiotic strain and a similar protocol were used. The majority of studies have shown no reduction in food allergy, allergic disease in general, or allergen sensitization. Few studies have assessed the long-term effects on respiratory-allergic disease (allergic rhinitis, asthma) in children greater than four to five years of age, and the results have been variable [49,58,60,71,73-75,81]. In addition to variability in results, there is also considerable heterogeneity in virtually all aspects of the conducted studies, including the strains used (different single strains or combinations of strains); the dose, the timing, and duration of administration; the method of probiotic delivery; and the outcome measures. The results are conflicting even for L. rhamnosus GG (LGG), the most extensively studied strain [50-52,58,69,70].

A meta-analysis from 2012 [44] included 13 prevention studies [50,52,54-57,59,61-63,65,66,82] and demonstrated that probiotic treatment reduced the incidence of eczema by 21 percent (risk ratio [RR] 0.79, 95% CI 0.71-0.88), from approximately 37 percent in the placebo group to 30 percent in the probiotic group. This effect was still present when the analysis was restricted to studies of patients with IgE-associated eczema (RR 0.80, 95% CI 0.66-0.96). However, this effect was seen only in the first two years of age and was lost by four years of age. Studies were reported to be fairly homogenous, and no clear difference emerged across various strata. A meta-analysis of trials through February 2014 that included 17 prevention studies reported nearly identical findings [46]. However, two subsequent randomized trials did not find a protective effect [76,79].

The definition of probiotics indicates that the microorganisms must be alive to exert their effects (see 'Definitions' above). However, there is preliminary evidence that dead microorganisms may also be beneficial. In a multicenter randomized trial, 129 infants at high risk of allergy were allocated to intake of a nonhydrolyzed formula with or without the addition of heat-killed Bifidobacterium breve C50 and Streptococcus thermophilus from birth until 12 months of age for prevention of early manifestations of allergic disease [83]. There was no decrease in the incidence of cow's milk allergy, but fewer infants were sensitized to cow's milk at 12 months in the active compared with the placebo group (1.7 versus 12.5 percent, respectively).

Target population — The allergic phenotype is often expressed within the first months of life [64]. Thus, prevention strategies are focused on pregnancy and/or the immediate postnatal period. Preventive measures may address the general population or target infants at high risk of developing allergic disease, characterized by a positive immediate family history of allergic disease. The latter group is more commonly targeted. It is not yet clear if specific subgroups may benefit from probiotic intervention.

The majority of studies have selectively recruited infants at high risk of allergic disease. Several studies have also compared outcomes according to maternal sensitization since maternal allergy confers a higher risk than paternal allergy. Reduced sensitization with L. reuteri and L. rhamnosus, respectively, was observed in two studies in the subgroup of high-risk infants with maternal sensitization compared with those with no maternal sensitization, although there was no benefit in the unsegregated primary analysis [53,56].

Only a few studies have included an unselected population (ie, participants both with and without a family history of allergic disease) [57,65,76]. Two showed a marked reduction in eczema risk, but the other showed no difference in risk. In one study, 415 pregnant mothers from an unselected population were assigned to a combination of B. lactis, L. rhamnosus, and L. acidophilus or placebo started at approximately 36 weeks of gestation and continued during breastfeeding for three months. The incidence of eczema was reduced at two years compared with placebo, and a subgroup analysis found that this preventive effect on eczema was most obvious in children without a family history of allergic disease [57].

Strain(s) and dose — Many different strains and combinations of probiotics have been used. Results are variable, even when the same strain is used. As an example, LGG reduced eczema in one study [50,51,58] but had no effect in two others [52,82], even though one of them [52] used a very similar protocol to that of the first study [50,51,58].

Several studies reported a significant reduction in eczema with a combination of probiotics (B. bifidum, B. lactis, and Lactococcus lactis [61]; B. bifidum, B. lactis, and L. acidophilus [59]; LGG, L. acidophilus, and B. lactis [57]; L. rhamnosus LPR and B. longum or L. paracasei and B. longum [72]) compared with placebo. However, combinations were not similarly effective in other studies (LGG and B. lactis [53,63]; L. rhamnosus LPR and B. longum [55]; L. salivarius, L. paracasei, B animalis subspecies lactis, and B. bifidum [76]). Treatment with a combination of L. rhamnosus, B. breve, and Propionibacterium freudenreichii was associated with decreased eczema at two, but not five, years of age compared with placebo [54,60].

