Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

CLINICAL GASTROENTEROLOGY AND HEPATOLOGY 2012;10:960 –968

PERSPECTIVES IN CLINICAL
GASTROENTEROLOGY AND HEPATOLOGY

A Gastroenterologist’s Guide to Probiotics

MATTHEW A. CIORBA
Department of Medicine, Division of Gastroenterology, Washington University, St Louis School of Medicine, St Louis, Missouri

The Human Microbiome and Probiotic


Podcast interview: www.gastro.org/cghpodcast. Mechanisms
Also available on iTunes.
To understand the role that probiotics may have in
influencing health, it is important to have an appreciation of
The enteric microbiota contribute to gastrointestinal the roles of the normal intestinal microbiome (commensal
health, and their disruption has been associated with many microbiota). The human GI tract is host to over 500 bacterial
disease states. Some patients consume probiotic products species as well as a less well-described virome. These microbiota
in attempts to manipulate the intestinal microbiota for form a virtual bioreactor facilitating digestion, nutrient provi-
health benefit. It is important for gastroenterologists to sion, and the shaping of our immune system.2 Our intestinal
improve their understanding of the mechanisms of probi- bacteria weigh up to 1 kg and bacterial cells outnumber human
otics and the evidence that support their use in practice. cells by 10:1. The bacterial genome may outnumber the human
Clinical trials have assessed the therapeutic effects of pro- genome by 100:1. Nutritional factors including several B vita-
biotic agents for several disorders, including antibiotic- or mins, vitamin K, folate, and short-chain fatty acids are pro-
Clostridium difficile–associated diarrhea, irritable bowel syn- duced by these bacteria. Up to 10% of an individual’s daily
drome, and the inflammatory bowel diseases. Although energy needs can be derived from the by-products of bacterial
probiotic research is a rapidly evolving field, there are suf- fermentation. Gastrointestinal microbiota are also critical for
ficient data to justify a trial of probiotics for treatment or normal immune system development.3 The physiologic impact
prevention of some of these conditions. However, the ca- mediated by our resident microbes is substantial enough to
pacity of probiotics to modify disease symptoms is likely to have earned the label of “other organ” from some.4
be modest and varies among probiotic strains—not all pro- Beyond contributing to or modifying the metabolic and
biotics are right for all diseases. The current review pro- nutritional functions of the commensal microbiota, probiotic
vides condition-specific rationale for using probiotic ther- bacteria have several putative mechanisms by which they may
apy and literature-based recommendations. confer specific beneficial effects. General categories include
Keywords: Clostridium difficile; IBD; IBS; Crohn’s Disease and modulation of immune or sensory-motor function, enhance-
Colitis; Yogurt; Pouchitis. ment of mucosal barrier function, and antipathogen effects
(Figure 1).5–7 Some of these mechanisms have been worked out
in animal models and/or in vitro systems only.
Soluble products secreted or shed by probiotics also mediate

F or more than a hundred years it has been recognized that


certain microorganisms may impart health benefits to the
host when administered in adequate amounts. These microor-
important physiologic benefits; thus, viable bacteria are not
necessarily required for all benefits.8,9 The mechanisms by
which probiotics exert benefit varies by specific probiotic strain
ganisms, termed probiotics, have recently become a topic of and likely depends on the clinical indication.10,11 Therefore, as
significant focus in basic and clinical investigation. Relevant to with antibiotic prescribing, clinical use of probiotics should
the practice of gastroenterology, probiotics are commonly used focus on matching the probiotic strain and dosage to the
by patients with gastrointestinal (GI) complaints or diseases. condition for which it has shown benefit in clinical trials. In the
Increasingly, probiotics are also being recommended by the future, greater understanding of probiotic-specific mechanisms
clinicians who treat these conditions.1
The goal of this review is to provide clinicians with an
overview of the rationale and data which support or refute the Abbreviations used in this paper: AAD, antibiotic-associated diar-
role of probiotics for treating commonly encountered GI dis- rhea; CD, Crohn’s disease; CDAD, Clostridium difficile–associated di-
arrhea; ECN, Escherichia coli Nissle 1917; GI, gastrointestinal; IBS,
orders. The information provided is based on review of primary
irritable bowel syndrome; LGG, Lactobacillus rhamnosus GG; NNT,
literature from randomized controlled trials (RCTs), meta-anal-
number needed-to-treat; RCT, randomized controlled trial; UC, ulcer-
yses, expert consensus panel recommendations, and society- ative colitis.
based practice recommendations. References are provided for © 2012 by the AGA Institute
more in-depth reading and tables or figures summarize key 1542-3565/$36.00
information. http://dx.doi.org/10.1016/j.cgh.2012.03.024
September 2012 PROBIOTICS IN PRACTICE 961

Figure 1. Mechanisms of action of probiotics in the gastrointestinal tract.

could allow for precise selection of a particular probiotic strain synbiotic product. Lactobacillus and Bifidobacterium species are
to target a patient’s specific pathogenic defect and clinical the most commonly used probiotics. However, 1 of the first
problem. probiotics, which is still in use, is the nonpathogenic Esche-
richia coli Nissle 1917 (ECN). Most probiotics were initially
Probiotic Concepts for Practice cultured from humans and resemble known commensal gut
bacteria. However, the commensal population they resemble
What Makes a Probiotic a Probiotic? typically represents only a fraction of the total luminal
Definitions of the terms probiotic, prebiotic, and syn- bacteria. Saccharomyces boulardii is a probiotic yeast strain
biotic are provided in Table 1. This review focuses on probi- with the potential advantage of having resistance to most
otics, though some probiotics have been tested as part of a antibiotics.
962 MATTHEW A. CIORBA CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 10, No. 9

