12
Copyright 2016 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited. Probiotics for the Prevention of Antibiotic-Associated Diarrhea in Children Hania Szajewska, yz Roberto Berni Canani, y Alfredo Guarino, § Iva Hojsak, jj Flavia Indrio, § Sanja Kolacek, ô Rok Orel, # Raanan Shamir, Yvan Vandenplas, yy Johannes B. van Goudoever, and zz Zvi Weizman, on Behalf of the ESPGHAN Working Group for Probiotics/Prebiotics ABSTRACT This article provides recommendations, developed by the Working Group (WG) on Probiotics of the European Society for Pediatric Gastro- enterology, Hepatology, and Nutrition, for the use of probiotics for the prevention of antibiotic-associated diarrhea (AAD) in children based on a systematic review of previously completed systematic reviews and of randomized controlled trials published subsequently to these reviews. The use of probiotics for the treatment of AAD is not covered. The recommendations were formulated only if at least 2 randomized controlled trials that used a given probiotic (with strain specification) were available. The quality of evidence (QoE) was assessed using the Grading of Recommendations Assessment, Development, and Evaluation guidelines. If the use of probiotics for preventing AAD is considered because of the existence of risk factors such as class of antibiotic(s), duration of antibiotic treatment, age, need for hospitalization, comorbidities, or previous episodes of AAD diarrhea, the WG recommends using Lactobacillus rhamnosus GG (moderate QoE, strong recommendation) or Saccharomyces boulardii (mod- erate QoE, strong recommendation). If the use of probiotics for preventing Clostridium difficile-associated diarrhea is considered, the WG suggests using S boulardii (low QoE, conditional recommendation). Other strains or combinations of strains have been tested, but sufficient evidence is still lacking. Received December 7, 2015; accepted December 10, 2015. From the Medical University of Warsaw, Department of Paediatrics, Warsaw, Poland, the y Department of Translational Medical Science, the z European Laboratory for The Investigation of Food Induced Diseases and CEINGE Advanced Biotechnology, University of Naples ‘‘Federico II,’’ Italy, the § Department of Paediatrics, Children’s Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia, the jj Department of Pediatrics, University Hospital Policlinico, University of Bari, Bari, Italy, the ô Department of Gastroenterology, Hepatology and Nutrition, Children’s Hospital, University Medical Centre, Ljubljana, Slovenia, the # Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel, the Department of Pediatrics, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium, the yy Department of Pediatrics, Emma Children’s Hospital-AMC and VU Universit Medical Center, Amsterdam, The Netherlands, and the zz Pediatric Gastroenterology and Nutrition Unit, Soroka Medical Center. Ben-Gurion University, Beer-Sheva, Israel. Address correspondence and reprint requests to Hania Szajewska, MD, Department of Paediatrics, Medical University of Warsaw, 02–091 Warsaw, Zwirki i Wigury 63A, Poland (e-mail: [email protected]). Supplemental digital content is available for this article. Direct URL citations appear in the printed text, and links to the digital files are provided in the HTML text of this article on the journal’s Web site (www.jpgn.org). H.S. has received research support (study products only) from Biogaia and Dicofarm; has been a clinical investigator for Arla Foods (ongoing), Danone, Nestle; has participated as a speaker on probiotics/microbiota-related subjects for Arla, Biogaia, Biocodex, Danone, Dicofarm, Hipp, Nestle, Nestle Nutrition Institute, Nutricia, Mead Johnson, Merck, Sequoia, and Yakult; has been a member of Nestle Nutrition Institute faculty; and has served as an advisory board member for Fundacja Nutricia (Nutricia Foundation), funding research grants in the field of human nutrition. R.B.C. has participated as a clinical investigator, and/or speaker for Dicofarm, Heinz, Mead Johnson Nutrition, Menarini, Nutricia, and Wyeth (none related to the work submitted). A.G. was supported by Biocodex, Dicofarm, and Mead Johnson Nutrition. Others (such as personal fees for advisory boards, consultancy; personal speakers fee related to any of the products mentioned in the article and companies using/or selling them, as well as competitors)none declared. Other industry relations not related to the content of the article: Astellas Pharma. I.H. has participated as a clinical investigator for Biogaia and Chr Hansen. F.I. has participated as a clinical investigator, consultant, and speaker for Arla Food, Biogaia, Noos, Nestle, and Nestle Nutrition Institute. S.K. received personal speaker fee for topics within the probiotic-related fields (never for probiotic product) from Biogaia, Medis, and Arla Foods. Clinical investigator without any personal fee or fee provided to the institution in the studies related to probiotic product of Dukat. Scientific grants delivered entirely and only to the Institution for probiotic- related clinical studies received from Chr. Hansen and Biogaia. Other activities nonrelated to probiotics were as follows: personal speaker fee received from Abbott, Danone, Nestle, Nutricia and MSD, and nonrestricted educational grants delivered to the Institution from Nestle, Nutricia, Podravka, AbbVie, Falk, Merck/MSD, Hospira and Pharmas. R.O. has participated as a clinical investigator for United Pharmaceuticals (probiotics were not involved) and for BioGaia (ongoing; investigator’s initiated study); has participated as a speaker for Medis, Nutricia, Ewopharma, Biogaia, United Pharmaceuticals, Danone, Abbvie, and MSD. R.S. has a clinical investigator, and/or advisory board member, and/or consultant, and/or speaker for Abbott, Danone Institute International, Danone, Enzymotec, Nestle, Nestle Nutrition Institute, and Nutricia; none related to the work submitted. Y.V. is consultant or member of advisory board for ASPEN, Ausnutria, Biocodex, Danone Belgium, and United Pharmaceuticals; has been a clinical investigator and speaker (sometimes probiotic related, sometimes not) for Abbott Nutrition, ARLA foods, ASPEN, Biogaia, Biocodex, Danone, Dumex, Hero, Hipp, Nestle Nutrition Institute, Nutricia, Mead Johnson, Merck, Menarini, Orafti, Pfizer, Phacobel, Sari Husada, Shire (Movetis), Sucampo, Takeda, United Pharmaceuticals, Wyeth and Yakult (grants, study, and advisory fees always paid to the institution). J.B.G. is member of the Dutch National Health Council, the Breastfeeding Council and founder and director of the Dutch Human Milk Bank; his institute received research grants from Danone and MJN; he received compensation for lectures by Danone, Nutricia, Nestle Nutrition Institute; he is consultant for Nutricia Nederland and holds 3 patents on the amino acid composition of infant formula. Z.W. has participated as a clinical investigator, speaker, and consultant for Biocodex and Biogaia. Copyright # 2016 by European Society for Pediatric Gastroenterology, Hepatology, and Nutrition and North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition DOI: 10.1097/MPG.0000000000001081 CLINICAL GUIDELINE JPGN Volume 62, Number 3, March 2016 495