Few studies have directly compared the effects of single strains or combinations of strains. One study showed differences in efficacy between two species [66]. In that study, a L. rhamnosus strain significantly reduced the prevalence of eczema at two, four, and six years of age while a B. lactis strain did not [66,75,81].

In studies designed for allergy prevention, doses have typically ranged between 108 to 1010 colony-forming units (CFU). However, dose-response data are scarce. A study designed to investigate the dose-response relationship on intestinal colonization reported successful temporary colonization with LGG at all three doses used (108, 109, or 1010 CFU/day) [84]. These findings cannot be extrapolated to other probiotic strains, because probiotic strains differ in their capacity to temporarily colonize the intestine. In addition, evidence of temporary colonization does not necessarily translate into a clinically relevant effect.

Timing and duration of treatment — The majority of studies have included prenatal treatment, typically for four to six weeks, but the period of postnatal supplementation has ranged from three months to two years. In some studies, the mother is treated antenatally rather than the infant [72]. Animal studies with probiotics show antenatal effects [85], but effects of maternal probiotics on cord blood immune responses are conflicting [86,87]. The only randomized trial to specifically examine the prenatal effects of probiotics gave LGG or placebo for the last two to four weeks of pregnancy with no postnatal supplementation found no effect [82]. Only one study [65] has specifically examined the effects of probiotic supplementation with a L. paracasei strain during weaning from breastfeeding (from 4 to 13 months) and found a reduction in eczema. Another study reported a significantly reduced incidence of eczema in infants who received L. rhamnosus and B. animalis subspecies lactis during six months starting in late infancy (mean age 10 months) compared with placebo [88]. These studies suggest that prenatal administration is not necessary for clinical benefit.

Two studies assessed the effects of LGG in the secondary prevention of allergic disease and found little to no effect on the outcomes measured [68,89]. In one study, 131 children 6 to 24 months old with recurrent wheeze and an atopic family history were randomized to LGG or placebo for six months. There was no difference in rates of atopic eczema or asthma-related events after six months of therapy and after six months of follow-up compared with placebo [68]. Another study evaluated the effects of probiotics for treatment of eczema and secondary prevention of sensitization and respiratory allergic disease [89]. A total of 208 infants aged three to six months with diagnosed eczema were randomized to L. paracasei, Bifidobacterium lactis, or placebo while receiving an extensively hydrolyzed formula. The study showed no clinical benefit through three years of age.

One trial of 220 children aged 1 to 12 months with double-blind, placebo-controlled food challenge (DBPCFC) confirmed cow's milk allergy randomly assigned to extensively hydrolyzed formula with or without LGG found a decreased risk of any other allergic manifestation including eczema, urticaria, asthma, and rhinoconjunctivitis in the LGG group over 36 months (absolute risk difference -0.23; 95% CI, -0.36 to -0.10) [90]. However, the trial was not blinded, and randomization was not concealed.

PREBIOTICS — There are limited data on the use of prebiotics to prevent allergic disease. A systematic review and meta-analysis found no effect on the development of asthma (two studies, 226 infants) but did find a significant reduction in eczema (four studies, 1218 infants; risk ratio [RR] 0.68, 95% CI 0.48-0.97; number needed to treat to benefit, 25 [95% CI 14 to >100]) [91]. Further studies, particularly in areas such as population to target (high- versus low-risk infants), best combination of prebiotics, and effect on development of allergic diseases other than eczema, are needed before routine administration of prebiotics to prevent allergic disease can be recommended.

SYNBIOTICS — One large study has specifically assessed a synbiotic combination in the primary prevention of allergic disease. In this randomized trial, 1223 pregnant mothers whose infants were at high risk for atopic disease were assigned to a synbiotic combination or placebo [54]. The synbiotic consisted of a prebiotic (galactooligosaccharides [GOS]) in combination with three different probiotic strains. This was administered to mothers for two to four weeks prior to delivery and then to their infants for the first six months postnatally. A reduction in eczema (odds ratio [OR] 0.74, 95% CI 0.55-0.98), particularly atopic eczema (OR 0.66, 95% CI 0.46-0.95), was seen in the synbiotic group at two years of age compared with placebo. There were no effects on other allergic outcomes or sensitization, although IgE-associated (atopic) diseases tended to occur less frequently in the synbiotic group. A follow-up at 5 and 13 years of age did not show any differences in the cumulative incidence of eczema, atopic eczema, sensitization, allergic rhinitis, or asthma between the treatment and placebo group [49,60]. However, in a subgroup analysis, there was some reduction in allergic outcomes in infants born by cesarean section [60], again illustrating that more studies are needed to evaluate if specific subgroups are more likely to benefit from interventions that modify infant gut microbiota. (See 'Target population' above and 'Proposed mechanisms' above.)