Table 1. Definitions otic strains do not colonize the gut and are no longer recover-
able in stool 1 to 4 weeks after stopping consumption.13 For
Probiotics Live microorganisms that confer a health benefit
on the host when administered in adequate example, McNulty and colleagues recently evaluated a fer-
amounts mented milk product with probiotic strains matching the com-
Prebiotic Dietary substances that nurture specific changes mercially available Activia (Dannon, White Plains, New York).
in the composition and/or activity of the GI The investigators showed that the probiotic product did not
microbiota (favoring beneficial bacteria), thus change the gut’s overall bacterial composition, but instead
conferring benefit(s) upon host health altered gene expression patterns relevant to carbohydrate me-
Synbiotics Products that contain both probiotics and tabolism in the host’s resident gut microbes.14 These changes in
prebiotics the human fecal “metatranscriptome” were transient, confined
NOTE. Adapted from Guarner F, Khan AG, Garisch J, et al. Probiotics only to the time of the probiotic consumption. Thus, if sus-
and prebiotics: world gastroenterology organisation global guidelines, tained benefit from a probiotic is desired, continued consump-
2011. Available at: http://www.worldgastroenterology.org/probiotics- tion is likely required.
prebiotics.html.48
Where Can Probiotics Fit Into a Therapeutic
Algorithm?
According to current definitions, probiotics should survive Data for probiotic use in several GI disorders are re-
both gastric acid and bile to reach the small intestine and colon viewed in the following section. For AAD and viral gastroen-
where they exert their effects. Clinical and basic investigations teritis, supporting data are strong and probiotics are among the
on probiotics have used a multitude of probiotic species, both only treatment modalities available. However, the duration of
as single strains and multispecies products. Many of these symptoms in these conditions is typically short regardless of
probiotics are available in a lyophilized (freeze-dried) pill form, probiotic use. In ulcerative colitis (UC), pouchitis, and irritable
though some are available in yogurt or as packets (sachets) bowel syndrome (IBS), adequate data exist for clinicians to
which can be mixed into noncarbonated drinks. Whether syn- consider recommending a therapeutic trial of specific probiotic
ergism or antagonism exists between probiotic species when strains or preparations in selected patients. In these conditions
offered together has not been examined in clinical studies, probiotics are usually administered as adjunctive therapy,
though both scenarios are theoretically possible. Though not rather than primary or first-line therapy. The decision to rec-
exhaustive, Table 2 lists several of the more commonly available ommend probiotic therapy ultimately depends on the clinical
probiotic preparations which have shown benefit in human scenario, patient interest, and clinician preference. In hepatic
trials. Probiotics are considered dietary supplements; thus, they encephalopathy, Crohn’s disease (CD), and Clostridium difficile–
are not covered by medical insurance and their production is associated diarrhea (CDAD), conventional medical therapies
not regulated by the Food and Drug Administration. As such, remain the gold standard. Practice relevant probiotic concepts
product quality, purity, and viability have been reported to be are summarized in Table 3.
variable.12 However, several clinically tested probiotic products
with quality-controlled production are now marketed by repu-
table companies.
Probiotic Therapy for Gastrointestinal
Conditions
Does Any Yogurt Work Just Like a Probiotic? Acute Onset Infectious Diarrhea
Lactic acid–producing bacteria have been used for cen- Several RCTs have evaluated the use of probiotics in
turies in food fermentation. Many yogurts contain live-active acute infectious diarrhea. The data are largely from pediatric
lactobacillus cultures and are considered functional food prod- studies where both prevention and treatment were examined.
ucts; however, most are not considered probiotics per se. This Trials were conducted across the world with durations of up to
term is reserved for products with an adequate number of 1 year. In the pediatric population, rotavirus has been the most
microorganisms at time of consumption specifically shown to common cause of infectious diarrhea. Data suggest that the
confer health benefits in controlled human trials. Yogurts for- benefit of probiotics in preventing acute infectious diarrhea is
tified with an adequate number of viable bacteria shown to modest.15,16 Lactobacillus rhamnosus GG (LGG), Lactobacillus reu-
exert benefit in controlled trials are classified as probiotics. teri and Lactobacillus casei all have shown benefit, with an ap-
Given this information, and the knowledge that probiotic ben- proximate number needed-to-treat (NNT) of 7 children to pre-
efits appear species-specific, expected clinical end points may vent 1 case of rotavirus in the child care center setting.17–19 With
not be achieved by generically recommending yogurt to patients the currently available rotavirus vaccine in consideration, the
in whom a purported probiotic benefit is desired. It should be American Academy of Pediatrics states that probiotics for pre-
noted, however, that yogurt consumption has other benefits venting acute infectious diarrhea are not universally endorsed,
including improved lactose tolerance and the provision of pro- but acknowledges that they may have a role in special circum-
tein, vitamin D, and calcium. stances.15 According to the US Center for Disease Control, data
are not sufficient to support the use of probiotics such as LGG
How Long Does One Have to Take a to prevent traveler’s diarrhea of bacterial origin.
Probiotic? The data supporting treatment of acute infectious diarrhea
As viable microorganisms, probiotics can survive in the with probiotics are stronger. LGG is the most effective probiotic
human gut and impact microbes which colonize the gut. Pro- reported to date, reducing both severity and duration of diar-
biotics are often detectable in the stool by culture or gene-based rhea by approximately 1 day.20,21 The American Academy of
assays during periods of consumption. However, many probi- Pediatrics supports the recommendation of LGG early in the
September 2012
Table 2. Available Probiotic Products Specifically Tested for Gastrointestinal Disorders

Practice
Brand name (Company) Bacterial species Clinical condition Effectiveness32,42,a guidelines15,48,84,b Bacteria count/dosing Cost/quantity

Activia (Dannon, White B lactis DN-173010 (plus IBS C 1bc 4 oz/cup, 1–4 qd $10–$18/24
Plains, NY) yogurt starters L bulgaricus, count
L lactis, and Streptococcus
thermophilus)
Align (Proctor and Gamble, B infantis 35624 IBS B 1bc 1 Billion/1 qd $29.99/28
Cincinnati, OH) count
BioGaia (Everidis Health L reuteri Protectis Infectious diarrhea treatment A 1ad 100 Million qd $22–$30/5 mL
Sciences, St Louis, MO) SD2112 (ATCC 55730) IBS C 1bd (29 servings)
Bio-K⫹ (Bio-K Plus L acidophilus CL1285 and L AAD prevention 1bc 50 Billion/capsule bid $29.99/15
International, Inc, Laval, casei LBC80R CDAD prevention 1bc count
QC, Canada)
Culturelle (Valio, Helsinki, LGG (LGG also included in AAD prevention A AAP, 1bc, 1bd 10 Billion/1 qd $18–$25/30
Finland/i-Health, Inc, Danimals yogurt; Dannon) Infectious diarrhea treatment A AAP, 1ad, 2bc count
Cromwell, CT) Infectious diarrhea prevention B 1bc, 1bd
CDAD prevention B/C
CDAD prevention of recurrence B/C
Crohn’s disease C
IBS B/C (children) 1ad,1bc,e

