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

Probiotics for gastrointestinal diseases

Probiotics for gastrointestinal diseases
Literature review current through: Jan 2024.
This topic last updated: Jan 24, 2022.

INTRODUCTION — The intestinal tract is host to a vast ecology of microbes that are necessary for health, but also have the potential to contribute to the development of diseases in susceptible individuals by a variety of mechanisms. Genetic or environmentally triggered perturbations in intestinal epithelial barrier function or innate immune bacterial killing, for example, can lead to an inflammatory response caused by increased uptake of bacterial and food antigens that stimulate the mucosal immune system [1-4]. (See "Immune and microbial mechanisms in the pathogenesis of inflammatory bowel disease".)

Interactions between intestinal microbes and the host are the subject of intensive ongoing research since they may influence a variety of diseases. Part of this research involves the deliberate manipulation of the intestinal microbiota with a therapeutic intention. The greatest experience has been in the inflammatory bowel diseases, ulcerative colitis, Crohn disease, pouchitis, and irritable bowel syndrome, although clinical trials are emerging in several other conditions.

There are four general methods by which the intestinal microbiota can be altered: administration of antibiotics, therapeutic diets and prebiotics (ie, dietary components that promote the growth and metabolic activity of beneficial bacteria), probiotics (ie, beneficial bacteria), or fecal microbial transplant (FMT). Combination of these methods is also possible (synbiotics). Interest in these approaches has extended well beyond the clinical sciences since a role for intestinal microbes in health and disease has been recognized in alternative and complementary forms of medicine for many years [5]. In a case control study, 51 percent of ulcerative colitis patients and 43 percent of patients with Crohn disease used probiotics compared with 21 percent of healthy controls [6]. In comparison, systematic evaluation of the efficacy of probiotics is relatively recent. This topic review focuses on clinical trials of probiotics in gastrointestinal diseases.

DEFINITION — Probiotics are microorganisms that have beneficial properties for the host. Most commercial products have been derived from food sources, especially cultured milk products. The list of such microorganisms continues to grow and includes many strains of lactic acid bacilli (eg, Lactobacillus and Bifidobacterium), a nonpathogenic strain of Escherichia coli (eg, E. coli Nissle 1917), Clostridium butyricum, Streptococcus salivarius, and Saccharomyces boulardii (a nonpathogenic strain of yeast). Also under development are strains of bacteria that have been genetically engineered to secrete immunomodulators (such as interleukin-10, trefoil factors, or defensins, or express altered surface proteins such as lipoteichoic acid), which have the potential to favorably influence the immune system [7-9]. More recently, the concept of restoring levels of protective commensal bacterial species that are diminished in certain disorders, such as Crohn disease, has been advanced [4,10-14].

Studies of selected probiotic species (given alone or in combination) have suggested potential efficacy in several gastrointestinal illnesses, the best studied of which are the inflammatory bowel diseases (particularly pouchitis) [4,15-22]. Therapeutic benefit has also been suggested in several other disorders, including antibiotic-related diarrhea, Clostridioides difficile toxin-induced colitis, infectious diarrhea, hepatic encephalopathy, irritable bowel syndrome, and allergy [23-27].

Some of the more commonly available probiotics include:

VSL#3 (Bifidobacterium breve, B. longum, B. infantis, Lactobacillus acidophilus, L. plantarum, L. paracasei, L. bulgaricus, Streptococcus thermophilus) [Note: Studies before 2015 were all performed with the original Italian (DeSimone) preparation]). The United States VSL#3 formulation is somewhat different and has different functional properties [28].

Align (B. infantis)

Culturelle (L. rhamnosus GG)

DanActive (L. casei)

Mutaflor (E. coli Nissle 1917)

Florastor (S. boulardii)

MECHANISMS OF BENEFIT — Mechanisms for the benefits of probiotics are incompletely understood. However, four general benefits have been described [4,29-31]:

Suppression of growth or epithelial binding/invasion by pathogenic bacteria [32].

Improvement of intestinal barrier function [33-35].

Modulation of the immune system. Several probiotic species or their products induce protective cytokines, including IL-10 and TGF-beta, and suppress proinflammatory cytokines, such as TNF, in the mucosa of patients with pouchitis, ulcerative colitis, and Crohn disease, in murine experimental colitis, and in isolated splenocytes [10,11,30,32,34,36-41]. S. boulardii limited the migration of T-helper 1 cells in inflamed colon tissue in a mouse colitis model and Lactobacillus casei protects from experimental colitis by inducing TLR-2-mediated regulatory T cells [42,43].

Modulation of pain perception. Some Lactobacillus strains appear to induce expression of micro-opioid and cannabinoid receptors in intestinal epithelial cells and mediate analgesic functions in the gut in a manner similar to the effects of morphine [44].

Probiotics differ in their ability to resist gastric acid and bile acids, colonize the intestinal tract, and influence cytokines secreted by intestinal epithelial cells [34,36,41,45-50]. Thus, not all probiotics are alike; as a result, benefits observed clinically with one species or with a combination of species are not necessarily generalizable to another [51].

Although yogurt is commonly recommended as a source of probiotics, not all of the live cultures contained in yogurt survive well in an acidic environment nor do they colonize the microbiota efficiently [52,53]. Furthermore, the residual lactose contained in yogurt can increase symptoms in patients with lactose intolerance, including those who develop secondary lactose intolerance due to an antecedent gastroenteritis (a setting in which probiotics have been recommended). Fermented dairy beverages, such as kefir, that contain much higher concentrations of live cultures than yogurt or cultures that are relatively resistant to gastric acid are also available [54]. Of interest, a fermented food diet increased diet microbiota diversity and decreased inflammatory markers to a greater extent than did dietary fiber [55].

Interesting observations indicate that it may not be necessary to administer living organisms to achieve a benefit. Secreted proteins and DNA of one probiotic preparation (VSL#3, Nature's Pharmaceuticals, Inc.) blocked cytokine activation and prevented apoptosis of epithelial cells [56,57]. The effects depended upon the specific DNA from the different bacterial species that were components of the preparation [56]. Non-methylated DNA from VSL#3, as well as other randomly selected E. coli strains, suppressed experimental colitis in several animal models [58]. These therapeutic effects are mediated through toll-like receptor 9 and with induction of type 1 interferons alpha/beta [59]. Defined molecular weight proteins from other probiotic species, including Lactobacillus GG, can also inhibit proinflammatory signaling and inflammatory cytokine-induced apoptosis in colonic epithelial cells through an epidermal growth factor receptor (EGFR)-dependent mechanism [34,35], while secreted products from a variety of species can inhibit cytokine production [10]. Similarly, heat-killed Lactobacillus casei Lbs2 induces regulatory T cell via TLR2 signaling [43], while native microbial products, such as flagellin from Roseburia intestinalis, can target the NLRP3 inflammasome and pyroptosis [60].

Some studies demonstrate that products of endogenous resident microbiota can have beneficial effects, creating opportunities for therapeutic manipulation beyond the traditional Lactobacillus and Bifidobacterium species. For example, Faecalibacterium prausnitzii, which is decreased in Crohn disease, secretes yet to be defined products that suppress experimental colitis and proinflammatory cytokines [10], and polysaccharide A of Bacteroides fragilis stimulates regulatory T cells in a TLR2, IL-10-dependent fashion [61]. Likewise, outer membrane vesicles from E. coli Nissle attenuated experimental colitis [62]. Clostridium species comprising Groups IV and XIVA, which are selectively decreased in active IBD patients, can induce IL-10 producing regulatory T cells [11,63]. This approach will likely be available for therapeutic use in the near future [13,14].