One study suggests that synbiotics may have a role in secondary prevention, with a reduced risk of developing wheeze seen in children with existing eczema [92]. In the original study, 90 infants less than seven months of age with atopic eczema were randomized to receive an extensively hydrolyzed whey formula with or without synbiotics (B. breve and a mixture of 90 percent GOS and 10 percent fructooligosaccharides [FOS]) for 12 weeks [93]. There was no effect of the synbiotic on the severity of eczema. However, 75 infants were reevaluated one year after the intervention, and a reduced prevalence of frequent wheezing was seen in the synbiotic group. Fewer infants had started to use asthma medication compared with placebo [92]. These results are preliminary, and larger, long-term studies are needed.

SIDE EFFECTS AND SAFETY — Generally, the safety profile of both prebiotics and probiotics is good, with few side effects observed in the previously reported clinical studies.

Prebiotics — Two meta-analyses that analyzed the safety of prebiotics in infancy concluded that formula with added prebiotics is well tolerated in full-term [94] and preterm infants [95].

Prebiotics are designed to selectively stimulate beneficial microbiota. However, preterm infants may not have an established microbiota that can use prebiotics for promoting health. There is at least a theoretical concern that the substrate may promote the growth of either potential pathogens or commensal luminal organisms that could translocate across the immature gut epithelial barrier and cause systemic disease. Animal models have demonstrated increased bacterial translocation in the intestine of immature pups fed a milk formula containing GOS and inulin [96].

No adverse effects were observed in one randomized prebiotic trial [27]. Another randomized prebiotic trial reported normal and similar growth in both the treatment and placebo groups [97]. Results from a primary prevention study using a combination of prebiotics and probiotics showed no differences in side effects or growth between children given the synbiotic versus placebo up to five years of age [60,98].

Probiotics — Administration of probiotics appears to be a safe intervention in the prenatal and postnatal periods in healthy individuals.

Probiotics are a diverse group of microorganisms. The most commonly used probiotics in allergy prevention are bifidobacteria and lactobacilli, normal commensals of the mammalian microbiota. They have been available in various types of foods for many years and are "generally recognized as safe" (GRAS) [14]. Theoretically, probiotics may cause systemic infections, deleterious metabolic activity, excessive immune stimulation, and gene transfer [99]. Lactobacillus septicemia was reported in children with short bowel syndrome receiving LGG supplementation [100]. In addition, caution is advised when using probiotics in immunocompromised patients, including premature infants, due to an increased risk of adverse effects and, in rare cases, infection caused by fungal contamination [101,102].

The safety of probiotics during pregnancy was evaluated in a meta-analysis [103]. Intake of lactobacilli and bifidobacteria during pregnancy had no effect on the incidence of cesarean section, birth weight, or gestational age. Several primary prevention studies have reported a paradoxical increase in possible allergy outcomes with probiotics, including sensitization [62], respiratory symptoms such as wheeze [52,62], and trends toward more rhinitis and asthma [58]. An actual causal relationship has not been confirmed, and follow-up studies that have evaluated lung function measures [73-75,78] and exhaled nitric oxide (eNO) [73-75,104] showed no harm with respect to probiotic treatment.

Products used in clinical trials are carefully screened for contaminating allergens. However, it is not uncommon for commercially available probiotics to be contaminated with cow's milk protein [105,106]. There are reports of sensitization to contaminating milk proteins [105] and of anaphylaxis to probiotics in children with cow's milk allergy due to contamination with cow's milk proteins [106]. Clinicians should promote awareness of these risks.

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or email these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Probiotics (The Basics)")

SUMMARY AND RECOMMENDATIONS

Definitions – Prebiotics are nondigestible carbohydrates that stimulate the growth and/or activity of beneficial colonic bacteria. Probiotics are live microorganisms that benefit the host. Synbiotics are a combination of prebiotics and probiotics. (See 'Definitions' above.)