Danactive (Dannon) L casei DN-114001 AAD prevention A 1bc 3.1 oz/cup; 10 $5.00/8 count
Infectious diarrhea prevention 1bd billion/cup
CDAD prevention 1bc
Florastor (Biocodex, Inc, Saccharomyces boulardii AAD prevention A AAP, 1ad, 1bc 250 mg/1 bid $19.99/20
Creswell, OR) (yeast) Infectious diarrhea treatment A 1ad, 1bc count
Infectious diarrhea prevention B
CDAD prevention B/C
CDAD prevention of recurrence B/C 1bc
Crohn’s disease C
Mutaflor (Ardeypharm, ECN UC induction B 100 mg/capsule bid $62–$81/60
Herdecke, Germany) UC maintenance A 1bc, BSG “A” Canadaf
VSL*3 (Sigma-Tau Combination probiotic product IBS B/C 122.5 Billion/capsule; $86/30
Pharmaceuticals, Inc, (Streptococcus UC induction B 1bc 450 billion/sachet; sachets
Towson, MA) thermophilus, B breve, B UC maintenance A IBS: ½–1 sachet/d; $52/60 count
longum, B infantis, L Pouchitis: prevention and A 1bc, BSG “B” Pouchitis: 2–4
acidophilus, L plantarum, L maintaining remission sachets/d; UC: 1–8
paracasei, L delbreuckii/ sachets/d

PROBIOTICS IN PRACTICE
bulgaricus)

aEffectiveness based on expert panel recommendations where: A ⫽ strong, positive, well-conducted, controlled studies in the primary literature; B ⫽ some positive, controlled studies but presence

of some negative studies or inadequate amount of work to establish the certainty; C ⫽ some positive studies but clearly inadequate amount of work to establish the certainty.
bPractice guidelines include those of (1) World Gastroenterology Organisation’s global guidelines evidence level assignment48 where 1a ⫽ systematic review with homogeneity of RCTs; 1b ⫽

individual RCT with narrow confidence interval; and 2b ⫽ individual cohort study (including low quality RCT)85; (2) American Academy of Pediatrics recommended (AAP)15; (3) British Society of
Gastroenterology guidelines Grade of Evidence (BSG)84: grade A indicates consistent results among RCTs, grade B indicates consistent cohort studies or smaller RCTs.
cAdult.
dPediatric.
eAs part of a multispecies probiotic product.

963
fECN is currently removed from the US market respecting the FDA’s decision on the classification of the product as a “biologic” instead of its former status as a “medical food.”
964 MATTHEW A. CIORBA CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 10, No. 9

Table 3. Practical Considerations Relevant to Probiotics in treatment paradigm.28 Recurrence of CDAD remains a clinical
Practice problem. In 1994, a trial reported that S boulardii (500 mg twice
a day) offered for 4 weeks after antibiotic therapy reduced
Common side effects of probiotics are typically transient but
include gas and bloating
overall CDAD recurrence rates.29 However, the finding was only
Different probiotic strains possess unique properties for benefiting significant for those with a history of recurrent CDAD. A
host physiology follow-up study, designed to be confirmatory, did not find S
One probiotic does not fit all GI illnesses; probiotic selection should boulardii to significantly reduce CDAD recurrence after standard
be based on the clinical indication and take into consideration therapy.30 Though a favorable trend was found in patients
the strain and dosage used in clinical trials treated with high-dose vancomycin (2 g/d) in the latter study,
Symptomatic benefits offered by probiotics are likely to be modest; the clinical significance of this is less clear. Lactobacillus pro-
thus, probiotic therapies may best be used to supplement rather biotics have been tested as single species and as combination
than replace conventional therapies probiotic products for preventing CDAD recurrence. While
Continuous consumption throughout the period of desired effect
some results have been promising, most studies are underpow-
appears required for probiotics
Avoid probiotics in the critically ill and those with severe immune
ered, have methodological flaws, or have not been reproduced.31
compromise Probiotic-based primary prevention still may be an approach
to the current scourge of C difficile. The 2 recent probiotic trials
discussed previously in the AAD section suggest that this may
be feasible. The Hickson study reported that DanActive (Dan-
course of acute infectious diarrhea to reduce symptom dura- non) supplementation in older hospitalized adults reduced
tion.15 AAD, but also CDAD (0% vs 17% placebo).25 The study evalu-
ating the combination probiotic Bio-K⫹ (Bio-K Plus Interna-