Probiotics for the Prevention of Antibiotic-Associated ... · Diarrhea in Children Hania ... European Society for Pediatric Gastro-enterology, Hepatology, and Nutrition, for the use

  • Upload
    vothuy

  • View
    223

  • Download
    2

Embed Size (px)

Citation preview

Co

CLINICAL GUIDELINE

Probiotics for the Prevention of Antibiotic-Associated

Diarrhea in Children�Hania Szajewska, yzRoberto Berni Canani, yAlfredo Guarino, §Iva Hojsak, jjFlavia Indrio,

§Sanja Kolacek, �Rok Orel, #Raanan Shamir, ��Yvan Vandenplas, yyJohannes B. van Goudoever,

pyright 2016 by

and zzZvi W alf of the ESPGHAN Working Group for Probiotics/Prebiotics

available. The quality of e

Recommendations Assess

Received December 7, 20From the �Medical Univ

Laboratory for The I§Department of PaedUniversity Hospital PUniversity Medical CeIsrael, Sackler FacultBrussels, Belgium, thethe zzPediatric Gastro

Address correspondence aWigury 63A, Poland

Supplemental digital conttext of this article on

H.S. has received researchhas participated as aInstitute, Nutricia, Memember for Fundacjainvestigator, and/or spsupported by Biocoderelated to any of the prnot related to the conteclinical investigator, ctopics within the probifee provided to the insrelated clinical studiesAbbott, Danone, NestlMerck/MSD, HospiraBioGaia (ongoing; invAbbvie, and MSD. RInternational, Danoneadvisory board for ASprobiotic related, somNutricia, Mead JohnsoYakult (grants, study,and founder and direclectures by Danone, Nformula. Z.W. has par

Copyright # 2016 byGastroenterology, He

DOI: 10.1097/MPG.0000

JPGN � Volume 62, N

eizman, on Beh

combinations of strains have been tested, but sufficient evidence is still

lacking.

ABSTRACT

This article provides recommendations, developed by the Working

Group (WG) on Probiotics of the European Society for Pediatric Gastro-

enterology, Hepatology, and Nutrition, for the use of probiotics for the

prevention of antibiotic-associated diarrhea (AAD) in children based on a

systematic review of previously completed systematic reviews and of

randomized controlled trials published subsequently to these reviews.

The use of probiotics for the treatment of AAD is not covered.

The recommendations were formulated only if at least 2 randomized

controlled trials that used a given probiotic (with strain specification) were

ESPGHAN and NASPGHAN. Un

vidence (QoE) was assessed using the Grading of

ment, Development, and Evaluation guidelines. If

15; accepted December 10, 2015.ersity of Warsaw, Department of Paediatrics, Warsaw,nvestigation of Food Induced Diseases and CEINGEiatrics, Children’s Hospital Zagreb, University of Zagoliclinico, University of Bari, Bari, Italy, the �Departntre, Ljubljana, Slovenia, the #Institute of Gastroentero

y of Medicine, Tel-Aviv University, Tel-Aviv, Israel,yyDepartment of Pediatrics, Emma Children’s Hospitaenterology and Nutrition Unit, Soroka Medical Centend reprint requests to Hania Szajewska, MD, Departme

(e-mail: [email protected]).ent is available for this article. Direct URL citations appthe journal’s Web site (www.jpgn.org).support (study products only) from Biogaia and Dicofa

speaker on probiotics/microbiota-related subjects for Aad Johnson, Merck, Sequoia, and Yakult; has been a mem