Proposed mechanisms of action – There is evidence from animal models and in vitro studies that gut microbiota modulate immune programming and can prevent the allergic phenotype. However, there is still an incomplete understanding of the optimal patterns of colonization for promoting immune tolerance and the possible impact of prebiotics and probiotics in this process. (See 'Rationale for prebiotics and probiotics in allergic disease' above and 'Proposed mechanisms' above.)

Outcomes – Meta-analyses suggest a benefit of probiotics in reducing the development of eczema, but not any other allergic outcomes, in at-risk infants (defined as presence of a biologic parent or sibling with asthma, allergic rhinitis, eczema, or food allergy). However, the effect is moderate, and the only probiotic strain with reproducible data is Lactobacillus rhamnosus GG (LGG). In addition, the great heterogeneity of the studies makes it difficult to advise on specifics regarding therapy (eg, strains, dose, timing, and duration). Prebiotics and synbiotics, which are anticipated to have a more global effect on the gut microbiota, have shown some promise, but further studies are needed. (See 'Prebiotics' above and 'Probiotics' above and 'Synbiotics' above.)

Probiotics for prevention of eczema – We suggest not giving probiotics during pregnancy, lactation, and infancy for the prevention of eczema (Grade 2C). However, we do not discourage this approach if parents/caregivers express strong interest and there is a family history of atopic disease, given the low risk of adverse events and modest potential for benefit. Parents/caregivers who choose this approach should first discuss it with the appropriate clinician (eg, obstetrician, pediatrician, other primary care provider). (See 'Our approach' above.)