Antibiotic Associated Diarrhea tional) also showed a reduction in CDAD in the treated cohort
Antibiotic use is common in children, and diarrhea (1.2% vs 23.8% placebo).26 The high incidence rate of C difficile
develops in approximately 20% of those taking antibiotics. positivity in the placebo groups (17% and 23.8%) is a criticism
Prevention of non–C difficile-related antibiotic-associated diar- for both of these studies. Nonetheless, if confirmatory studies
rhea (AAD) with probiotics has been assessed in RCTs. A 2011 show similar results, these intriguing findings may lead to a
Cochrane Review evaluating more than 3400 patients from 16 paradigm shift in managing older adults requiring antibiotic
studies concluded that the overall evidence suggests a protective therapy.
effect of probiotics in preventing AAD.22 Studies using LGG While controversy exists, current society guidelines and ex-
and S boulardii produced the most convincing results.23 The pert opinion panels state that existing data are not sufficient to
NNT to prevent 1 case of AAD was approximately 7 in the justify recommending available probiotics for preventing pri-
Cochrane Review. The American Academy of Pediatrics sup- mary or recurrent CDAD.31–33
ports the recommendation of probiotics for prevention of, but
not treatment of, AAD.15 Irritable Bowel Syndrome
In the adult population probiotics also appear effective in Irritable bowel syndrome is characterized by symptoms
limiting AAD. A meta-analysis evaluating studies on various of abdominal pain and altered bowel habits which occur over at
probiotics and antibiotic regimens published between 1977 and least 3 months. This common disorder is managed with varying
2005 found that both LGG and S boulardii offered a reduction clinical styles as no dominant therapeutic strategy has
in risk of AAD development (combined RR 0.31 and 0.37, emerged.34 The pathophysiology of IBS remains unknown, but
respectively).24 Two recent placebo-controlled RCTs evaluated several lines of evidence link symptomatic expression of this
combination probiotic products for the prevention of AAD as disorder with the intestinal microbiota. IBS patients may have
their primary end point. Hickson et al used the probiotic subtle differences in their luminal and mucosal-associated in-
mixture currently marketed as DanActive (Dannon) in the testinal microbiota compared with controls.35,36 New-onset IBS
United States and found that it significantly reduced AAD (12% symptoms can develop in up to a third of individuals after
vs 34%) in an older cohort of hospitalized patients.25 A second recovery from a self-limited episode of infectious gastroenteri-
study evaluated a combination probiotic containing both L casei tis.37,38 Small-bowel bacterial overgrowth has been reported in a
and Lactobacillus acidophilus (Bio-K⫹; Bio-K Plus International, proportion of IBS patients, and antibiotics offer relief of IBS
Laval, Quebec, Canada) in 255 patients. Patients given the symptoms in some individuals.39,40 Although controversy exists,
higher dose of probiotic concurrent with antibiotics (and for 5 bacteria likely contribute to at least some symptoms of IBS.41
days afterward) had fewer occurrences of AAD (15.5% vs Several clinical trials have investigated the potential for pro-
44.1%).26 As a secondary end point, both of these studies also biotics as therapy in IBS. These trials are the subject of several
showed a reduction in development of CDAD (discussed be- single-topic reviews.42– 44 Systematic summarization of these
low). results is complicated by the inclusion of several probiotic
strains/species, single or combination preparations, dosing reg-
C Difficile–Associated Diarrhea imens, and unique study designs. Several studies included end
C difficile–associated diarrhea is a common nosocomial points which were not clinically applicable, or demonstrated
and community-based medical condition. Typically linked to improvement over baseline, but not compared with placebo.42
antibiotic-induced disturbance of the intestinal microbiota, Most studies were short-term only; data on long-term efficacy
CDAD is now increasingly identified in patients without recent are still lacking.
antibiotic exposure.27 Antibiotic therapy with metronidazole, A meta-analysis of 3 RCTs suggests that LGG moderately
oral vancomycin, and now fidaxomicin make up the current improves pain symptoms in children with IBS (NNT ⫽ 4).45
September 2012 PROBIOTICS IN PRACTICE 965