Nutricia (Nutricia Foundation), funding research graeaker for Dicofarm, Heinz, Mead Johnson Nutrition, Mx, Dicofarm, and Mead Johnson Nutrition. Others (sucoducts mentioned in the article and companies using/or snt of the article: Astellas Pharma. I.H. has participated aonsultant, and speaker for Arla Food, Biogaia, Noos, Notic-related fields (never for probiotic product) from Biotitution in the studies related to probiotic product of Dukreceived from Chr. Hansen and Biogaia. Other activitiee, Nutricia and MSD, and nonrestricted educational granand Pharmas. R.O. has participated as a clinical inve

estigator’s initiated study); has participated as a speaker.S. has a clinical investigator, and/or advisory board, Enzymotec, Nestle, Nestle Nutrition Institute, and NuPEN, Ausnutria, Biocodex, Danone Belgium, and Unitetimes not) for Abbott Nutrition, ARLA foods, ASPENn, Merck, Menarini, Orafti, Pfizer, Phacobel, Sari Husa

and advisory fees always paid to the institution). J.B.G.tor of the Dutch Human Milk Bank; his institute receutricia, Nestle Nutrition Institute; he is consultant for Nuticipated as a clinical investigator, speaker, and consuEuropean Society for Pediatric Gastroenterology,patology, and Nutrition000000001081

umber 3, March 2016

the use of probiotics for preventing AAD is considered because of the

existence of risk factors such as class of antibiotic(s), duration of antibiotic

treatment, age, need for hospitalization, comorbidities, or previous episodes

of AAD diarrhea, the WG recommends using Lactobacillus rhamnosus GG

(moderate QoE, strong recommendation) or Saccharomyces boulardii (mod-

erate QoE, strong recommendation). If the use of probiotics for preventing

Clostridium difficile-associated diarrhea is considered, the WG suggests

using S boulardii (low QoE, conditional recommendation). Other strains or

authorized reproduction of this article is prohibited.

Poland, the yDepartment of Translational Medical Science, the zEuropeanAdvanced Biotechnology, University of Naples ‘‘Federico II,’’ Italy, thereb School of Medicine, Zagreb, Croatia, the jjDepartment of Pediatrics,ment of Gastroenterology, Hepatology and Nutrition, Children’s Hospital,logy, Nutrition and Liver Diseases, Schneider Children’s Medical Center ofthe ��Department of Pediatrics, UZ Brussel, Vrije Universiteit Brussel,

l-AMC and VU Universit Medical Center, Amsterdam, The Netherlands, andr. Ben-Gurion University, Beer-Sheva, Israel.nt of Paediatrics, Medical University of Warsaw, 02–091 Warsaw, Zwirki i

ear in the printed text, and links to the digital files are provided in the HTML

rm; has been a clinical investigator for Arla Foods (ongoing), Danone, Nestle;rla, Biogaia, Biocodex, Danone, Dicofarm, Hipp, Nestle, Nestle Nutritionber of Nestle Nutrition Institute faculty; and has served as an advisory board

nts in the field of human nutrition. R.B.C. has participated as a clinicalenarini, Nutricia, and Wyeth (none related to the work submitted). A.G. wash as personal fees for advisory boards, consultancy; personal speakers feeelling them, as well as competitors)—none declared. Other industry relationss a clinical investigator for Biogaia and Chr Hansen. F.I. has participated as aestle, and Nestle Nutrition Institute. S.K. received personal speaker fee for

gaia, Medis, and Arla Foods. Clinical investigator without any personal fee orat. Scientific grants delivered entirely and only to the Institution for probiotic-s nonrelated to probiotics were as follows: personal speaker fee received fromts delivered to the Institution from Nestle, Nutricia, Podravka, AbbVie, Falk,

stigator for United Pharmaceuticals (probiotics were not involved) and forfor Medis, Nutricia, Ewopharma, Biogaia, United Pharmaceuticals, Danone,member, and/or consultant, and/or speaker for Abbott, Danone Institutetricia; none related to the work submitted. Y.V. is consultant or member ofed Pharmaceuticals; has been a clinical investigator and speaker (sometimes, Biogaia, Biocodex, Danone, Dumex, Hero, Hipp, Nestle Nutrition Institute,da, Shire (Movetis), Sucampo, Takeda, United Pharmaceuticals, Wyeth andis member of the Dutch National Health Council, the Breastfeeding Councilived research grants from Danone and MJN; he received compensation fortricia Nederland and holds 3 patents on the amino acid composition of infantltant for Biocodex and Biogaia.Hepatology, and Nutrition and North American Society for Pediatric

495

Co

Key Words: Clostridium difficile, dysbiosis, guideline, infants, microbiota,

probiotics, RCT, systematic review

(JPGN 2016;62: 495–506)

Szajewska et al

A ntibiotic-associated diarrhea (AAD) is a common and chal-lenging complication observed in the ambulatory and hospi-

tal settings alike that occurs in up to a third of all patients treatedwith antibiotics (1). It is defined as diarrhea that occurs in relation toantibiotic treatment with the exclusion of other etiologies. Thisrelation does not necessarily translate into an immediate adversereaction to antibiotics, because AAD may occur after a few weeksand even up to a few months after the administration of theantibiotics (2). Thus, in the latter situation, caution is needed todifferentiate AAD from an episode of infectious gastroenteritis. Therisk of AAD is higher when there is a use of aminopenicillinswithout/with clavulanate, cephalosporins, clindamycin, and, ingeneral, any antibiotic that is active against anaerobes (3). Almostany oral and intravenous antibiotic treatment can, however, causeAAD (3). Clinically, AAD may present as mild diarrhea, but itcan present as well as fulminant pseudomembranous colitis.Usually, no pathogen is identified. In the most severe forms andin an increasing number of patients with chronic conditions such asthose with inflammatory bowel diseases, cystic fibrosis, and cancer,however, the causative agent is often identified as Clostridiumdifficile (4).