  1. West CE, Renz H, Jenmalm MC, et al. The gut microbiota and inflammatory noncommunicable diseases: associations and potentials for gut microbiota therapies. J Allergy Clin Immunol 2015; 135:3.
  2. Björkstén B, Sepp E, Julge K, et al. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol 2001; 108:516.
  3. Kalliomäki M, Kirjavainen P, Eerola E, et al. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol 2001; 107:129.
  4. Pan SJ, Kuo CH, Lam KP, et al. Probiotics and allergy in children--an update review. Pediatr Allergy Immunol 2010; 21:e659.
  5. Tang ML, Lahtinen SJ, Boyle RJ. Probiotics and prebiotics: clinical effects in allergic disease. Curr Opin Pediatr 2010; 22:626.
  6. Ozdemir O. Various effects of different probiotic strains in allergic disorders: an update from laboratory and clinical data. Clin Exp Immunol 2010; 160:295.
  7. Yao TC, Chang CJ, Hsu YH, Huang JL. Probiotics for allergic diseases: realities and myths. Pediatr Allergy Immunol 2010; 21:900.
  8. Sherman PM, Cabana M, Gibson GR, et al. Potential roles and clinical utility of prebiotics in newborns, infants, and children: proceedings from a global prebiotic summit meeting, New York City, June 27-28, 2008. J Pediatr 2009; 155:S61.
  9. Thomas DW, Greer FR, American Academy of Pediatrics Committee on Nutrition, American Academy of Pediatrics Section on Gastroenterology, Hepatology, and Nutrition. Probiotics and prebiotics in pediatrics. Pediatrics 2010; 126:1217.
  10. Fiocchi A, Burks W, Bahna SL, et al. Clinical Use of Probiotics in Pediatric Allergy (CUPPA): A World Allergy Organization Position Paper. World Allergy Organ J 2012; 5:148.
  11. West CE, Dzidic M, Prescott SL, Jenmalm MC. Bugging allergy; role of pre-, pro- and synbiotics in allergy prevention. Allergol Int 2017; 66:529.
  12. Gibson GR, Hutkins R, Sanders ME, et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol 2017; 14:491.
  13. Food and Agriculture Organization of the United Nations. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. http://www.who.int/foodsafety/publications/fs_management/en/probiotics.pdf (Accessed on July 22, 2010).
  14. Food and Agriculture Organization of the United Nations. Guidelines for the evaluation of probiotics in food ftp://ftp.fao.org/es/esn/food/wgreport2.pdf (Accessed on July 22, 2010).
  15. Gerrard JW, Geddes CA, Reggin PL, et al. Serum IgE levels in white and metis communities in Saskatchewan. Ann Allergy 1976; 37:91.
  16. Wold AE. The hygiene hypothesis revised: is the rising frequency of allergy due to changes in the intestinal flora? Allergy 1998; 53:20.
  17. Hill DA, Siracusa MC, Abt MC, et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med 2012; 18:538.
  18. Prescott SL, Björkstén B. Probiotics for the prevention or treatment of allergic diseases. J Allergy Clin Immunol 2007; 120:255.
  19. Sudo N, Sawamura S, Tanaka K, et al. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol 1997; 159:1739.
  20. Penders J, Stobberingh EE, van den Brandt PA, Thijs C. The role of the intestinal microbiota in the development of atopic disorders. Allergy 2007; 62:1223.
  21. Adlerberth I, Strachan DP, Matricardi PM, et al. Gut microbiota and development of atopic eczema in 3 European birth cohorts. J Allergy Clin Immunol 2007; 120:343.
  22. Penders J, Thijs C, van den Brandt PA, et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut 2007; 56:661.
  23. Wang M, Karlsson C, Olsson C, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol 2008; 121:129.
  24. Abrahamsson TR, Jakobsson HE, Andersson AF, et al. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol 2012; 129:434.
  25. Bisgaard H, Li N, Bonnelykke K, et al. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J Allergy Clin Immunol 2011; 128:646.
  26. Boehm G, Lidestri M, Casetta P, et al. Supplementation of a bovine milk formula with an oligosaccharide mixture increases counts of faecal bifidobacteria in preterm infants. Arch Dis Child Fetal Neonatal Ed 2002; 86:F178.
  27. Moro G, Arslanoglu S, Stahl B, et al. A mixture of prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the first six months of age. Arch Dis Child 2006; 91:814.
  28. Maslowski KM, Vieira AT, Ng A, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461:1282.
  29. Wong JM, de Souza R, Kendall CW, et al. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 2006; 40:235.
  30. Schouten B, van Esch BC, Hofman GA, et al. Cow milk allergy symptoms are reduced in mice fed dietary synbiotics during oral sensitization with whey. J Nutr 2009; 139:1398.
  31. Scholtens PA, Alliet P, Raes M, et al. Fecal secretory immunoglobulin A is increased in healthy infants who receive a formula with short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides. J Nutr 2008; 138:1141.