The use of probiotics to manage gastrointestinal disorders has received intense interest for the past several decades, but documentation of their efficacy and safety has suffered from the lack of large rigorous trials. This, in part, is because probiotics are classified as food additives and, therefore, are not required to undergo US Food and Drug Administration approval for use. Efficacy of existing data is summarized by two rigorously structured technical reviews by the American Gastroenterological Association (AGA) and the Cochrane Collaboration [25,51].

GASTROINTESTINAL INFLAMMATION

Pouchitis — For the surgical treatment of ulcerative colitis and familial adenomatous polyposis, proctocolectomy with ileal pouch-anal anastomosis (IPAA) is the favored alternative to proctocolectomy with permanent ileostomy since it preserves intestinal continuity and sphincter function and removes almost all of the colorectal mucosa. This procedure consists of total abdominal colectomy and the construction of a J or S-shaped ileal pouch that is anastomosed to the anus.

The most frequently observed long-term complication of IPAA is acute and/or chronic inflammation of the ileal reservoir, called pouchitis. Symptoms of pouchitis include increased stool frequency, urgency, hematochezia, abdominal pain, fever, and extraintestinal manifestations of inflammatory bowel diseases (see "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis"). Pouchitis affects approximately 20 percent of ulcerative colitis patients in the first year after creation of the IPAA and 50 percent of patients after five years, but less than 1 percent of patients with colectomies to treat familial polyposis.

Detailed studies of the microbiota in patients with pouchitis have demonstrated unique patterns, including the persistence of Fusobacteria and enteric species, increased Clostridium perfringens, and the absence of Streptococcus and Faecalibacterium species in the inflamed pouch [64-67]. These observations and a high rate of response to various antibiotics support an important role for bacteria in the pathogenesis of pouchitis and provide a rationale for clinical trials aimed at altering the microbiota with probiotics.

Small controlled trials have suggested that at least one probiotic preparation (VSL#3; original Italian formulation) containing 5 X 10(11) per gram of four Lactobacillus species, three Bifidobacterium species, and one strain of Streptococcus salivarius subspecies thermophilus may be effective in prevention of recurrent pouchitis after antibiotic induction of remission.

A randomized, placebo-controlled trial included 40 patients with a history of chronic, relapsing pouchitis who were placed into clinical and endoscopic remission with broad-spectrum antibiotics [68]. Patients were randomly assigned to VSL#3 6 g/day or placebo. After nine months of daily treatment, significantly fewer patients in the probiotic group had experienced a relapse (15 versus 100 percent). Within three months of stopping treatment, all patients in the VSL#3 group had relapsed. Likewise, fecal Lactobacillus and bifidobacteria concentrations returned to pretreatment levels within one month after therapy was discontinued, indicating that permanent colonization with the probiotic species did not occur.

A second study from the same group and investigators in London included 36 patients with recurrent or refractory pouchitis who had required antibiotics at least twice in the previous year [69]. After achieving remission with antibiotics, patients were randomly assigned to VSL#3 or placebo once daily for one year. Significantly more patients in the probiotic group remained in remission (85 versus 6 percent). Patients randomized to VSL#3 also had significantly better quality of life.

A third study from the Gionchetti group included 40 consecutive patients who underwent IPAA for ulcerative colitis [70]. Patients were randomly assigned to receive VSL#3 3 g/day or placebo immediately after ileostomy closure for one year. Patients receiving the probiotic had significantly fewer episodes of pouchitis (10 versus 40 percent). Probiotic treatment was also associated with significant improvement in quality of life compared with placebo.

Different conclusions were reached in an observational study in the United States that involved 31 patients with antibiotic-dependent pouchitis who were treated with VSL#3 after achieving remission with ciprofloxacin [71]. After eight months, only a minority of patients remained on probiotic therapy and in symptomatic remission (most having stopped it due to recurrence of symptoms or adverse effects).

Other studies have demonstrated a benefit from other probiotics including Lactobacillus rhamnosus GG, Lactobacillus plantarum 299, and B. infantis [44,72,73]. Several systematic reviews have suggested that VSL#3 may be more effective than placebo for preventing relapse of chronic pouchitis and preventing onset of pouchitis [21,74]. However, both the AGA Systematic Review and two Cochrane database systematic reviews judged that the quality of the supporting evidence was very low [21,51,75].

Ulcerative colitis — Convincing data to support the use of probiotic preparations in ulcerative colitis are lacking [51,76-79]. Although various probiotic species have shown promise in the treatment of ulcerative colitis, because of the small number of patients in these studies and the risks associated with probiotics, systematic reviews have reached variable conclusions regarding the use of probiotics for the induction and maintenance of remission of ulcerative colitis [22,25,80,81], but publication bias may influence results.

The following observations were made in randomized trials:

E. coli 1917 Nissle was as effective as low dose 5-ASA in preventing relapse of ulcerative colitis in adults [82,83] and in an open label trial in children [84]. Rectal administration of E. coli 1917 Nissle enemas had equivocal results [85].

Lactobacillus GG appeared to be more effective than standard treatment involving mesalazine in prolonging relapse-free time, but did not influence relapse rates in patients with quiescent ulcerative colitis [86].

The combination of a prebiotic and a probiotic (B. longum) was associated with improvement in histologic scores and measures of immune activation in a one-month randomized controlled trial [87].

The combination of the original VSL#3 preparation plus balsalazide was slightly more effective than balsalazide or mesalazine alone in a randomized trial of patients with acute mild-to-moderate ulcerative colitis [88]. A randomized trial involving 29 children with ulcerative colitis found that VSL#3 was more effective than placebo in maintaining remission (73 versus 21 percent at one year) when given in conjunction with steroid induction and mesalamine maintenance treatment [89].

Emerging evidence suggests that VSL#3 can induce remission and reduce disease activity in patients with mild/moderate active ulcerative colitis [90-93]. A randomized trial of 77 patients found that VSL#3 was more effective than placebo in improving the ulcerative colitis disease activity index (UCDAI) by 50 percent at six weeks (33 versus 10 percent) and inducing remission at 12 weeks (43 versus 16 percent) [90]. A second randomized trial examined 144 patients who were receiving a 5-ASA, azathioprine, and/or methotrexate [93]. The trial similarly found that patients who received VSL#3 were more likely to have at least a 50 percent decrease in the UCDAI after eight weeks compared with patients who received placebo (63 versus 41 percent). Remission rates were also higher in the VSL#3 group (48 versus 32 percent). However, histologic scores were not significantly improved with VSL#3 therapy.

A small, double-blind, placebo-controlled trial showed no significant difference in the rates of maintenance of remission in patients with left-sided ulcerative colitis randomized to 52 weeks of L. acidophilus La-5 and Bifidobacterium animalis subsp. lactis Bb-12 or placebo (relapse rate 75 versus 92 percent) [94].

Another small, double-blind, placebo-controlled study reported significantly lower clinical and endoscopic disease activity in pediatric patients with ulcerative colitis treated with Lactobacillus reuteri ATCC 55730 [95].

A two-year comparison of low-dose mesalamine alone versus mesalamine plus L. salivarius, L. acidophilus, and B. bifidus BGN4 showed improved outcomes with adjunctive probiotics [96].

Crohn disease — Clinical trials of probiotics in Crohn disease have shown mixed results. The reasons for the heterogeneity are unclear, but could be due to several factors such as the specific probiotics (and doses) used, differences in study duration, characteristics of the included patients (eg, location of disease), and endpoints that were measured. Likewise, use of probiotics in prevention of postoperative recurrence of Crohn disease has been unsuccessful [97].