The use of probiotics, defined as ‘‘live microorganisms that,when administered in adequate amounts, confer a health benefit onthe host,’’ (5) and/or fermented products such as yogurt has beenreported as a measure to prevent the occurrence of AAD. Therationale for the use of these products relies on the hypothesis thatAAD is caused by dysbiosis that is triggered by antibiotic use andthat the probiotic intervention favorably modulates the intestinalmicrobiota (1).

The aim of this position paper by the European Society forPediatric Gastroenterology, Hepatology, and Nutrition (ESP-GHAN) Working Group (WG) on Probiotics and Prebiotics is toprovide recommendations for the use of probiotics for preventingAAD in children.

METHODSThe same methodology that had been used previously by the

WG for developing guidelines on the use of probiotics for themanagement of acute gastroenteritis (6) was applied for developingthe present position paper. In brief, the document provides a reviewof previously completed systematic reviews and of randomizedcontrolled trials (RCTs) published subsequently to these reviews.For systematic reviews/meta-analyses, the Cochrane Database ofSystematic Reviews and the Database of Abstracts of Reviews ofEffects (DARE) were searched. For subsequently published trials(starting from the date of the most recent search in the includedreviews), CENTRAL (Cochrane Central Register of ControlledTrials), MEDLINE, and EMBASE were searched up to July2015 and again in November 2015.

The focus was on 6 taxonomic groups (Lactobacillus,Bifidobacterium, Saccharomyces, Streptococcus, Enterococcus,and/or Bacillus). The list of individual probiotics to be consideredwas established based on the results of the Cochrane reviewevaluating probiotics for preventing AAD in children (7) and thelist of commonly used probiotics developed by the World Gastro-enterology Organization (8).

The WG is aware that taxonomically equivalent probiotic

pyright 2016 by ESPGHAN and NASPGHAN. Un

microorganisms may be supplied by different manufacturers.At least 1 study indicated that the manufacturing process may

496

influence properties of probiotic bacteria (9). At present, whetheror not these manufacturing differences translate into differences invivo, as well as clinical outcomes, however, remains unclear.Consequently, the taxonomically equivalent probiotics are pre-sented jointly, regardless of the manufacturer. The WG alsorealizes that the same brand may have a different compositionin different locations; nevertheless, this position paper deals withstrain(s) rather than brands or commercial names. Finally, depend-ing on the country, the same probiotic microorganism(s) may beavailable as food supplements, available as registered pharmaceu-tical products, and/or incorporated into foods (10). In this docu-ment, the effectiveness of probiotics was analyzed regardless of theregistration status. Health care professionals and consumersshould, however, be aware of possible variations in the manufac-turing and safety profiles of the products, which may be differentwhen the strain is registered as a drug and also with regard to theclaims allowed.

The primary outcome measures were diarrhea/AAD andC difficile-associated diarrhea (all as defined by the investigators).

To assess the methodological quality of the included RCTs(included in the previously published meta-analyses and sub-sequently published RCTs not included in the systematic reviews),the Cochrane Collaboration’s tool for assessing risk of bias wasused. This tool includes the following criteria: adequacy ofsequence generation, allocation concealment, and blinding ofparticipants, personnel and outcome assessors; incomplete outcomedata; and selective reporting (11).

For reporting the effect, the results for individual studiesand pooled statistics are reported as the risk ratio (RR) betweenthe experimental and control groups with 95% confidence intervals(95% CIs). In other circumstances, we report the findings asreported by the authors of the included studies.

When synthesizing the evidence, each section presentsa summary of the evidence followed by the key recommen-dations. The GRADE system, developed by the Grading ofRecommendations Assessment, Development and EvaluationsWorking Group (12), was used to grade the strength of evidenceand grades of recommendations used in these guidelines. In brief,the GRADE system offers 4 categories of the quality of theevidence (ie, high, moderate, low, and very low) and 2 categoriesof the strength of recommendation (ie, strong or conditional[weak]) (Table 1). The GRADE system suggests presentingrecommendations in the active voice (13). Thus, we used thewording ‘‘the WG recommends’’ for strong recommendations,and ‘‘the WG suggests’’ for conditional [weak] recommen-dations.

As in our previous document (6), the WG adopted theposition of the US Food and Drug Administration Guidance forIndustry (14) that at least 2 adequate and well-controlled studies,each convincing on its own, are needed to establish the effec-tiveness of an intervention. Consequently, the recommendationswere formulated only if at least 2 RCTs that used a given probioticwere available. If there was only 1 RCT, regardless of whether ornot it showed a benefit, no recommendation was formulated.Moreover, if the strain specification was not given and/or theprobiotic product was not otherwise identifiable, no recommen-dation was made.

For the sake of completeness, we report the pooled data(meta-analysis) of all probiotic trials. No recommendation onthe use of probiotics in general was, however, made, becausepooling data on different probiotics has been repeatedly ques-tioned (15). Instead, because various probiotic strains differ intheir effects, preference was given to reporting evidence and

JPGN � Volume 62, Number 3, March 2016

authorized reproduction of this article is prohibited.

recommendations related to a specific probiotic strain or theircombinations separately.

www.jpgn.org

Co

of C difficile-associated diarrhea (4 RCTs, n¼ 938, RR 0.34, 95%

QUALITY OF EVIDENCE: Moderate.

TABLE 1. The grades of the quality of evidence and strength of recommendation set by the GRADE Working Group

Quality of evidence High quality We are very confident that the true effect lies close to that of the estimate of the effect.