  32. van Hoffen E, Ruiter B, Faber J, et al. A specific mixture of short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides induces a beneficial immunoglobulin profile in infants at high risk for allergy. Allergy 2009; 64:484.
  33. Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nat Immunol 2004; 5:104.
  34. Rosenfeldt V, Benfeldt E, Valerius NH, et al. Effect of probiotics on gastrointestinal symptoms and small intestinal permeability in children with atopic dermatitis. J Pediatr 2004; 145:612.
  35. Kopp MV, Goldstein M, Dietschek A, et al. Lactobacillus GG has in vitro effects on enhanced interleukin-10 and interferon-gamma release of mononuclear cells but no in vivo effects in supplemented mothers and their neonates. Clin Exp Allergy 2008; 38:602.
  36. Muraro A, Halken S, Arshad SH, et al. EAACI food allergy and anaphylaxis guidelines. Primary prevention of food allergy. Allergy 2014; 69:590.
  37. Halken S, Muraro A, de Silva D, et al. EAACI guideline: Preventing the development of food allergy in infants and young children (2020 update). Pediatr Allergy Immunol 2021; 32:843.
  38. NIAID-Sponsored Expert Panel, Boyce JA, Assa'ad A, et al. Guidelines for the diagnosis and management of food allergy in the United States: report of the NIAID-sponsored expert panel. J Allergy Clin Immunol 2010; 126:S1.
  39. Cuello-Garcia CA, Fiocchi A, Pawankar R, et al. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Prebiotics. World Allergy Organ J 2016; 9:10.
  40. Fiocchi A, Pawankar R, Cuello-Garcia C, et al. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Probiotics. World Allergy Organ J 2015; 8:4.
  41. Osborn DA, Sinn JK. Probiotics in infants for prevention of allergic disease and food hypersensitivity. Cochrane Database Syst Rev 2007; :CD006475.
  42. Lee J, Seto D, Bielory L. Meta-analysis of clinical trials of probiotics for prevention and treatment of pediatric atopic dermatitis. J Allergy Clin Immunol 2008; 121:116.
  43. Doege K, Grajecki D, Zyriax BC, et al. Impact of maternal supplementation with probiotics during pregnancy on atopic eczema in childhood--a meta-analysis. Br J Nutr 2012; 107:1.
  44. Pelucchi C, Chatenoud L, Turati F, et al. Probiotics supplementation during pregnancy or infancy for the prevention of atopic dermatitis: a meta-analysis. Epidemiology 2012; 23:402.
  45. Azad MB, Coneys JG, Kozyrskyj AL, et al. Probiotic supplementation during pregnancy or infancy for the prevention of asthma and wheeze: systematic review and meta-analysis. BMJ 2013; 347:f6471.
  46. Zuccotti G, Meneghin F, Aceti A, et al. Probiotics for prevention of atopic diseases in infants: systematic review and meta-analysis. Allergy 2015; 70:1356.
  47. Panduru M, Panduru NM, Sălăvăstru CM, Tiplica GS. Probiotics and primary prevention of atopic dermatitis: a meta-analysis of randomized controlled studies. J Eur Acad Dermatol Venereol 2015; 29:232.
  48. Jiang W, Ni B, Liu Z, et al. The Role of Probiotics in the Prevention and Treatment of Atopic Dermatitis in Children: An Updated Systematic Review and Meta-Analysis of Randomized Controlled Trials. Paediatr Drugs 2020; 22:535.
  49. Kallio S, Kukkonen AK, Savilahti E, Kuitunen M. Perinatal probiotic intervention prevented allergic disease in a Caesarean-delivered subgroup at 13-year follow-up. Clin Exp Allergy 2019; 49:506.
  50. Kalliomäki M, Salminen S, Arvilommi H, et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet 2001; 357:1076.
  51. Kalliomäki 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.
  52. 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.
  53. Huurre A, Laitinen K, Rautava S, et al. Impact of maternal atopy and probiotic supplementation during pregnancy on infant sensitization: a double-blind placebo-controlled study. Clin Exp Allergy 2008; 38:1342.
  54. Kukkonen K, Savilahti E, Haahtela T, et al. Probiotics and prebiotic galacto-oligosaccharides in the prevention of allergic diseases: a randomized, double-blind, placebo-controlled trial. J Allergy Clin Immunol 2007; 119:192.
  55. Soh SE, Aw M, Gerez I, et al. Probiotic supplementation in the first 6 months of life in at risk Asian infants--effects on eczema and atopic sensitization at the age of 1 year. Clin Exp Allergy 2009; 39:571.
  56. Abrahamsson TR, Jakobsson T, Böttcher MF, et al. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 2007; 119:1174.
  57. Dotterud CK, Storrø O, Johnsen R, Oien T. Probiotics in pregnant women to prevent allergic disease: a randomized, double-blind trial. Br J Dermatol 2010; 163:616.
  58. Kalliomäki M, Salminen S, Poussa T, et al. Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 2003; 361:1869.