In the aggregate, the available data do not support clinical effectiveness of probiotic therapy for either induction or maintenance of remission in patients with Crohn disease [25,26,51,78,80,98-100]. Whether certain patient subgroups might benefit remains to be determined. A report of clinical improvement with combination of a probiotic, B. longum, and a prebiotic [101] suggested the possibility of using a synbiotic approach to treating Crohn disease, although previous reports with other agents were less positive. While probiotics administration is generally regarded as safe, toxicity can occur in the setting of immunosuppression and enhanced mucosal permeability, as evidenced by Lactobacillus-induced sepsis in a Crohn disease and HIV-infected patient taking a self-made yogurt [102].

Diverticular colitis — Infrequently, patients with diverticular disease develop a segmental colitis, most commonly in the sigmoid colon, which can occasionally be symptomatic. Combination therapy with VSL#3 and oral beclomethasone dipropionate (a locally acting corticosteroid not available in the United States) was beneficial in a case series. (See "Segmental colitis associated with diverticulosis".)

Radiation enteritis — A meta-analysis of six randomized controlled trials of probiotics in radiation-induced diarrhea detected slight overall benefit in diarrhea, but no difference in Bristol Stool Scales or antidiarrheal mediation use [103]. Results of individual studies were variable. Basic research indicates that intestinal bacteria contribute to radiation-induced injury and repair, so this therapeutic application is open to investigation [104]. Lachnospiraceae and their short-chain fatty acids and tryptophan metabolites were associated with survival to total body irradiation in a mouse model [105].

DIARRHEAL ILLNESSES

C. difficile infection — Issues related to probiotics and C. difficile infection are discussed separately. (See "Clostridioides difficile infection in adults: Treatment and prevention", section on 'Alternative therapies'.)

Infectious diarrhea — The use of probiotics in children with presumed acute infectious diarrheal illness has no benefit. Probiotic use in adults with acute infectious gastroenteritis is unproven. Older systematic reviews have demonstrated a modest reduction in the duration of infectious diarrhea with the use of probiotics, although there was heterogeneity among studies [25,106-111]. However, two large prospective, randomized multicenter trials demonstrated no benefit of various commonly used probiotic strains on clinical progression or duration of symptoms in infants and young children seen in emergency rooms for early acute gastroenteritis [112-114]. These studies led a European Society for Pediatric Gastroenterology, Hepatology, and Nutrition working group to conclude that evidence supporting benefits of various probiotics in managing acute gastroenteritis in children was weak, low to very low certainty, while evidence of the lack of benefit for combinations of Lactobacillus rhamnosus R0011 and Lactobacillus helveticus R0052 was strong [115]. Likewise, the AGA Technical Review on probiotics in gastrointestinal diseases concluded that probiotics were not beneficial for treating children with acute gastroenteritis (moderate evidence) [51].

Data to support the specific type, dose, or duration of probiotic and the mechanism of protection from specific etiologic agents are limited. It is also unclear whether probiotics reduce important complications of diarrheal illness such as dehydration and malnutrition.

A 2010 meta-analysis that included 63 randomized controlled trials (using several different probiotic preparations) in adults and children found that probiotics reduced the overall risk of diarrhea lasting four or more days by 59 percent (relative risk 0.41, 95% CI 0.32-0.53) and the mean duration of diarrhea by 25 hours (95% CI 16-34 hours) [110]. The two most commonly studied probiotics were Lactobacillus GG and S. boulardii.

In a randomized double-blinded trial of 971 children three months to four years of age presenting to an emergency room with acute gastroenteritis symptoms, a five-day course of L. rhamnosus GG did not significantly alter symptom severity or duration relative to placebo [112]. In a similarly designed study of 886 children ages 3 to 48 months, L. rhamnosus R0011 and L. helveticus R0052 given for five days did not alter gastroenteritis clinical progression, duration of symptoms, or health care utilization compared with placebo [113]. An analysis of fecal specimens from the L. rhamnosus R0011 and L. helveticus R0052 study showed no viral-specific benefits or accelerated viral clearance by the probiotics [114].

The role of probiotics in the prevention of traveler's diarrhea is discussed in detail separately [116]. (See "Travelers' diarrhea: Treatment and prevention".)

Collagenous colitis — Collagenous colitis, a subtype of microscopic colitis, is a diarrheal illness characterized by the presence of a thickened subepithelial collagenous plate and lymphocytic infiltrate in the colonic mucosa. (See "Microscopic (lymphocytic and collagenous) colitis: Clinical manifestations, diagnosis, and management".)

A possible benefit of E. coli strain Nissle 1917 was suggested in an open-label study of 14 patients [117]. The authors hypothesized that the benefit may have been due to an antagonistic effect of the probiotic against strains of Yersinia species. In a second placebo-controlled trial, a combination of L. acidophilus and B. animalis strains had no significant effect on primary endpoints but were associated with some improvement in symptoms [118].

Celiac disease — Probiotics can degrade or alter gluten and gliadin [119-123], and Lactobacillus and Bifidobacterium concentrations are decreased in patients with celiac disease [124]. However, there is no evidence to support the use of probiotics in patients with celiac disease [125]. In a randomized controlled trial, 22 adults with celiac disease on a gluten-containing diet were randomized to treatment with B. infantis NLS super strain or placebo [126]. Although patients treated with B. infantis reported a significant improvement in constipation, indigestion, and gastroesophageal reflux as compared with placebo, there was no significant difference in diarrhea, abdominal pain, or celiac serologies. In contrast, in a randomized trial in which 33 children were assigned to treatment with B. longum CECT7347 or placebo added to a gluten-free diet, treatment with B. longum CECT7347 resulted in a reduction of fecal secretory IgA [127]. Suggestions that probiotics might be clinically effective are provided by a lack of activation of mucosal cytokine responses following consumption of bread degraded ex vivo by 10 probiotic Lactobacillus strains [122] and probiotic-digested gliadin-improved function of cultured epithelial cell lines [123]. (See "Epidemiology, pathogenesis, and clinical manifestations of celiac disease in adults", section on 'Clinical manifestations'.)

CONSTIPATION — Evidence to support the use of single or combination probiotics in children or adults with functional constipation are lacking [128,129]. Multiple small, randomized, placebo-controlled trials of probiotics in patients with chronic constipation without irritable bowel syndrome, and in normal subjects with a tendency toward infrequent stools, suggest improvement in defecation frequency, stool consistency, and intestinal transit time with Bifidobacterium lactis DN-173 010, B. lactis BB12, Lactobacillus casei Shirota, L. reuteri DSM 19738 and E. coli Nissle 1917 [130-135]. However, these results using probiotics in the management of severe constipation should be interpreted with caution due to marked heterogeneity in study design and results, as well as publication bias [136,137]. Limited heterogeneous studies do not support use of probiotics in children with functional constipation [138,139].

IRRITABLE BOWEL SYNDROME — Evidence remains unconvincing for benefits of probiotics for treating irritable bowel syndrome (IBS), probably due to marked heterogeneity in this disorder and variability between the agents studied [51]. A number of controlled trials of probiotics in IBS have been published, many of which can be criticized for methodologic limitations [51,140-154]. All were short-term studies and none has provided clear evidence as to the potential role of probiotic treatment. Furthermore, the magnitude of benefit in studies with positive results was modest, suggesting that the probiotics under consideration are unproven, but may have the potential to impact the management of IBS [25,99,143,155-158]. It is possible that a clinically important benefit might be achieved in certain subgroups of patients, particularly those with diarrhea-predominant symptoms in light of data of alterations in the composition of resident enteric bacterial species in this subset of IBS [159] and its response to Rifaximin [158]. As in inflammatory bowel disease, a preferred approach may be to correct the altered microbiome in an individual based on observed compositional changes, rather than use a generic exogenous probiotic preparation for all patients.