Moderate quality We are moderately confident in the effect estimate: the true effect is likely to be close to the

estimate of the effect, but there is a possibility that it is substantially different.

Low quality Confidence in the effect estimate is limited: the true effect may be substantially different from

the estimate of the effect.

Very low quality Very little confidence in the effect estimate: the true effect is likely to be substantially different

from the estimate of the effect.

Strength of recommendation Strong When the evidence showed that the benefit of the intervention clearly outweighs the

undesirable effects.

Conditional (weak) When the trade-offs were less certain (either because of the low quality of evidence or because

the evidence suggests that desirable and undesirable effects are closely balanced).

GRADE¼Grading of Recommendations Assessment, Development and Evaluations.

JPGN � Volume 62, Number 3, March 2016 Probiotics for the Prevention of AAD

A draft of the position paper was sent to the WG members forreview and further comments. All of the critical feedback wasdiscussed through e-mail or during personal contacts, and changeswere incorporated as necessary. Recommendations were formulatedand graded. The WG members voted anonymously on each recom-mendation using an online electronic survey tool (SurveyMonkeyInc, Palo Alto, CA, www.surveymonkey.com). Any disagreementfollowing voting was resolved by discussion, and for all recom-mendations, a full consensus was reached. A finalized documentwas submitted to the ESPGHAN Council for final acceptancebefore publication.

The WG recommendations may need to be modified bydifferent countries considering differences in health care systems,local values and preferences, including availability, quality, andcosts of probiotics, and should help local policy makers to decidewhether to use routinely probiotics with documented efficacy forpreventing AAD in children receiving antibiotics based on localcost-effectiveness analysis. This is particularly important in low-and middle-income countries.

Clearly, an individual patient’s risk of developing AAD or Cdifficile-associated diarrhea depends on a number of factors such asclass of antibiotic(s), duration of antibiotic treatment, age, need forhospitalization, comorbidities, and previous episodes of AAD or Cdifficile-associated diarrhea (1–3). These risk factors should beconsidered when making decisions on the use of probiotics inchildren for preventing AAD or C difficile-associated diarrhea.The WG acknowledges that the judicious use of antibiotics remainsthe best method of preventing AAD.

The conclusions of this document may require revision in thefuture as new information becomes available. It is the intention ofthe WG to revise the recommendations not later than 5 years fromnow and produce an updated document.

PROBIOTICS OVERALLA number of systematic reviews and meta-analyses have

shown that probiotics as a group are effective in preventing AAD(7,16,17).

A 2012 meta-analysis by Hempel et al (16) collecteddata from 82 RCTs that evaluated the efficacy of probiotics forpreventing AAD in subjects of all ages. Probiotics, as a group,reduced the risk of AAD (63 RCTs, n¼ 11,811 participants, RR0.58, 95% CI 0.50–0.68). Sixteen RCTs were carried out in infantsand young children and reported a reduced risk of AAD withprobiotic administration (RR 0.55, 95% CI 0.38–0.80). In themajority of trials, Lactobacillus-based interventions, alone or in

pyright 2016 by ESPGHAN and NASPGHAN. Un

combination with other genera, were used. Strains were poorlydocumented. The quality of evidence was low. Of 63 included

www.jpgn.org

trials, 59 lacked adequate information to assess the overall risk ofbias. There was no placebo group in some trials. Included trialsused different definitions of diarrhea/AAD, and in some, nodefinition of these outcomes was provided. Moreover, significantheterogeneity between trials for both primary and secondary out-comes was detected. The authors concluded that the evidence isinsufficient to determine whether this association varies system-atically by population, antibiotic characteristic, or probioticpreparation.

A 2013 systematic review with a meta-analysis assessedthe efficacy and safety of probiotics for preventing C difficile-associated diarrhea or C difficile infection in adults and children(17). A complete case analysis (ie, participants who completedthe study) showed that compared with placebo or no treatment,administration of probiotics reduced the risk of C difficile-associated diarrhea by 64% (23 RCTs, n¼ 4213, RR 0.36, 95%CI 0.26–0.51) in adults and children. In children, probioticadministration reduced the risk of C difficile-associated diarrheafrom 5.9% to 2.3% (3 RCTs, n¼ 605, RR 0.40, 95% CI 0.17–0.96)(17).

For this report, 21 RCTs involving 3255 children wereincluded (18–38). Among them, 11 RCTs were included in 2strain-specific systematic reviews initiated as part of the develop-ment of these guidelines (39,40). One unpublished study (29) wasidentified in the systematic review by Johnston et al (7). Forcharacteristics of the included RCTs, see Table 2, and for amethodological quality summary, see Figure 1. The pooled resultsof 21 RCTs showed that compared with placebo or no intervention,probiotics as a class reduced the risk of AAD by 52% (21.2% vs9.1%, respectively; RR 0.48, 95% CI 0.37–0.61) (Fig. 2). Only 2probiotics were evaluated in >1 RCT. These were Lactobacillusrhamnosus GG (LGG) and Saccharomyces boulardii. Comparedwith placebo, the administration of probiotics also reduced the risk

CI 0.15–0.76) (Fig. 3).

PROBIOTICS WITH RECOMMENDATIONS

L rhamnosus GG (LGG)

RECOMMENDATION. If the use of probiotics forpreventing AAD in children is considered, the WGrecommends using L rhamnosus GG.

authorized reproduction of this article is prohibited.