  59. Kim JY, Kwon JH, Ahn SH, et al. Effect of probiotic mix (Bifidobacterium bifidum, Bifidobacterium lactis, Lactobacillus acidophilus) in the primary prevention of eczema: a double-blind, randomized, placebo-controlled trial. Pediatr Allergy Immunol 2010; 21:e386.
  60. Kuitunen M, Kukkonen K, Juntunen-Backman K, et al. Probiotics prevent IgE-associated allergy until age 5 years in cesarean-delivered children but not in the total cohort. J Allergy Clin Immunol 2009; 123:335.
  61. Niers L, Martín R, Rijkers G, et al. The effects of selected probiotic strains on the development of eczema (the PandA study). Allergy 2009; 64:1349.
  62. Taylor AL, Dunstan JA, Prescott SL. Probiotic supplementation for the first 6 months of life fails to reduce the risk of atopic dermatitis and increases the risk of allergen sensitization in high-risk children: a randomized controlled trial. J Allergy Clin Immunol 2007; 119:184.
  63. Rautava S, Arvilommi H, Isolauri E. Specific probiotics in enhancing maturation of IgA responses in formula-fed infants. Pediatr Res 2006; 60:221.
  64. Prescott SL, Wiltschut J, Taylor A, et al. Early markers of allergic disease in a primary prevention study using probiotics: 2.5-year follow-up phase. Allergy 2008; 63:1481.
  65. West CE, Hammarström ML, Hernell O. Probiotics during weaning reduce the incidence of eczema. Pediatr Allergy Immunol 2009; 20:430.
  66. Wickens K, Black PN, Stanley TV, et al. A differential effect of 2 probiotics in the prevention of eczema and atopy: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 2008; 122:788.
  67. Lodinová-Zádníková R, Prokesová L, Kocourková I, et al. Prevention of allergy in infants of allergic mothers by probiotic Escherichia coli. Int Arch Allergy Immunol 2010; 153:201.
  68. Rose MA, Stieglitz F, Köksal A, et al. Efficacy of probiotic Lactobacillus GG on allergic sensitization and asthma in infants at risk. Clin Exp Allergy 2010; 40:1398.
  69. Boyle RJ, Ismail IH, Kivivuori S, et al. Lactobacillus GG treatment during pregnancy for the prevention of eczema: a randomized controlled trial. Allergy 2011; 66:509.
  70. Ou CY, Kuo HC, Wang L, et al. Prenatal and postnatal probiotics reduces maternal but not childhood allergic diseases: a randomized, double-blind, placebo-controlled trial. Clin Exp Allergy 2012; 42:1386.
  71. Jensen MP, Meldrum S, Taylor AL, et al. Early probiotic supplementation for allergy prevention: long-term outcomes. J Allergy Clin Immunol 2012; 130:1209.
  72. Rautava S, Kainonen E, Salminen S, Isolauri E. Maternal probiotic supplementation during pregnancy and breast-feeding reduces the risk of eczema in the infant. J Allergy Clin Immunol 2012; 130:1355.
  73. Abrahamsson TR, Jakobsson T, Björkstén B, et al. No effect of probiotics on respiratory allergies: a seven-year follow-up of a randomized controlled trial in infancy. Pediatr Allergy Immunol 2013; 24:556.
  74. West CE, Hammarström ML, Hernell O. Probiotics in primary prevention of allergic disease--follow-up at 8-9 years of age. Allergy 2013; 68:1015.
  75. Wickens K, Stanley TV, Mitchell EA, et al. Early supplementation with Lactobacillus rhamnosus HN001 reduces eczema prevalence to 6 years: does it also reduce atopic sensitization? Clin Exp Allergy 2013; 43:1048.
  76. Allen SJ, Jordan S, Storey M, et al. Probiotics in the prevention of eczema: a randomised controlled trial. Arch Dis Child 2014; 99:1014.
  77. Loo EX, Llanora GV, Lu Q, et al. Supplementation with probiotics in the first 6 months of life did not protect against eczema and allergy in at-risk Asian infants: a 5-year follow-up. Int Arch Allergy Immunol 2014; 163:25.
  78. Gorissen DM, Rutten NB, Oostermeijer CM, et al. Preventive effects of selected probiotic strains on the development of asthma and allergic rhinitis in childhood. The Panda study. Clin Exp Allergy 2014; 44:1431.
  79. Cabana MD, McKean M, Caughey AB, et al. Early Probiotic Supplementation for Eczema and Asthma Prevention: A Randomized Controlled Trial. Pediatrics 2017; 140.
  80. Peldan PS, Kukkonen AK, Savilahti E, Kuitunen M. Perinatal Probiotic Mixture and Development of Allergic Sensitization up to 13 Years of Age. Int Arch Allergy Immunol 2020; 181:270.
  81. Wickens K, Black P, Stanley TV, et al. A protective effect of Lactobacillus rhamnosus HN001 against eczema in the first 2 years of life persists to age 4 years. Clin Exp Allergy 2012; 42:1071.
  82. Boyle RJ, Ismail IH, Kivivuori S, et al. Lactobacillus GG treatment during pregnancy for the prevention of eczema: a randomized controlled trial. Allergy 2011; 66:509.