Meta-analyses of randomized controlled trials found important methodologic limitations of most of the studies [143,158]. There was some evidence of efficacy for B. infantis 35624 in two appropriately designed studies. The following illustrate the range of findings in some of the largest controlled trials:

The probiotic B. infantis was significantly more effective than placebo at four weeks in a controlled trial of 362 patients with IBS [142]. However, the benefit was confined to only one of three doses tested, and there was no clear dose-response relationship.

In one trial, 77 patients with IBS were randomly assigned to a malted milk drink containing Lactobacillus salivarius UCC4331 or B. infantis 35624 or to a malted milk drink alone [144]. Symptoms were significantly improved at most time points in the group receiving B. infantis. There was a corresponding normalization in the ratio of serum IL-10/IL-12 suggesting that the probiotic may help reduce a proinflammatory state associated with IBS.

Improvement in abdominal pain and a trend towards normalization of stool frequency in constipated patients was found in the probiotic-treated group in a placebo-controlled trial of 40 patients randomly assigned to L. plantarum 299V or placebo [160]. By contrast, no benefit was observed in a second placebo-controlled trial [161]. No overall improvement was observed in a randomized, placebo-controlled trial involving 100 patients with IBS treated with a combination of four probiotic species [162].

In a trial with 122 patients with IBS, Bifidobacterium bifidum MIMBb75 was compared with placebo. Patients who received B. bifidum MIMBb75 had a 52 percent overall response rate compared with 21 percent for patients who received placebo [163].

A placebo-controlled blinded trial of a low fermentable oligo- di- and monosaccharides and polyols (FODMAP) diet with or without multi-strain probiotics in 104 IBS patients showed reduced symptom scores with the low FODMAP diet, but no added benefit of the probiotics [150].

A multicenter controlled trial of B. infantis 35624 in 302 non-patients with abdominal discomfort and bloating showed no benefit in symptoms beyond a significant placebo effect [151].

A multicenter German controlled trial reported that nonviable Bifidobacterium bifidum HI-MIMBb75 daily for eight weeks in 443 IBS patients led to at least 30 percent improvement in abdominal pain and decreased activity of other IBS symptoms in at least four of the eight weeks of treatment in 34 percent of subjects, compared with improvement in 19 percent of placebo-treated subjects (risk ratio 1.7, 95% CI 1.3-2.4) [164].

LACTOSE INTOLERANCE — Ingestion of lactase-containing probiotics has the potential to aid lactose digestion in patients with lactose intolerance. Several studies have evaluated the benefit of various probiotics in patients with lactose intolerance [145,165]. A systematic review of 15 controlled trials found inconsistent results among strains, but a positive effect of probiotics and suggested further studies on specific strains in which a benefit was suggested [165]. A study of a lactose-fermenting Lactobacillus acidophilus strain showed reduced symptoms after an in vivo lactose challenge, laying the foundation for further long-term trials [166].

HEPATIC ENCEPHALOPATHY — The role of probiotics in the treatment of hepatic encephalopathy is discussed separately. (See "Hepatic encephalopathy in adults: Treatment", section on 'Modification of colonic flora (prebiotics and probiotics)'.)

PANCREATITIS — A multicenter, double-blind, placebo-controlled randomized trial of multispecies probiotic preparation and placebo demonstrated that probiotics did not reduce the risk of infectious complications and actually increased mortality from mesenteric ischemia in patients with pancreatitis [167,168]. As a result, probiotics are not recommended in severe acute pancreatitis.

SMALL BOWEL BACTERIAL OVERGROWTH — The role of probiotics in small bowel bacterial overgrowth is unproven. (See "Small intestinal bacterial overgrowth: Management".)

ALLERGY — Probiotics have the potential to reduce intestinal permeability and the generation of proinflammatory cytokines that are elevated in patients with a variety of allergic disorders. The role of probiotics in the treatment of allergic disease is discussed separately. (See "Prebiotics and probiotics for prevention of allergic disease".)

H. PYLORI — The role of probiotics in treatment of H. pylori infection is discussed separately. (See "Treatment regimens for Helicobacter pylori in adults".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Probiotics".)

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 e-mail 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

Several probiotic preparations have promise in preventing or treating various conditions. However, most studies have been small and use highly variable probiotic preparations, and many have important methodologic limitations, making it difficult to make unequivocal conclusions regarding efficacy, especially when compared with proven therapies. Furthermore, considerable differences exist in composition, doses, and biologic activity between various commercial preparations, so that results with one preparation cannot be applied to other probiotic preparations. Finally, costs to the patient may be considerable since no preparation is FDA approved and most are not reimbursed by insurers. Enthusiasm for use of probiotics has outpaced the scientific evidence. Large, well-designed multicenter controlled clinical trials with replication cohorts are needed to clarify the role of specific probiotics in different well-defined patient populations.

The decision to use a probiotic rests mostly upon the degree of anticipated benefit, available alternatives, the clarity of the available data in showing a benefit, costs, and patient preferences. No probiotic strategy is currently considered to represent either the standard of care or primary treatment for any of the conditions described above. The following recommendations are based upon the author's overall appraisal of the quality and consistency of the available evidence.

Pouchitis – Data from small, controlled trials suggest a benefit from the original (DeSimone) VSL#3 consortium in the primary and secondary prevention of pouchitis. Thus, this is a reasonable adjunctive option in addition to standard medical therapy, although long-term efficacy is uncertain and access to this preparation in the United States is difficult. (See 'Pouchitis' above.)

Ulcerative colitis – Benefits of probiotics in ulcerative colitis remain unproven, but E. coli Nissle 1917 shows promise in maintaining remission and could be considered as an alternative to biologic approaches in patients with mild recurrent disease who are intolerant or resistant to 5-ASA preparations. The original VSL#3 preparation may have some efficacy in treating active disease as an adjunctive approach, but the quality of data is poor. No probiotic preparations have been validated for clinical use in ulcerative colitis. (See 'Ulcerative colitis' above.)

Crohn disease – A benefit of standard probiotics in Crohn disease remains unproven. (See 'Crohn disease' above.)

Infectious diarrhea – The use of probiotics in children with presumed acute infectious diarrheal illness has no benefit. Probiotic use in adults with acute infectious gastroenteritis is unproven. (See 'Infectious diarrhea' above.)

Constipation – Limited randomized controlled trials suggest possible improvement in defecation frequency and stool consistency in adult and older adult patients with chronic constipation, but limited benefit in children. However, these studies are likely subject to publication bias and larger studies are needed before probiotics can be routinely recommended in the management of severe chronic constipation. (See 'Constipation' above.)

Irritable bowel syndrome – Evidence remains unconvincing for benefits of probiotics for treating irritable bowel syndrome, probably due to marked heterogeneity in this disorder and variability between the agents studied. A definitive therapeutic role remains unproven and needs to be further investigated in defined patient subsets. (See 'Irritable bowel syndrome' above.)

Lactose intolerance – A benefit of probiotics for lactose intolerance remains unproven. (See 'Lactose intolerance' above.)

Hepatic encephalopathy – Initial studies were associated with an improvement in hepatic encephalopathy. However, a large meta-analysis has shown no demonstrable benefit with regard to clinically relevant outcomes (eg, mortality and quality of life). (See 'Hepatic encephalopathy' above and "Hepatic encephalopathy in adults: Treatment", section on 'Modification of colonic flora (prebiotics and probiotics)'.)

Allergy – A definitive role of probiotics for allergic conditions remains unproven, although initial results in studies of children with a variety of preparations for atopic dermatitis are promising. (See 'Allergy' above and "Prebiotics and probiotics for prevention of allergic disease".)