Traditional IBS treatment end points have not been adequately induction of remission and maintenance of remission typically by
met in studies of other single strain lactobacillus species in comparing the probiotic with oral mesalamine or adding the
adults.42 A Bifidobacterium infantis strain (B infantis 35624; Align, probiotic to standard therapy. ECN at 200 mg/d was similar in
Proctor and Gamble, Cincinnati, Ohio) was evaluated in 2 efficacy to 1500 mg of mesalamine for maintaining UC in
clinical trials. One study found significant reductions in pain, remission.58
bloating, bowel movement difficulty, and composite symptom High dose VSL#3 (3.6 trillion colony-forming units per day;
score vs placebo and a lactobacillus species.46 In a larger follow-up Sigma-Tau Pharmaceuticals, Inc, Towson, MA) has shown ther-
study, reduction in pain and global relief of IBS symptoms were apeutic efficacy in 2 RCTs evaluating patients with mild-to-
significantly greater in the B infantis–treated group compared moderately active UC. When offered to UC patients having a
with placebo.47 flare while taking mesalamine or an immunomodulator, the
General recommendations from the American College of probiotic cohort demonstrated improved symptom-based dis-
Gastroenterology as well as expert consensus panels from both ease activity indexes and rectal bleeding, but not endoscopic
the United States and in Europe are similar.32,34,48 There is scores, compared with the placebo group.59 A study conducted
reasonable rationale for why probiotics may work as treatment in India included 144 adults with relapsing UC and showed the
for IBS. There are at least some positive controlled studies VSL#3 group to have significantly higher remission rates (42.9%
showing that probiotic supplementation reduces IBS symptoms vs 15.9%) and endoscopic healing (32% vs 14.7%).60 Most pa-
in some patients. The evidence of benefit is not sufficiently tients in both groups remained taking a stable dose of mesala-
strong to support the general recommendation of probiotics mine therapy. A high dropout rate in both groups (29% VSL#3,
for IBS; however, the benefit appears greatest for bifidobacteria 59% placebo) was a limitation of the latter study. VSL#3 was
species and certain combinations of probiotics which include also shown to improve rates of induction and maintenance of
bifidobacteria species rather than single species lactobacillus pro- remission in children with UC (n ⫽ 29 total).61 Recent Co-
biotics. chrane reviews conclude that there are insufficient data to
With probiotics, patients might experience a global im- demonstrate that probiotics have efficacy in maintaining remis-
provement in symptomatology rather than specific improve- sion in UC; however, they have not recently addressed induction
ment in bowel function. Because treatment options for IBS of remission in UC.62 Single strain Lactobacillus and Bifidobacte-
remain limited in both number and efficacy, a therapeutic rium (infantis 35624, Align, Proctor and Gamble) probiotics did
trial of probiotics is reasonable for patients interested in this not show efficacy for maintaining UC remission in clinical
approach. trials.63,64
In summary, the overall evidence suggests that ECN and
Inflammatory Bowel Diseases VSL#3 have modest efficacy, similar to and perhaps comple-
Evidence points to the intestinal microbiome being a mentary to mesalamine, in inducing and maintaining remission
key player in the development and perpetuation of the inflam- for mild-to-moderately active UC.
matory bowel diseases.49 Defects in the innate immune re-
sponse to commensal intestinal bacteria resulting in an exag- Pouchitis
gerated adaptive immune response to these organisms are Chronic or recurrent pouchitis is an important compli-
implicated in the pathogenesis of CD.50 Several key CD risk cation occurring in approximately 10% to 20% of UC patients
genes have functions related to bacterial killing, and antibiotics after ileal anal pouch formation surgery. VSL#3 (Sigma-Tau
have therapeutic efficacy in CD and pouchitis.51,52 Compared Pharmaceuticals, Inc) was shown beneficial in prophylaxis
with CD, a central role for gut bacteria is less strongly impli- against pouchitis onset after surgical take-down65 and in main-
cated in the pathogenesis of UC.53 However, the evidence sup- taining clinical remission after antibiotic induction.66,67 These
porting probiotics in patient management is better for UC and trials were conducted in Europe and included approximately 20
pouchitis than for CD. patients per group. A practice-based report from the Cleveland
Several limitations exist with trials which have evaluated Clinic found only 19% of patients (6 of 31) who started taking
probiotic therapy in the inflammatory bowel diseases. These VSL#3 after treatment with antibiotics to still be taking the
include small cohort sizes, use of different probiotic doses and probiotic at 8 months.68 A single study from the Netherlands
strains, varied treatment durations, and differences in concur- found that compared with a historical cohort, patients taking
rent conventional treatments. Regardless, patients with inflam- LGG had a delayed onset of pouchitis at 3 years (7% vs 29%).69
matory bowel disease often take or consider taking probiotics Clinical expert-generated guidelines concur that probiot-
and appreciate their clinician having knowledge of the topic. ics (VSL#3) can be effective for preventing recurrence of
pouchitis.32,70,71
Crohn’s Disease
Probiotic use in the management of CD is not sup- Complications of Chronic Liver
ported by currently available RCT data. Trials have found LGG Disease/Hepatic Encephalopathy
and other lactobacilli not superior to placebo as an additive to Luminal microbiota play an important role in the
standard care for inducing or maintaining remission in CD or pathogenesis of both spontaneous bacterial peritonitis and he-
for preventing postoperative relapse.54 –56 There are also no solid patic encephalopathy. Ammonia produced by gut bacteria is
data to support the use of ECN or S boulardii in CD.57 believed to play a key role in hepatic encephalopathy. Antibiot-
ics are employed in clinical practice to reduce severity or fre-
Ulcerative Colitis quency of both these chronic liver disease complications. Lac-
Several published RCTs have shown benefit of probiot- tulose, a mainstay of therapy, is a prebiotic for lactobacilli which
ics in the management of UC. These studies have examined can limit bacterial ureases.
966 MATTHEW A. CIORBA CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 10, No. 9