STRENGTH OF RECOMMENDATION: Strong

497

Copyright 2016 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

TABLE

2.

Ch

ara

cterist

ics

of

the

incl

ud

ed

tria

ls

Ref

eren

ceP

arti

cip

ants

(age

)

Pro

bio

tic

do

se

(CF

U/d

ay)

Du

rati

on

of

inte

rven

tio

nF

oll

ow-u

p

An

tib

ioti

c/s

(ad

min

istr

atio

n)

Defi

nit

ion

of

dia

rrhea

or

AA

DM

anuf

actu

rer

Sp

onso

r

La

cto

ba

cill

us

rha

mno

sus

GG

Arv

ola

etal

(19

)N¼

11

9,

inp

atie

nts

,

(2w

k–

13

y)

2�

10

10

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

ther

apy

3m

oP

enic

illi

n,

Am

ox

icil

lin

,

Cep

halo

spo

rin

s,

Ery

thro

my

cin

,

Tri

met

op

rim

-su

lfa

�3

wat

ery

stoo

ls/d

ay

for

min

imu

mo

f2

con

secu

tiv

ed

ays

No

tre

po

rted

Fin

nis

h

Fo

un

dat

ion

for

Gas

troen

tero

-

log

ical

Res

earc

h

Kin

get

al(2

0)

15

,in

pat

ien

ts

(21

day

s–1

1y

)

30�

10

9F

or

the

du

rati

on

of

anti

bio

tic

ther

apy

Not

men

tioned

Var

ious

�3

loo

sest

ools

in

24

h

No

tre

po

rted

No

tre

port

ed

Sza

jew

ska

etal

(22

)N¼

66

,in

pat

ien

ts,

(mea

nag

e1

2y

;

age

ran

ge

no

t

repo

rted

)

1�

10

9F

or

the

du

rati

on

of

anti

bio

tic

ther

apy

(7d

ays)

6w

kA

mox

icil

lin,

Cla

rith

rom

yci

n

(ora

l)

�3

loo

seo

rw

ater

y

sto

ols

per

day

for

atle

ast

48

h

Dic

ofa

rm,

Rom

e,

Ital

y

Med

ical

Un

iver

sity

of

War

saw

Vai

san

enet

al(2

1)

59

,o

utp

atie

nts

(5m

o–

11

y)

4�

10

8F

or

the

du

rati

on

of

anti

bio

tic

ther

apy

(7d

ays)

No

tm

enti

on

edA

mox

icil

lin

(ora

l)D

efin

edb

yp

aren

tsN

ot

rep

ort

edN

ot

repo

rted

Van

der

ho

ofet

al(1

8)

18

8,

ou

tpat

ien

ts

(6m

o–

10

y)

1–

2�

10

10

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

ther

apy

(10

day

s)

10

day

sA

mox

icil

lin,

amox

icil

lin/

clav

ula

nat

e,

cefp

rozi

l,

clar

ith

rom

yci

n,

oth

er(o

ral)

.

�2

liq

uid

stoo

ls/d

ayC

AG

Fu

ncti

on

al

Fo

od

Gra

nt

fro

mC

AG

Nu

trit

ion

,a

div

isio

no

f

Co

nA

gra

Sa

cch

aro

myc

esb

ou

lard

ii

Bin

etal

(27

)N¼

20

5,

exac

td

ata

no

tg

iven

(22

mo

–1

6y

)

25

0m

g(0

.5�

10

10)�

Fo

rth

ed

ura

tio

n

of

anti

bio

tic

trea

tmen

t

(14

day

s)

No

dat

aA

mox

icil

lin,

clar

ith

rom

yci

n,

met

ron

idaz

ole

Dia

rrh

ea:

incr

ease

in

the

freq

uen

cyo

f

bo

wel

mo

vem

ents

(>3

/day

)o

ra

dec

reas

ein

sto

ol

consi

sten

cy(B

rist

ol

sto

ol

scal

e5

or

6)

No

info

rmat

ion

giv

en

Bio

cod

ex,

Par

is,

Fra

nce

,C

hin

ese

bra

nd

nam

e:

YiH

uo.

S

bo

ula

rdii

CN

CM

I-74

5

Cas

emet

al(2

4)

140,

hosp

ital

ized

and

ou

tpat

ien

ts

(6m

o–

18

y)

50

0m

g(1�

10

10)�

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

trea

tmen

t(S

B

gro

up

7.2

9�

0.9

2

day

s;co

ntr

ol

gro

up

7.5

9�

1.1

7

day

s)

No

dat

aV

ario

us

(ora

lor

intr

aven

ous

)

Dia

rrh

ea:�

3lo

ose

or

wat

ery

stoo

lsp

er

day

for

am

inim

um

of

48

hd

uri

ng

and

/or

up

to2

wk

afte

rth

een

do

f

anti

bio

tic

trea

tmen

t

No

tre

po

rted

No

tre

port

ed

Erd

eve

etal

(25

)N¼

46

6,

exac

td

ata

no

tg

iven

(1–

15

y)

25

0m

g(0

.5�

10

10)�

Ex

act

dat

an

ot

giv

enN

od

ata

Su

lbac

tam

-am

pic

illi

n,

azit

hro

myci

n(n

ot

men

tio

ned

)

Ex

act

defi

nit

ion

no

t

giv

en

No

tre

po

rted

No

tre

port

ed

Szajewska et al JPGN � Volume 62, Number 3, March 2016

498 www.jpgn.org

Copyright 2016 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Ref

eren

ceP

arti

cip

ants

(age

)