  1. Sartor RB. The intestinal microbiota in inflammatory bowel diseases. Nestle Nutr Inst Workshop Ser 2014; 79:29.
  2. Huttenhower C, Kostic AD, Xavier RJ. Inflammatory bowel disease as a model for translating the microbiome. Immunity 2014; 40:843.
  3. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014; 146:1489.
  4. Sartor RB, Wu GD. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017; 152:327.
  5. Cheifetz AS, Gianotti R, Luber R, Gibson PR. Complementary and Alternative Medicines Used by Patients With Inflammatory Bowel Diseases. Gastroenterology 2017; 152:415.
  6. Hedin CR, Mullard M, Sharratt E, et al. Probiotic and prebiotic use in patients with inflammatory bowel disease: a case-control study. Inflamm Bowel Dis 2010; 16:2099.
  7. Steidler L, Hans W, Schotte L, et al. Treatment of murine colitis by Lactococcus lactis secreting interleukin-10. Science 2000; 289:1352.
  8. Vandenbroucke K, Hans W, Van Huysse J, et al. Active delivery of trefoil factors by genetically modified Lactococcus lactis prevents and heals acute colitis in mice. Gastroenterology 2004; 127:502.
  9. Mohamadzadeh M, Pfeiler EA, Brown JB, et al. Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc Natl Acad Sci U S A 2011; 108 Suppl 1:4623.
  10. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008; 105:16731.
  11. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013; 500:232.
  12. El Hage R, Hernandez-Sanabria E, Van de Wiele T. Emerging Trends in "Smart Probiotics": Functional Consideration for the Development of Novel Health and Industrial Applications. Front Microbiol 2017; 8:1889.
  13. Mishima Y, Sartor RB. Manipulating resident microbiota to enhance regulatory immune function to treat inflammatory bowel diseases. J Gastroenterol 2020; 55:4.
  14. Oka A, Sartor RB. Microbial-Based and Microbial-Targeted Therapies for Inflammatory Bowel Diseases. Dig Dis Sci 2020; 65:757.
  15. Fedorak RN, Dieleman LA. Probiotics in the treatment of human inflammatory bowel diseases: update 2008. J Clin Gastroenterol 2008; 42 Suppl 2:S97.
  16. Naidoo K, Gordon M, Fagbemi AO, et al. Probiotics for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2011; :CD007443.
  17. Bernstein CN. Antibiotics, probiotics and prebiotics in IBD. Nestle Nutr Inst Workshop Ser 2014; 79:83.
  18. Shen J, Zuo ZX, Mao AP. Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn's disease, and pouchitis: meta-analysis of randomized controlled trials. Inflamm Bowel Dis 2014; 20:21.
  19. Kruis W. Probiotics. Dig Dis 2013; 31:385.
  20. Lichtenstein L, Avni-Biron I, Ben-Bassat O. Probiotics and prebiotics in Crohn's disease therapies. Best Pract Res Clin Gastroenterol 2016; 30:81.
  21. Singh S, Stroud AM, Holubar SD, et al. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev 2015; :CD001176.
  22. Derwa Y, Gracie DJ, Hamlin PJ, Ford AC. Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Aliment Pharmacol Ther 2017; 46:389.
  23. Rowland I, Capurso L, Collins K, et al. Current level of consensus on probiotic science--report of an expert meeting--London, 23 November 2009. Gut Microbes 2010; 1:436.
  24. Ritchie ML, Romanuk TN. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS One 2012; 7:e34938.
  25. Parker EA, Roy T, D'Adamo CR, Wieland LS. Probiotics and gastrointestinal conditions: An overview of evidence from the Cochrane Collaboration. Nutrition 2018; 45:125.
  26. Wilkins T, Sequoia J. Probiotics for Gastrointestinal Conditions: A Summary of the Evidence. Am Fam Physician 2017; 96:170.
  27. Rondanelli M, Faliva MA, Perna S, et al. Using probiotics in clinical practice: Where are we now? A review of existing meta-analyses. Gut Microbes 2017; 8:521.
  28. Palumbo P, Lombardi F, Cifone MG, Cinque B. The Epithelial Barrier Model Shows That the Properties of VSL#3 Depend from Where it is Manufactured. Endocr Metab Immune Disord Drug Targets 2019; 19:199.
  29. Vanderpool C, Yan F, Polk DB. Mechanisms of probiotic action: Implications for therapeutic applications in inflammatory bowel diseases. Inflamm Bowel Dis 2008; 14:1585.
  30. Maldonado Galdeano C, Cazorla SI, Lemme Dumit JM, et al. Beneficial Effects of Probiotic Consumption on the Immune System. Ann Nutr Metab 2019; 74:115.
  31. Sanders ME, Merenstein DJ, Reid G, et al. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol 2019; 16:605.
  32. Jones SE, Versalovic J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol 2009; 9:35.
  33. Seth A, Yan F, Polk DB, Rao RK. Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruption by a PKC- and MAP kinase-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 2008; 294:G1060.
  34. Yan F, Cao H, Cover TL, et al. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007; 132:562.
  35. Yan F, Cao H, Cover TL, et al. Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J Clin Invest 2011; 121:2242.
  36. McCarthy J, O'Mahony L, O'Callaghan L, et al. Double blind, placebo controlled trial of two probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. Gut 2003; 52:975.
  37. Pathmakanthan S, Li CK, Cowie J, Hawkey CJ. Lactobacillus plantarum 299: beneficial in vitro immunomodulation in cells extracted from inflamed human colon. J Gastroenterol Hepatol 2004; 19:166.
  38. Borruel N, Carol M, Casellas F, et al. Increased mucosal tumour necrosis factor alpha production in Crohn's disease can be downregulated ex vivo by probiotic bacteria. Gut 2002; 51:659.
  39. Lin YP, Thibodeaux CH, Peña JA, et al. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-Jun. Inflamm Bowel Dis 2008; 14:1068.
  40. Ng SC, Plamondon S, Kamm MA, et al. Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis. Inflamm Bowel Dis 2010; 16:1286.
  41. Rossi O, van Berkel LA, Chain F, et al. Faecalibacterium prausnitzii A2-165 has a high capacity to induce IL-10 in human and murine dendritic cells and modulates T cell responses. Sci Rep 2016; 6:18507.
  42. Dalmasso G, Cottrez F, Imbert V, et al. Saccharomyces boulardii inhibits inflammatory bowel disease by trapping T cells in mesenteric lymph nodes. Gastroenterology 2006; 131:1812.
  43. Thakur BK, Saha P, Banik G, et al. Live and heat-killed probiotic Lactobacillus casei Lbs2 protects from experimental colitis through Toll-like receptor 2-dependent induction of T-regulatory response. Int Immunopharmacol 2016; 36:39.
  44. Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med 2007; 13:35.
  45. Fujii T, Ohtsuka Y, Lee T, et al. Bifidobacterium breve enhances transforming growth factor beta1 signaling by regulating Smad7 expression in preterm infants. J Pediatr Gastroenterol Nutr 2006; 43:83.
  46. Riedel CU, Foata F, Philippe D, et al. Anti-inflammatory effects of bifidobacteria by inhibition of LPS-induced NF-kappaB activation. World J Gastroenterol 2006; 12:3729.
  47. O'Hara AM, O'Regan P, Fanning A, et al. Functional modulation of human intestinal epithelial cell responses by Bifidobacterium infantis and Lactobacillus salivarius. Immunology 2006; 118:202.