The role for probiotics in these disorders is an area of tigators in an effort to encompass both beneficial microbes and
ongoing investigation.72 Treatment of hepatic encephalopathy their products.83
with L acidophilus was studied as early as 1965.73 More recently,
Liu and colleagues offered a probiotic and prebiotic mixture to
Conclusions
97 patients with minimal hepatic encephalopathy and observed
a reduction in ammonia levels and improvement in encepha- Evidence supports a role for considering the recommen-
lopathy.74 Another group found that a yogurt-based probiotic dation of conventional probiotics for some clinical conditions.
supplement significantly improved quantitative measurements Probiotic strain selection should focus on quality-tested prod-
of minimal hepatic encephalopathy in patients with nonalco- ucts with clinically demonstrated benefit for the given disorder.
holic cirrhosis.75 The latter group is now completing a more Patients and physicians should expect modest effects and con-
comprehensive trial using the probiotic LGG in a similar pa- sider using probiotics as a supplement to, rather than a replace-
tient cohort.76 ment for, conventional therapy. Though challenges exist, ongo-
Society guidelines and expert consensus panels do not cur- ing investigations offer great promise for the future. Perhaps
rently support a recommendation of probiotic use for any one day clinicians will have the opportunity to use directed
chronic liver disease–associated condition. selection of a probiotic or probiotic-derived product to specif-
ically address a patient’s unique disease-causing physiologic or
genetic defect.
Safety of Probiotics
For most populations, probiotic consumption is con-
sidered safe and complications rare. A review on the safety of
Supplementary Material
probiotics by Snydman points out that although case reports of Note: To access the supplementary material accompa-
bacteremia and endocarditis (LGG) as well as cases of fungemia nying this article, visit the online version of Clinical Gastroenter-
(S boulardii) exist, epidemiologic evidence suggests that there is ology and Hepatology at www.cghjournal.org, and at http://dx.
no overall increase in population risk based on usage data.77 doi.org/10.1016/j.cgh.2012.03.024.
This position is substantiated by a recent US government com-
missioned review panel report.78 As a caveat, however, a high References
profile multicenter placebo controlled Dutch RCT examining 1. Williams MD, Ha CY, Ciorba MA. Probiotics as therapy in gastro-
probiotic supplementation in severe acute pancreatitis found a enterology: a study of physician opinions and recommendations.
higher incidence of mesenteric ischemia and death in the treat- J Clin Gastroenterol 2010;44:631– 636.
ment group.79 This is the only trial to date to infer such a 2. Kau AL, Ahern PP, Griffin NW, et al. Human nutrition, the gut
relationship, but supports the concept that probiotics should microbiome and the immune system. Nature 2011;474:327–
be avoided in critically ill patients. Indwelling central vein 336.
3. Macpherson AJ, Harris NL. Interactions between commensal in-
catheters and perhaps cardiac valvular disease may be relative
testinal bacteria and the immune system. Nat Rev Immunol
contraindications.80
2004;4:478 – 485.
4. Sekirov I, Russell SL, Antunes LC, et al. Gut microbiota in health
What Lies Ahead for Probiotics in and disease. Physiol Rev 2010;90:859 –904.
Digestive Diseases 5. Ng SC, Hart AL, Kamm MA, et al. Mechanisms of action of
probiotics: recent advances. Inflamm Bowel Dis 2009;15:300 –
Inspired investigators and technical advances in genom- 310.
ics are facilitating in-depth scientific investigation of the hu- 6. Madsen KL. Interactions between microbes and the gut epithe-
man microbiome and the functional capacities of probiotics. lium. J Clin Gastroenterol 2011;45(Suppl):S111–S114.
These advances are sure to bring paradigmatic changes to our 7. Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus
fundamental understanding of how microbiota influence modulates intestinal pain and induces opioid and cannabinoid
health and how they can be manipulated to combat disease and receptors. Nat Med 2007;13:35–37.
improve quality of life. Future indications and therapeutic 8. Ciorba MA, Riehl TE, Rao MS, et al. Lactobacillus probiotic pro-
directions for probiotics may include conditions as diverse as tects intestinal epithelium from radiation injury in a TLR-2/cyclo-
oxygenase-2-dependent manner. Gut 2012;61:829 – 838.
mood disorders, obesity, autism, and diabetes. Recent clinical
9. Yan F, Cao H, Cover TL, et al. Colon-specific delivery of a probiotic-
trials and translational studies suggest that lactobacillus pro- derived soluble protein ameliorates intestinal inflammation in
biotics may offer epithelial cytoprotection to limit symptoms of mice through an EGFR-dependent mechanism. J Clin Invest 2011;
radiation enteritis, a dose-limiting side effect for patients re- 121:2242–2253.
ceiving abdominal radiation therapy for malignancy.8,81,82 10. Shanahan F. Probiotics in perspective. Gastroenterology 2010;
Promise is held for confirmative testing of helminth-based 139:1808 –1812.
therapy and “turbo-probiotics” designed to secrete human cy- 11. Mileti E, Matteoli G, Iliev ID, et al. Comparison of the immuno-
tokines. Gene-based bacterial profiling studies from disease modulatory properties of three probiotic strains of lactobacilli
affected humans have identified what may be novel “probiotics” using complex culture systems: prediction for in vivo efficacy.
such as Faecalibacterium prausnitizii and Clostridium species IV PLoS One 2009;4:e7056.
12. Berman S, Spicer D. Safety and reliability of lactobacillus sup-
and XIVa. Finally, the identification, purification, and repack-
plements in Seattle, Washington. Internet J Altern Med 2003;
aging of probiotic-derived soluble factors possessing proven 1(2).
capacity to modify biological function may allow us to harness 13. Sanders ME. Impact of probiotics on colonizing microbiota of the
the power of probiotics while averting the potential risks asso- gut. J Clin Gastroenterol 2011;45(Suppl):S115–S119.
ciated with live bacteria. As these advances progress to the 14. McNulty NP, Yatsunenko T, Hsiao A, et al. The impact of a
clinic, the term pharmabiotic has been proposed by some inves- consortium of fermented milk strains on the gut microbiome of
September 2012 PROBIOTICS IN PRACTICE 967