Pro

bio

tic

do

se

(CF

U/d

ay)

Du

rati

on

of

inte

rven

tio

nF

oll

ow-u

p

An

tib

ioti

c/s

(ad

min

istr

atio

n)

Defi

nit

ion

of

dia

rrhea

or

AA

DM

anuf

actu

rer

Sp

onso

r

Ko

tow

ska

etal

(23

)N¼

26

9,

inp

atie

nts

and

ou

tpat

ien

ts

(6m

o–

14

y)

50

0m

g(1�

10

10)�

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

trea

tmen

t(S

B

gro

up

7.8�

1

day

s;co

ntr

ol

gro

up

8.1�

1

day

s)

2w

kV

ario

us

(ora

lor

intr

aven

ous

)

Dia

rrh

ea:�

3lo

ose

or

wat

ery

stoo

lsp

er

day

for

am

inim

um

of

48

hd

uri

ngan

d/

or

up

to2

wk

afte

r

the

end

of

anti

bio

tic

trea

tmen

t.A

AD

:A

s

abo

ve,

cau

sed

by

C

dif

fici

leo

rfo

r

oth

erw

ise

un

expl

ain

ed

dia

rrh

ea

No

info

rmat

ion

giv

en(E

nte

rol

Bio

cod

ex—

info

rmat

ion

from

the

auth

ors

)

No

info

rmat

ion

giv

en(M

edic

al

Un

iver

sity

of

War

saw

info

rmat

ion

fro

mth

e

auth

ors

)

Sh

anet

al(2

6)

33

3,

inp

atie

nts

(6m

o–

14

y)

50

0m

g(1�

10

10)�

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

trea

tmen

t(e

xact

dat

an

ot

giv

en)

2w

kV

ario

us

(intr

aven

ous)

Dia

rrhea

:�

3lo

ose

or

wat

ery

stoo

lsp

er

day

or

du

rin

g�

48

h,

occ

urr

ing

du

ring

and

/or

up

to2

wk

afte

rth

een

do

f

anti

bio

tic

trea

tmen

t

On

eo

fth

e

inve

stig

ators

serv

esas

a

con

sult

ant

in

Bio

cod

ex

Bio

flo

r,C

hin

a

Zh

aoet

al(2

8)

24

0(7

–9

y�

2y

)5

00

mg

(1�

10

10)�

Fo

rth

ed

ura

tio

no

f

anti

bio

tic

trea

tmen

t(1

4

day

s)

8w

kA

mox

icil

lin,

clar

ith

rom

yci

n

No

defi

nit

ion

giv

enN

ot

rep

ort

edN

ot

repo

rted

Ba

cill

us

cla

usi

i

Des

tura

etal

(29

)N¼

32

3,

inp

atie

nts

and

ou

tpat

ien

ts

(mea

nag

e4

y)

4�

10

9U

nti

len

do

f

anti

bio

tic

ther

apy

(7–

21

day

s)

Un

til

end

of

anti

bio

tic

ther

apy

(7–

21

day

s)

Pen

icil

lin

s,

ceph

alo

spo

rin

,

coam

ox

ycl

av/

ampi

cill

in-

sulb

acta

m,

and

oth

ers

Ch

ang

ein

bo

wel

hab

its

wit

hth

e

pas

sag

eo

f3

or

mo

reli

qu

idst

oo

ls

per

day

for

atle

ast2

con

secu

tiv

ed

ays

48

haf

ter

init

iati

on

of

anti

bio

tic

ther

apy

Stu

dy

fun

ded

by

indu

stry

Stu

dy

fun

ded

by

ind

ust

ry

Bifi

dobact

eriu

mla

ctis

and

Str

ther

mophil

us

Cor

rea

etal

(30

)N¼

15

7,

inp

atie

nts

(6–

36

mo

)

Bla

ctis

10

7C

FU

/g

and

Str

ther

mophil

us

10

6

CF

U/g

15

day

s30

day

sV

ario

us

Chan

ge

inbow

el

hab

its

wit

hth

e

pas

sag

eo

f3

or

mo

reli

qu

idst

oo

ls

per

day

for

atle

ast2

con

secu

tiv

ed

ays

Nes

tle

Nes

tle

JPGN � Volume 62, Number 3, March 2016 Probiotics for the Prevention of AAD

www.jpgn.org 499

Copyright 2016 by ESPGHAN and NASPGHAN. Unauthorized reproduction of this article is prohibited.

Ref

eren

ceP

arti

cip

ants

(age

)

Pro

bio

tic

do

se

(CF

U/d

ay)

Du

rati

on

of

inte

rven

tio

nF

oll

ow-u

p

An

tib

ioti

c/s

(ad

min

istr

atio

n)

Defi

nit

ion

of

dia

rrhea

or

AA

DM

anuf

actu

rer

Sp

onso

r

Bifi

do

ba

cter

ium

lon

gu

mP

L0

3/L

act

ob

aci

llu

srh

am

no

sus

KL

53

A/L

act

ob

aci

llu

sp

lan

taru

mP

L0

2

Szy

man

ski

etal

(34

)N¼

78

,in

pat

ien

ts

and

ou

tpat

ien

ts

(5m

o–

16

y)