  48. Schultz M, Veltkamp C, Dieleman LA, et al. Lactobacillus plantarum 299V in the treatment and prevention of spontaneous colitis in interleukin-10-deficient mice. Inflamm Bowel Dis 2002; 8:71.
  49. Madsen KL, Doyle JS, Tavernini MM, et al. Antibiotic therapy attenuates colitis in interleukin 10 gene-deficient mice. Gastroenterology 2000; 118:1094.
  50. Sougioultzis S, Simeonidis S, Bhaskar KR, et al. Saccharomyces boulardii produces a soluble anti-inflammatory factor that inhibits NF-kappaB-mediated IL-8 gene expression. Biochem Biophys Res Commun 2006; 343:69.
  51. Preidis GA, Weizman AV, Kashyap PC, Morgan RL. AGA Technical Review on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020; 159:708.
  52. Pedrosa MC, Golner BB, Goldin BR, et al. Survival of yogurt-containing organisms and Lactobacillus gasseri (ADH) and their effect on bacterial enzyme activity in the gastrointestinal tract of healthy and hypochlorhydric elderly subjects. Am J Clin Nutr 1995; 61:353.
  53. Kailasapathy K, Chin J. Survival and therapeutic potential of probiotic organisms with reference to Lactobacillus acidophilus and Bifidobacterium spp. Immunol Cell Biol 2000; 78:80.
  54. Yılmaz İ, Dolar ME, Özpınar H. Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk J Gastroenterol 2019; 30:242.
  55. Wastyk HC, Fragiadakis GK, Perelman D, et al. Gut-microbiota-targeted diets modulate human immune status. Cell 2021; 184:4137.
  56. Jijon H, Backer J, Diaz H, et al. DNA from probiotic bacteria modulates murine and human epithelial and immune function. Gastroenterology 2004; 126:1358.
  57. Yan F, Polk DB. Probiotic bacterium prevents cytokine-induced apoptosis in intestinal epithelial cells. J Biol Chem 2002; 277:50959.
  58. Rachmilewitz D, Katakura K, Karmeli F, et al. Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 2004; 126:520.
  59. Katakura K, Lee J, Rachmilewitz D, et al. Toll-like receptor 9-induced type I IFN protects mice from experimental colitis. J Clin Invest 2005; 115:695.
  60. Wu X, Pan S, Luo W, et al. Roseburia intestinalis‑derived flagellin ameliorates colitis by targeting miR‑223‑3p‑mediated activation of NLRP3 inflammasome and pyroptosis. Mol Med Rep 2020; 22:2695.
  61. Round JL, Lee SM, Li J, et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011; 332:974.
  62. Fábrega MJ, Rodríguez-Nogales A, Garrido-Mesa J, et al. Intestinal Anti-inflammatory Effects of Outer Membrane Vesicles from Escherichia coli Nissle 1917 in DSS-Experimental Colitis in Mice. Front Microbiol 2017; 8:1274.
  63. Atarashi K, Tanoue T, Shima T, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011; 331:337.
  64. Komanduri S, Gillevet PM, Sikaroodi M, et al. Dysbiosis in pouchitis: evidence of unique microfloral patterns in pouch inflammation. Clin Gastroenterol Hepatol 2007; 5:352.
  65. Sokol H, Lay C, Seksik P, Tannock GW. Analysis of bacterial bowel communities of IBD patients: what has it revealed? Inflamm Bowel Dis 2008; 14:858.
  66. Reshef L, Kovacs A, Ofer A, et al. Pouch Inflammation Is Associated With a Decrease in Specific Bacterial Taxa. Gastroenterology 2015; 149:718.
  67. Dubinsky V, Reshef L, Bar N, et al. Predominantly Antibiotic-resistant Intestinal Microbiome Persists in Patients With Pouchitis Who Respond to Antibiotic Therapy. Gastroenterology 2020; 158:610.
  68. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology 2000; 119:305.
  69. Mimura T, Rizzello F, Helwig U, et al. Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut 2004; 53:108.
  70. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology 2003; 124:1202.
  71. Shen B, Brzezinski A, Fazio VW, et al. Maintenance therapy with a probiotic in antibiotic-dependent pouchitis: experience in clinical practice. Aliment Pharmacol Ther 2005; 22:721.
  72. Kuisma J, Mentula S, Jarvinen H, et al. Effect of Lactobacillus rhamnosus GG on ileal pouch inflammation and microbial flora. Aliment Pharmacol Ther 2003; 17:509.
  73. Bengtsson J, Adlerberth I, Östblom A, et al. Effect of probiotics (Lactobacillus plantarum 299 plus Bifidobacterium Cure21) in patients with poor ileal pouch function: a randomised controlled trial. Scand J Gastroenterol 2016; 51:1087.
  74. Holubar SD, Cima RR, Sandborn WJ, Pardi DS. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev 2010; :CD001176.
  75. Nguyen N, Zhang B, Holubar SD, et al. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev 2019; 11:CD001176.
  76. Kaur L, Gordon M, Baines PA, et al. Probiotics for induction of remission in ulcerative colitis. Cochrane Database Syst Rev 2020; 3:CD005573.
  77. Iheozor-Ejiofor Z, Kaur L, Gordon M, et al. Probiotics for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2020; 3:CD007443.
  78. Limketkai BN, Akobeng AK, Gordon M, Adepoju AA. Probiotics for induction of remission in Crohn's disease. Cochrane Database Syst Rev 2020; 7:CD006634.
  79. Tian C, Huang Y, Wu X, et al. The Efficacy and Safety of Mesalamine and Probiotics in Mild-to-Moderate Ulcerative Colitis: A Systematic Review and Meta-Analysis. Evid Based Complement Alternat Med 2020; 2020:6923609.
  80. Ganji-Arjenaki M, Rafieian-Kopaei M. Probiotics are a good choice in remission of inflammatory bowel diseases: A meta analysis and systematic review. J Cell Physiol 2018; 233:2091.
  81. Dong J, Teng G, Wei T, et al. Methodological Quality Assessment of Meta-Analyses and Systematic Reviews of Probiotics in Inflammatory Bowel Disease and Pouchitis. PLoS One 2016; 11:e0168785.
  82. Rembacken BJ, Snelling AM, Hawkey PM, et al. Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet 1999; 354:635.
  83. Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 2004; 53:1617.
  84. Henker J, Müller S, Laass MW, et al. Probiotic Escherichia coli Nissle 1917 (EcN) for successful remission maintenance of ulcerative colitis in children and adolescents: an open-label pilot study. Z Gastroenterol 2008; 46:874.
  85. Matthes H, Krummenerl T, Giensch M, et al. Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complement Altern Med 2010; 10:13.
  86. Zocco MA, dal Verme LZ, Cremonini F, et al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther 2006; 23:1567.
  87. Furrie E, Macfarlane S, Kennedy A, et al. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut 2005; 54:242.
  88. Tursi A, Brandimarte G, Giorgetti GM, et al. Low-dose balsalazide plus a high-potency probiotic preparation is more effective than balsalazide alone or mesalazine in the treatment of acute mild-to-moderate ulcerative colitis. Med Sci Monit 2004; 10:PI126.
  89. Miele E, Pascarella F, Giannetti E, et al. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol 2009; 104:437.
  90. Sood A, Midha V, Makharia GK, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol 2009; 7:1202.
  91. Bibiloni R, Fedorak RN, Tannock GW, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol 2005; 100:1539.
  92. Huynh HQ, deBruyn J, Guan L, et al. Probiotic preparation VSL#3 induces remission in children with mild to moderate acute ulcerative colitis: a pilot study. Inflamm Bowel Dis 2009; 15:760.
  93. Tursi A, Brandimarte G, Papa A, et al. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J Gastroenterol 2010; 105:2218.