gnotobiotic mice and monozygotic twins. Sci Transl Med Bowel Syndrome, Brandt LJ, Chey WD, et al. An evidence-based
2011;3;106ra106. position statement on the management of irritable bowel
15. Thomas DW, Greer FR, American Academy of Pediatrics Commit- syndrome. Am J Gastroenterol 2009;104(Suppl 1):S1–35.
tee on Nutrition, et al. Probiotics and prebiotics in pediatrics. 35. Carroll IM, Ringel-Kulka T, Keku TO, et al. Molecular analysis of
Pediatrics 2010;126:1217–1231. the luminal- and mucosal-associated intestinal microbiota in di-
16. Guandalini S. Probiotics for prevention and treatment of diarrhea. arrhea-predominant irritable bowel syndrome. Am J Physiol Gas-
J Clin Gastroenterol 2011;45(Suppl):S149 –S153. trointest Liver Physiol 2011;301:G799 –G807.
17. Weizman Z, Asli G, Alsheikh A. Effect of a probiotic infant formula 36. Kassinen A, Krogius-Kurikka L, Mäkivuokko H, et al. The fecal
on infections in child care centers: comparison of two probiotic microbiota of irritable bowel syndrome patients differs signifi-
agents. Pediatrics 2005;115:5–9. cantly from that of healthy subjects. Gastroenterology 2007;133:
18. Pedone CA, Arnaud CC, Postaire ER, et al. Multicentric study of 24 –33.
the effect of milk fermented by Lactobacillus casei on the inci- 37. Marshall JK, Thabane M, Garg AX, et al. Incidence and epidemi-
dence of diarrhoea. Int J Clin Pract 2000;54:568 –571. ology of irritable bowel syndrome after a large waterborne out-
19. Szajewska H, Kotowska M, Mrukowicz JZ, et al. Efficacy of Lac- break of bacterial dysentery. Gastroenterology 2006;131:445–
tobacillus GG in prevention of nosocomial diarrhea in infants. 450.
J Pediatr 2001;138:361–365. 38. Spiller R, Garsed K. Postinfectious irritable bowel syndrome.
20. Szymański H, Pejcz J, Jawień M, et al. Treatment of acute infectious Gastroenterology 2009;136:1979 –1988.
diarrhoea in infants and children with a mixture of three Lactobacil- 39. Ford AC, Spiegel BM, Talley NJ, et al. Small intestinal bacterial
lus rhamnosus strains–a randomized, double-blind, placebo-con- overgrowth in irritable bowel syndrome: systematic review and
trolled trial. Aliment Pharmacol Ther 2006;23:247–253. meta-analysis. Clin Gastroenterol Hepatol 2009;7:1279 –
21. Allen SJ, Martinez EG, Gregorio GV, et al. Probiotics for treating 1286.
acute infectious diarrhoea. Cochrane Database Syst Rev 2010; 40. Pimentel M, Lembo A, Chey WD, et al. Rifaximin therapy for
CD003048. patients with irritable bowel syndrome without constipation.
22. Johnston BC, Goldenberg JZ, Vandvik PO, et al. Probiotics for the N Engl J Med 2011;364:22–32.
prevention of pediatric antibiotic-associated diarrhea. Cochrane 41. Spiegel BM. Questioning the bacterial overgrowth hypothesis of
Database Syst Rev 2011;CD004827. irritable bowel syndrome: an epidemiologic and evolutionary per-
23. Szajewska H, Ruszczyński M, Radzikowski A. Probiotics in the spective. Clin Gastroenterol Hepatol 2011;9:461– 469.
prevention of antibiotic-associated diarrhea in children: a meta-
42. Ringel Y, Ringel-Kulka T. The rationale and clinical effectiveness
analysis of randomized controlled trials. J Pediatr 2006;149:
of probiotics in irritable bowel syndrome. J Clin Gastroenterol
367–372.
2011;45(Suppl):S145–S148.
24. McFarland LV. Meta-analysis of probiotics for the prevention of
43. Moayyedi P, Ford AC, Talley NJ, et al. The efficacy of probiotics in
antibiotic associated diarrhea and the treatment of Clostridium
the treatment of irritable bowel syndrome: a systematic review.
difficile disease. Am J Gastroenterol 2006;101:812– 822.
Gut 2010;59:325–332.
25. Hickson M, D’Souza AL, Muthu N, et al. Use of probiotic lacto-
44. Brenner DM, Moeller MJ, Chey WD, et al. The utility of probiotics
bacillus preparation to prevent diarrhoea associated with antibi-
in the treatment of irritable bowel syndrome: a systematic review.
otics: randomised double blind placebo controlled trial. BMJ
Am J Gastroenterol 2009;104:1033–1049.
2007;335:80.
45. Horvath A, Dziechciarz P, Szajewska H. Meta-analysis: Lactoba-
26. Gao XW, Mubasher M, Fang CY, et al. Dose-response efficacy of
cillus rhamnosus GG for abdominal pain-related functional gas-
a proprietary probiotic formula of Lactobacillus acidophilus
trointestinal disorders in childhood. Aliment Pharmacol Ther
CL1285 and Lactobacillus casei LBC80R for antibiotic-associ-
2011;33:1302–1310.
ated diarrhea and Clostridium difficile-associated diarrhea pro-
phylaxis in adult patients. Am J Gastroenterol 2010;105:1636 – 46. O’Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and Bifido-
1641. bacterium in irritable bowel syndrome: symptom responses and
27. Kelly CP, LaMont JT. Clostridium difficile–more difficult than ever. relationship to cytokine profiles. Gastroenterology 2005;128:
N Engl J Med 2008;359:1932–1940. 541–551.
28. Lo Vecchio A, Zacur GM. Clostridium difficile infection: an update 47. Whorwell PJ, Altringer L, Morel J, et al. Efficacy of an encapsu-
on epidemiology, risk factors, and therapeutic options. Curr Opin lated probiotic Bifidobacterium infantis 35624 in women with
Gastroenterol 2012;28:1–9. irritable bowel syndrome. Am J Gastroenterol 2006;101:1581–
29. McFarland LV, Surawicz CM, Greenberg RN, et al. A randomized 1590.
placebo-controlled trial of Saccharomyces boulardii in combina- 48. Guarner F, Khan AG, Garisch J, et al. World Gastroenterology
tion with standard antibiotics for Clostridium difficile disease. Organisation Practice Guideline—Probiotics and Prebiotics Octo-
JAMA 1994;271:1913–1918. ber 2011. Available at: http://www.worldgastroenterology.org/
30. Surawicz CM, McFarland LV, Greenberg RN, et al. The search for probiotics-prebiotics.html. Accessed January 1, 2012.
a better treatment for recurrent Clostridium difficile disease: use 49. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med
of high-dose vancomycin combined with Saccharomyces boular- 2009;361:2066 –2078.
dii. Clin Infect Dis 2000;31:1012–1017. 50. Sartor RB. Genetics and environmental interactions shape the
31. Na X, Kelly C. Probiotics in Clostridium difficile infection. J Clin intestinal microbiome to promote inflammatory bowel disease
Gastroenterol 2011;45(Suppl):S154 –S158. versus mucosal homeostasis. Gastroenterology 2010;139:
32. Floch MH, Walker WA, Madsen K, et al. Recommendations for 1816 –1819.
probiotic use-2011 update. J Clin Gastroenterol 2011;45(Suppl): 51. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of in-
S168 –S171. flammatory bowel disease. Nature 2011;474:307–317.
33. Cohen SH, Gerding DN, Johnson S, et al. Clinical practice guide- 52. Sartor RB. Therapeutic manipulation of the enteric microflora in
lines for Clostridium difficile infection in adults: 2010 update by inflammatory bowel diseases: antibiotics, probiotics, and prebi-
the society for healthcare epidemiology of America (SHEA) and otics. Gastroenterology 2004;126:1620 –1633.
the infectious diseases society of America (IDSA). Infect Control 53. Danese S, Fiocchi C. Ulcerative colitis. N Engl J Med 2011;365:
Hosp Epidemiol 2010;31:431– 455. 1713–1725.
34. American College of Gastroenterology Task Force on Irritable 54. Schultz M, Timmer A, Herfarth HH, et al. Lactobacillus GG in
968 MATTHEW A. CIORBA CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 10, No. 9