  83. Morisset M, Aubert-Jacquin C, Soulaines P, et al. A non-hydrolyzed, fermented milk formula reduces digestive and respiratory events in infants at high risk of allergy. Eur J Clin Nutr 2011; 65:175.
  84. Petschow BW, Figueroa R, Harris CL, et al. Effects of feeding an infant formula containing Lactobacillus GG on the colonization of the intestine: a dose-response study in healthy infants. J Clin Gastroenterol 2005; 39:786.
  85. Blümer N, Sel S, Virna S, et al. Perinatal maternal application of Lactobacillus rhamnosus GG suppresses allergic airway inflammation in mouse offspring. Clin Exp Allergy 2007; 37:348.
  86. Boyle RJ, Mah LJ, Chen A, et al. Effects of Lactobacillus GG treatment during pregnancy on the development of fetal antigen-specific immune responses. Clin Exp Allergy 2008; 38:1882.
  87. Prescott SL, Wickens K, Westcott L, et al. Supplementation with Lactobacillus rhamnosus or Bifidobacterium lactis probiotics in pregnancy increases cord blood interferon-gamma and breast milk transforming growth factor-beta and immunoglobin A detection. Clin Exp Allergy 2008; 38:1606.
  88. Schmidt RM, Pilmann Laursen R, Bruun S, et al. Probiotics in late infancy reduce the incidence of eczema: A randomized controlled trial. Pediatr Allergy Immunol 2019; 30:335.
  89. Gore C, Custovic A, Tannock GW, et al. Treatment and secondary prevention effects of the probiotics Lactobacillus paracasei or Bifidobacterium lactis on early infant eczema: randomized controlled trial with follow-up until age 3 years. Clin Exp Allergy 2012; 42:112.
  90. Berni Canani R, Di Costanzo M, Bedogni G, et al. Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestations in children with cow's milk allergy: 3-year randomized controlled trial. J Allergy Clin Immunol 2017; 139:1906.
  91. Osborn DA, Sinn JK. Prebiotics in infants for prevention of allergy. Cochrane Database Syst Rev 2013; :CD006474.
  92. van der Aa LB, van Aalderen WM, Heymans HS, et al. Synbiotics prevent asthma-like symptoms in infants with atopic dermatitis. Allergy 2011; 66:170.
  93. van der Aa LB, Heymans HS, van Aalderen WM, et al. Effect of a new synbiotic mixture on atopic dermatitis in infants: a randomized-controlled trial. Clin Exp Allergy 2010; 40:795.
  94. Rao S, Srinivasjois R, Patole S. Prebiotic supplementation in full-term neonates: a systematic review of randomized controlled trials. Arch Pediatr Adolesc Med 2009; 163:755.
  95. Srinivasjois R, Rao S, Patole S. Prebiotic supplementation of formula in preterm neonates: a systematic review and meta-analysis of randomised controlled trials. Clin Nutr 2009; 28:237.
  96. Barrat E, Michel C, Poupeau G, et al. Supplementation with galactooligosaccharides and inulin increases bacterial translocation in artificially reared newborn rats. Pediatr Res 2008; 64:34.
  97. Arslanoglu S, Moro GE, Schmitt J, et al. Early dietary intervention with a mixture of prebiotic oligosaccharides reduces the incidence of allergic manifestations and infections during the first two years of life. J Nutr 2008; 138:1091.
  98. Kukkonen K, Savilahti E, Haahtela T, et al. Long-term safety and impact on infection rates of postnatal probiotic and prebiotic (synbiotic) treatment: randomized, double-blind, placebo-controlled trial. Pediatrics 2008; 122:8.
  99. Borriello SP, Hammes WP, Holzapfel W, et al. Safety of probiotics that contain lactobacilli or bifidobacteria. Clin Infect Dis 2003; 36:775.
  100. Kunz AN, Noel JM, Fairchok MP. Two cases of Lactobacillus bacteremia during probiotic treatment of short gut syndrome. J Pediatr Gastroenterol Nutr 2004; 38:457.
  101. Liong MT. Safety of probiotics: translocation and infection. Nutr Rev 2008; 66:192.
  102. Centers for Disease Control and Prevention. Health Alert Network. Fatal gastrointestinal mucormycosis in an infant following ingestion of contaminated dietary supplement – Connecticut, 2014. http://www.bt.cdc.gov/han/han00373.asp (Accessed on November 25, 2014).
  103. Dugoua JJ, Machado M, Zhu X, et al. Probiotic safety in pregnancy: a systematic review and meta-analysis of randomized controlled trials of Lactobacillus, Bifidobacterium, and Saccharomyces spp. J Obstet Gynaecol Can 2009; 31:542.
  104. Kukkonen AK, Kuitunen M, Savilahti E, et al. Airway inflammation in probiotic-treated children at 5 years. Pediatr Allergy Immunol 2011; 22:249.
  105. Bruni FM, Piacentini GL, Peroni DG, et al. Cow's milk allergic children can present sensitisation to probiotics. Acta Paediatr 2009; 98:321.
  106. Lee TT, Morisset M, Astier C, et al. Contamination of probiotic preparations with milk allergens can cause anaphylaxis in children with cow's milk allergy. J Allergy Clin Immunol 2007; 119:746.
Topic 13581 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