  94. Wildt S, Nordgaard I, Hansen U, et al. A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis. J Crohns Colitis 2011; 5:115.
  95. Oliva S, Di Nardo G, Ferrari F, et al. Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment Pharmacol Ther 2012; 35:327.
  96. Palumbo VD, Romeo M, Marino Gammazza A, et al. The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:372.
  97. Doherty GA, Bennett GC, Cheifetz AS, Moss AC. Meta-analysis: targeting the intestinal microbiota in prophylaxis for post-operative Crohn's disease. Aliment Pharmacol Ther 2010; 31:802.
  98. Rolfe VE, Fortun PJ, Hawkey CJ, Bath-Hextall F. Probiotics for maintenance of remission in Crohn's disease. Cochrane Database Syst Rev 2006; :CD004826.
  99. Whelan K, Quigley EM. Probiotics in the management of irritable bowel syndrome and inflammatory bowel disease. Curr Opin Gastroenterol 2013; 29:184.
  100. Bourreille A, Cadiot G, Le Dreau G, et al. Saccharomyces boulardii does not prevent relapse of Crohn's disease. Clin Gastroenterol Hepatol 2013; 11:982.
  101. Steed H, Macfarlane GT, Blackett KL, et al. Clinical trial: the microbiological and immunological effects of synbiotic consumption - a randomized double-blind placebo-controlled study in active Crohn's disease. Aliment Pharmacol Ther 2010; 32:872.
  102. Haziri D, Prechter F, Stallmach A. [Yoghurt-induced Lactobacillus bacteremia in a patient with Crohn's disease on therapy with ustekinumab and concomitant HIV-Infection]. Z Gastroenterol 2021; 59:317.
  103. Liu MM, Li ST, Shu Y, Zhan HQ. Probiotics for prevention of radiation-induced diarrhea: A meta-analysis of randomized controlled trials. PLoS One 2017; 12:e0178870.
  104. Packey CD, Ciorba MA. Microbial influences on the small intestinal response to radiation injury. Curr Opin Gastroenterol 2010; 26:88.
  105. Guo H, Chou WC, Lai Y, et al. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites. Science 2020; 370.
  106. Van Niel CW, Feudtner C, Garrison MM, Christakis DA. Lactobacillus therapy for acute infectious diarrhea in children: a meta-analysis. Pediatrics 2002; 109:678.
  107. Szajewska H, Skórka A, Ruszczyński M, Gieruszczak-Białek D. Meta-analysis: Lactobacillus GG for treating acute diarrhoea in children. Aliment Pharmacol Ther 2007; 25:871.
  108. Britton RA, Versalovic J. Probiotics and gastrointestinal infections. Interdiscip Perspect Infect Dis 2008; 2008:290769.
  109. Szajewska H, Skórka A. Saccharomyces boulardii for treating acute gastroenteritis in children: updated meta-analysis of randomized controlled trials. Aliment Pharmacol Ther 2009; 30:960.
  110. Allen SJ, Martinez EG, Gregorio GV, Dans LF. Probiotics for treating acute infectious diarrhoea. Cochrane Database Syst Rev 2010; :CD003048.
  111. Bernaola Aponte G, Bada Mancilla CA, Carreazo NY, Rojas Galarza RA. Probiotics for treating persistent diarrhoea in children. Cochrane Database Syst Rev 2013; :CD007401.
  112. Schnadower D, Tarr PI, Casper TC, et al. Lactobacillus rhamnosus GG versus Placebo for Acute Gastroenteritis in Children. N Engl J Med 2018; 379:2002.
  113. Freedman SB, Williamson-Urquhart S, Farion KJ, et al. Multicenter Trial of a Combination Probiotic for Children with Gastroenteritis. N Engl J Med 2018; 379:2015.
  114. Freedman SB, Xie J, Nettel-Aguirre A, et al. A randomized trial evaluating virus-specific effects of a combination probiotic in children with acute gastroenteritis. Nat Commun 2020; 11:2533.
  115. Szajewska H, Guarino A, Hojsak I, et al. Use of Probiotics for the Management of Acute Gastroenteritis in Children: An Update. J Pediatr Gastroenterol Nutr 2020; 71:261.
  116. McFarland LV. Meta-analysis of probiotics for the prevention of traveler's diarrhea. Travel Med Infect Dis 2007; 5:97.
  117. Tromm A, Niewerth U, Khoury M, et al. The probiotic E. coli strain Nissle 1917 for the treatment of collagenous colitis: first results of an open-label trial. Z Gastroenterol 2004; 42:365.
  118. Wildt S, Munck LK, Vinter-Jensen L, et al. Probiotic treatment of collagenous colitis: a randomized, double-blind, placebo-controlled trial with Lactobacillus acidophilus and Bifidobacterium animalis subsp. Lactis. Inflamm Bowel Dis 2006; 12:395.
  119. De Angelis M, Rizzello CG, Fasano A, et al. VSL#3 probiotic preparation has the capacity to hydrolyze gliadin polypeptides responsible for Celiac Sprue. Biochim Biophys Acta 2006; 1762:80.
  120. Di Cagno R, Barbato M, Di Camillo C, et al. Gluten-free sourdough wheat baked goods appear safe for young celiac patients: a pilot study. J Pediatr Gastroenterol Nutr 2010; 51:777.
  121. Duar RM, Clark KJ, Patil PB, et al. Identification and characterization of intestinal lactobacilli strains capable of degrading immunotoxic peptides present in gluten. J Appl Microbiol 2015; 118:515.
  122. Francavilla R, De Angelis M, Rizzello CG, et al. Selected Probiotic Lactobacilli Have the Capacity To Hydrolyze Gluten Peptides during Simulated Gastrointestinal Digestion. Appl Environ Microbiol 2017; 83.
  123. Giorgi A, Cerrone R, Capobianco D, et al. A Probiotic Preparation Hydrolyzes Gliadin and Protects Intestinal Cells from the Toxicity of Pro-Inflammatory Peptides. Nutrients 2020; 12.
  124. Lorenzo Pisarello MJ, Vintiñi EO, González SN, et al. Decrease in lactobacilli in the intestinal microbiota of celiac children with a gluten-free diet, and selection of potentially probiotic strains. Can J Microbiol 2015; 61:32.
  125. Seiler CL, Kiflen M, Stefanolo JP, et al. Probiotics for Celiac Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am J Gastroenterol 2020; 115:1584.
  126. Smecuol E, Hwang HJ, Sugai E, et al. Exploratory, randomized, double-blind, placebo-controlled study on the effects of Bifidobacterium infantis natren life start strain super strain in active celiac disease. J Clin Gastroenterol 2013; 47:139.
  127. Olivares M, Castillejo G, Varea V, Sanz Y. Double-blind, randomised, placebo-controlled intervention trial to evaluate the effects of Bifidobacterium longum CECT 7347 in children with newly diagnosed coeliac disease. Br J Nutr 2014; 112:30.
  128. Harris RG, Neale EP, Ferreira I. When poorly conducted systematic reviews and meta-analyses can mislead: a critical appraisal and update of systematic reviews and meta-analyses examining the effects of probiotics in the treatment of functional constipation in children. Am J Clin Nutr 2019; 110:177.
  129. Martínez-Martínez MI, Calabuig-Tolsá R, Cauli O. The effect of probiotics as a treatment for constipation in elderly people: A systematic review. Arch Gerontol Geriatr 2017; 71:142.
  130. Koebnick C, Wagner I, Leitzmann P, et al. Probiotic beverage containing Lactobacillus casei Shirota improves gastrointestinal symptoms in patients with chronic constipation. Can J Gastroenterol 2003; 17:655.
  131. Yang YX, He M, Hu G, et al. Effect of a fermented milk containing Bifidobacterium lactis DN-173010 on Chinese constipated women. World J Gastroenterol 2008; 14:6237.