inducing and maintaining remission of Crohn’s disease. BMC 72. Lata J, Jurankova J, Kopacova M, et al. Probiotics in hepatology.
Gastroenterol 2004;4:5. World J Gastroenterol 2011;17:2890 –2896.
55. Bousvaros A, Guandalini S, Baldassano RN, et al. A randomized, 73. Macbeth WA, Kass EH, McDermott WV Jr. Treatment of hepatic
double-blind trial of Lactobacillus GG versus placebo in addition encephalopathy by alteration of intestinal flora with lactobacillus
to standard maintenance therapy for children with Crohn’s dis- acidophilus. Lancet 1965;1:399 – 403.
ease. Inflamm Bowel Dis 2005;11:833– 839. 74. Liu Q, Duan ZP, Ha DK, et al. Synbiotic modulation of gut flora:
56. Prantera C, Scribano ML, Falasco G, et al. Ineffectiveness of effect on minimal hepatic encephalopathy in patients with cirrho-
probiotics in preventing recurrence after curative resection for sis. Hepatology 2004;39:1441–1449.
Crohn’s disease: a randomised controlled trial with Lactobacillus 75. Bajaj JS, Saeian K, Christensen KM, et al. Probiotic yogurt for the
GG. Gut 2002;51:405– 409. treatment of minimal hepatic encephalopathy. Am J Gastroen-
57. Meijer BJ, Dieleman LA. Probiotics in the treatment of human terol 2008;103:1707–1715.
inflammatory bowel diseases: update 2011. J Clin Gastroenterol 76. Probiotic Lactobacillus GG (LGG) in Patients With Minimal He-
2011;45(Suppl):S139 –S144. patic Encephalopathy. Available at: http://clinicaltrials.gov/
58. Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of ulcer- ct2/show/NCT00992290. Accessed December 11, 2011.
ative colitis with the probiotic Escherichia coli Nissle 1917 is as 77. Snydman DR. The safety of probiotics. Clin Infect Dis 2008;
effective as with standard mesalazine. Gut 2004;53:1617–1623. 46(Suppl 2):S104 –S111; Discussion:S144 –S151.
59. Tursi A, Brandimarte G, Papa A, et al. Treatment of relapsing 78. Hempel S, Newberry S, Ruelaz A, et al. Safety of Probiotics to
mild-to-moderate ulcerative colitis with the probiotic VSL#3 as Reduce Risk and Prevent or Treat Disease. US Department of
adjunctive to a standard pharmaceutical treatment: a double- Health and Human Services, 2011. Available at: http://www.ahrq.
blind, randomized, placebo-controlled study. Am J Gastroenterol gov/clinic/tp/probiotictp.htm#Report.
2010;105:2218 –2227. 79. Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic pro-
60. Sood A, Midha V, Makharia GK, et al. The probiotic preparation, phylaxis in predicted severe acute pancreatitis: a randomised, dou-
VSL#3 induces remission in patients with mild-to-moderately ble-blind, placebo-controlled trial. Lancet 2008;371:651– 659.
active ulcerative colitis. Clin Gastroenterol Hepatol 2009:7; 80. Boyle RJ, Robins-Browne RM, Tang ML. Probiotic use in clinical
1202–1209, 1209.e1201. practice: what are the risks? Am J Clin Nutr 2006;83:1256 –
61. Miele E, Pascarella F, Giannetti E, et al. Effect of a probiotic 1264; Quiz:1446 –1257.
preparation (VSL#3) on induction and maintenance of remission 81. Ciorba MA, Stenson WF. Probiotic therapy in radiation-induced
in children with ulcerative colitis. Am J Gastroenterol 2009;104: intestinal injury and repair. Ann N Y Acad Sci 2009;1165:190 –
437– 443. 194.
62. Naidoo K, Gordon M, Fagbemi AO, et al. Probiotics for mainte- 82. Packey CD, Ciorba MA. Microbial influences on the small intesti-
nance of remission in ulcerative colitis. Cochrane Database Syst nal response to radiation injury. Curr Opin Gastroenterol 2010;
Rev 2011;CD007443. 26:88 –94.
63. Fujimori S, Gudis K, Mitsui K, et al. A randomized controlled trial 83. Shanahan F, Collins SM. Pharmabiotic manipulation of the mi-
on the efficacy of synbiotic versus probiotic or prebiotic treatment crobiota in gastrointestinal disorders, from rationale to reality.
to improve the quality of life in patients with ulcerative colitis. Gastroenterol Clin North Am 2010;39:721–726.
Nutrition 2009;25:520 –525. 84. Mowat C, Cole A, Windsor A, et al. Guidelines for the manage-
64. Shanahan F, Guaraner F, von Wright A, et al. A one year, ment of inflammatory bowel disease in adults. Gut 2011;60:
double-blind, placebo controlled trial of a Lactobacillus or a 571– 607.
Bidfidobacterium probiotic for maintenance of steroid-induced 85. Centre for Evidence Based Medicine. Levels of Evidence, 2009.
remission of ulcerative colitis. Gastroenterology 2006;130:A- Available at: http://www.cebm.net/index.aspx?o⫽1025. Ac-
44, 249. cessed Feb 1, 2012.
65. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis
onset with probiotic therapy: a double-blind, placebo-controlled
trial. Gastroenterology 2003;124:1202–1209.
66. Mimura T, Rizzello F, Helwig U, et al. Once daily high dose
probiotic therapy (VSL#3) for maintaining remission in recurrent Reprint requests
or refractory pouchitis. Gut 2004;53:108 –114. Address requests for reprints to: Matthew A. Ciorba, MD, Depart-
67. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as ment of Medicine, Division of Gastroenterology, Washington Univer-
maintenance treatment in patients with chronic pouchitis: a dou- sity, St Louis School of Medicine, 660 South Euclid Avenue, Box 8124,
ble-blind, placebo-controlled trial. Gastroenterology 2000;119: St Louis, Missouri 63110. e-mail: mciorba@dom.wustl.edu; fax: (314)
305–309. 362-8959.
68. Shen B, Brzezinski A, Fazio VW, et al. Maintenance therapy with
a probiotic in antibiotic-dependent pouchitis: experience in clini- Acknowledgments
cal practice. Aliment Pharmacol Ther 2005;22:721–728. The author thanks colleagues at Washington University for their
69. Gosselink MP, Schouten WR, van Lieshout LM, et al. Delay of the critical assessment of this review, and for the knowledge shared by
first onset of pouchitis by oral intake of the probiotic strain participants of FASEB’s Probiotics, Intestinal Microbiota and the Host:
Lactobacillus rhamnosus GG. Dis Colon Rectum 2004;47:876 – Physiological and Clinical Implications 2011 Summer Research Con-
884. ference.
70. Pardi DS, D’Haens G, Shen B, et al. Clinical guidelines for the
management of pouchitis. Inflamm Bowel Dis 2009;15:1424 – Conflicts of interest
1431. The author discloses the following: M.A.C. has received grant sup-
71. Holubar SD, Cima RR, Sandborn WJ, et al. Treatment and pre- port for laboratory research from VSL Pharmaceuticals, Inc.
vention of pouchitis after ileal pouch-anal anastomosis for
chronic ulcerative colitis. Cochrane Database Syst Rev 2010; Funding
CD001176. This work is partially supported by NIH grant DK089016.

You might also like