  132. Chmielewska A, Szajewska H. Systematic review of randomised controlled trials: probiotics for functional constipation. World J Gastroenterol 2010; 16:69.
  133. Nishida S, Gotou M, Akutsu S, et al. Effect of yogurt containing Bifidobacterium lactis BB-12 on improvement of defecation and fecal microflora of healthy female adults. Milk Science 2004; 53:71.
  134. Sadeghzadeh M, Rabieefar A, Khoshnevisasl P, et al. The effect of probiotics on childhood constipation: a randomized controlled double blind clinical trial. Int J Pediatr 2014; 2014:937212.
  135. Riezzo G, Orlando A, D'Attoma B, et al. Randomised double blind placebo controlled trial on Lactobacillus reuteri DSM 17938: improvement in symptoms and bowel habit in functional constipation. Benef Microbes 2018; 9:51.
  136. Dimidi E, Christodoulides S, Fragkos KC, et al. The effect of probiotics on functional constipation in adults: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2014; 100:1075.
  137. Miller LE, Ouwehand AC, Ibarra A. Effects of probiotic-containing products on stool frequency and intestinal transit in constipated adults: systematic review and meta-analysis of randomized controlled trials. Ann Gastroenterol 2017; 30:629.
  138. Wojtyniak K, Szajewska H. Systematic review: probiotics for functional constipation in children. Eur J Pediatr 2017; 176:1155.
  139. Huang R, Hu J. Positive Effect of Probiotics on Constipation in Children: A Systematic Review and Meta-Analysis of Six Randomized Controlled Trials. Front Cell Infect Microbiol 2017; 7:153.
  140. O'Sullivan MA, O'Morain CA. Bacterial supplementation in the irritable bowel syndrome. A randomised double-blind placebo-controlled crossover study. Dig Liver Dis 2000; 32:294.
  141. Nobaek S, Johansson ML, Molin G, et al. Alteration of intestinal microflora is associated with reduction in abdominal bloating and pain in patients with irritable bowel syndrome. Am J Gastroenterol 2000; 95:1231.
  142. Whorwell PJ, Altringer L, Morel J, et al. Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol 2006; 101:1581.
  143. Brenner DM, Moeller MJ, Chey WD, Schoenfeld PS. The utility of probiotics in the treatment of irritable bowel syndrome: a systematic review. Am J Gastroenterol 2009; 104:1033.
  144. O'Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology 2005; 128:541.
  145. Levri KM, Ketvertis K, Deramo M, et al. Do probiotics reduce adult lactose intolerance? A systematic review. J Fam Pract 2005; 54:613.
  146. Francavilla R, Miniello V, Magistà AM, et al. A randomized controlled trial of Lactobacillus GG in children with functional abdominal pain. Pediatrics 2010; 126:e1445.
  147. Ki Cha B, Mun Jung S, Hwan Choi C, et al. The effect of a multispecies probiotic mixture on the symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. J Clin Gastroenterol 2012; 46:220.
  148. Sisson G, Ayis S, Sherwood RA, Bjarnason I. Randomised clinical trial: A liquid multi-strain probiotic vs. placebo in the irritable bowel syndrome--a 12 week double-blind study. Aliment Pharmacol Ther 2014; 40:51.
  149. Stevenson C, Blaauw R, Fredericks E, et al. Randomized clinical trial: effect of Lactobacillus plantarum 299 v on symptoms of irritable bowel syndrome. Nutrition 2014; 30:1151.
  150. Staudacher HM, Lomer MCE, Farquharson FM, et al. A Diet Low in FODMAPs Reduces Symptoms in Patients With Irritable Bowel Syndrome and A Probiotic Restores Bifidobacterium Species: A Randomized Controlled Trial. Gastroenterology 2017; 153:936.
  151. Ringel-Kulka T, McRorie J, Ringel Y. Multi-Center, Double-Blind, Randomized, Placebo-Controlled, Parallel-Group Study to Evaluate the Benefit of the Probiotic Bifidobacterium infantis 35624 in Non-Patients With Symptoms of Abdominal Discomfort and Bloating. Am J Gastroenterol 2017; 112:145.
  152. Giannetti E, Maglione M, Alessandrella A, et al. A Mixture of 3 Bifidobacteria Decreases Abdominal Pain and Improves the Quality of Life in Children With Irritable Bowel Syndrome: A Multicenter, Randomized, Double-Blind, Placebo-Controlled, Crossover Trial. J Clin Gastroenterol 2017; 51:e5.
  153. Zhang Y, Li L, Guo C, et al. Effects of probiotic type, dose and treatment duration on irritable bowel syndrome diagnosed by Rome III criteria: a meta-analysis. BMC Gastroenterol 2016; 16:62.
  154. Lyra A, Hillilä M, Huttunen T, et al. Irritable bowel syndrome symptom severity improves equally with probiotic and placebo. World J Gastroenterol 2016; 22:10631.
  155. Moayyedi P, Ford AC, Talley NJ, et al. The efficacy of probiotics in the treatment of irritable bowel syndrome: a systematic review. Gut 2010; 59:325.
  156. Camilleri M. Probiotics and irritable bowel syndrome: rationale, mechanisms, and efficacy. J Clin Gastroenterol 2008; 42 Suppl 3 Pt 1:S123.
  157. McFarland LV, Dublin S. Meta-analysis of probiotics for the treatment of irritable bowel syndrome. World J Gastroenterol 2008; 14:2650.
  158. Chey WD. SYMPOSIUM REPORT: An Evidence-Based Approach to IBS and CIC: Applying New Advances to Daily Practice: A Review of an Adjunct Clinical Symposium of the American College of Gastroenterology Meeting October 16, 2016 • Las Vegas, Nevada. Gastroenterol Hepatol (N Y) 2017; 13:1.
  159. Ringel Y, Carroll IM. Alterations in the intestinal microbiota and functional bowel symptoms. Gastrointest Endosc Clin N Am 2009; 19:141.
  160. Carroll IM, Ringel-Kulka T, Keku TO, et al. Molecular analysis of the luminal- and mucosal-associated intestinal microbiota in diarrhea-predominant irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2011; 301:G799.
  161. Sen S, Mullan MM, Parker TJ, et al. Effect of Lactobacillus plantarum 299v on colonic fermentation and symptoms of irritable bowel syndrome. Dig Dis Sci 2002; 47:2615.
  162. Drouault-Holowacz S, Bieuvelet S, Burckel A, et al. A double blind randomized controlled trial of a probiotic combination in 100 patients with irritable bowel syndrome. Gastroenterol Clin Biol 2008; 32:147.
  163. Guglielmetti S, Mora D, Gschwender M, Popp K. Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life--a double-blind, placebo-controlled study. Aliment Pharmacol Ther 2011; 33:1123.
  164. Andresen V, Gschossmann J, Layer P. Heat-inactivated Bifidobacterium bifidum MIMBb75 (SYN-HI-001) in the treatment of irritable bowel syndrome: a multicentre, randomised, double-blind, placebo-controlled clinical trial. Lancet Gastroenterol Hepatol 2020; 5:658.
  165. Oak SJ, Jha R. The effects of probiotics in lactose intolerance: A systematic review. Crit Rev Food Sci Nutr 2019; 59:1675.
  166. Pakdaman MN, Udani JK, Molina JP, Shahani M. The effects of the DDS-1 strain of lactobacillus on symptomatic relief for lactose intolerance - a randomized, double-blind, placebo-controlled, crossover clinical trial. Nutr J 2016; 15:56.
  167. Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet 2008; 371:651.
  168. Capurso G, Marignani M, Piciucchi M, et al. Probiotics and severe acute pancreatitis. J Clin Gastroenterol 2008; 42 Suppl 3 Pt 1:S148.
Topic 2603 Version 37.0

References

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