2�

10

8F

or

the

du

rati

on

of

anti

bio

tic

trea

tmen

t

Du

ring

and

/or

up

to2

wee

ks

afte

r

the

end

of

the

anti

bio

tic

ther

apy

Am

oxic

illi

nw

ith

or

wit

ho

ut

clav

ula

nat

e,

ceph

alo

spo

rin

s,

pen

icil

lin

,

mac

roli

des

,

amin

og

lyco

sid

es

�3

loo

seo

rw

ater

y

sto

ols

per

day

for

am

inim

um

of

48

h,

occ

urr

ing

du

ring

and

/or

up

to

2w

eeks

afte

rth

e

end

of

the

anti

bio

tic

ther

apy

IBS

SB

iom

edIB

SS

Bio

med

SA

,

Cra

cow

,P

ola

nd

(stu

dy

pro

duc

ts)

La

cto

ba

cill

us

aci

do

ph

ilu

san

dL

act

oba

cill

us

bu

lga

ricu

s

Tan

kan

ow

etal

(31

)N¼

38

,o

utp

atie

nts

(5m

o–

6y

)

20

.4�

10

81

0d

ays

(min

imu

m5

day

s)

10

day

s

(min

imu

m5

day

s)

Am

oxic

illi

n>

1ab

no

rmal

loos

e

bo

wel

mo

vem

ent

per

day

Hy

nso

n,

Wes

tcott

,

and

Du

nn

ing

Pro

duct

s

Su

ppo

rted

infu

ll

by

Hy

nso

n,

Wes

tcott

&

Du

nn

ing

Pro

duct

s

La

cto

ba

cill

us

aci

do

ph

ilu

san

dB

ifid

oba

cter

ium

infa

nti

s

Jira

pin

yo

etal

(32

)N¼

18

,in

pat

ien

ts

(1–

36

mo

)

3ca

psu

les

dai

ly7

day

sN

ot

stat

edB

road

-sp

ectr

um

anti

bio

tics

(mai

nly

cefo

tax

ime)

No

tst

ated

No

tre

po

rted

No

tre

port

ed

La

cto

ba

cill

us

aci

do

ph

ilu

san

dB

ifid

oba

cter

ium

bre

ve

Con

tard

iet

al(3

7)

40

,o

utp

atie

nts

(1m

o–

3y

)

3�

10

9F

or

10

day

sN

ot

stat

edA

mox

icil

lin

No

tre

port

edN

ot

rep

ort

edN

ot

repo

rted

La

cto

ba

cill

us

rha

mno

sus

E/N

,O

xy

,P

en

Rus

zczy

nsk

iet

al(3

5)

24

0,

inp

atie

nts

and

ou

tpat

ien

ts

(3m

o–

14

y)

4�

10

10

Fo

rth

ed

ura

tio

no

f

the

anti

bio

tic

trea

tmen

t

Du

ring

and

/or

up

to2

wee

ks

afte

r

the

end

of

the

anti

bio

tic

ther

apy.

Pen

icil

lin

s;b

road

-

spec

tru

m

pen

icil

lin

s,

ceph

alo

spo

rin

s,

mac

roli

des

,

clin

dam

yci

n

�3

loo

sest

ools

per

day

for

am

inim

um

of

48

h,

occ

urr

ing

du

rin

gan

d/o

ru

pto

2w

kaf

ter

the

end

of

the

anti

bio

tic

ther

apy

Bio

med

Lu

bli

n

(Lak

cid

Fo

rte)

Fu

nded

inp

art

by

Bio

med

,

Lu

bli

n,

Po

lan

d,

and

the

Med

ical

Un

iver

sity

of

War

saw

La

cto

ba

cill

us

rha

mno

sus

GG

,B

ifid

ob

act

eriu

mla

ctis

Bb-

12

,an

dL

act

oba

cill

us

aci

do

ph

ilu

sL

a-5

Fo

xet

al(3

3)

70

,o

utp

atie

nts

(1y

–1

2y

)

LG

G(5

.2�

10

9),

Bb

-12

(5.9�

10

9)

and

La-

5

(8.3�

10

9)

Sam

ed

ura

tio

nas

thei

ran

tib

ioti

c

trea

tmen

t

Du

rati

on

of

trea

tmen

t

plu

s1

wee

k

b-l

acta

ms,

mac

roli

des

,

tetr

acycl

ines

Dia

rrhea

,cl

assi

fied

at

dif

fere

nt

lev

els

of

sev

erit

y:

for

exam

ple

,le

ss

sev

ere

(sto

ol

freq

uen

cy�

2/d

ay

for

2o

rm

ore

day

s

wit

hst

ool

consi

sten

cy�

5o

n

the

BS

S);

mo

re

sev

ere

(sto

ol

freq

uen

cy�

3/d

ay

for

2o

rm

ore

day

s

wit

hst

ool

consi

sten

cy�

6o

n

the

BS

S)

Par

mal

at

(Bri

sban

e,

Qu

een

slan

d,

Au

stra

lia)

Par

mal

at

(Bri

sban

e,

Qu

een

slan

d,

Au

stra

lia)

Szajewska et al JPGN � Volume 62, Number 3, March 2016

500 www.jpgn.org

Copyright 2016 by ESPGHAN and NASPGHAN. Un

Ref

eren

ceP

arti

cip

ants

(age

)

Pro

bio

tic

do

se

(CF

U/d

ay)

Du

rati

on

of

inte

rven

tio

nF

oll

ow-u

p

An

tib

ioti

c/s

(ad

min

istr

atio

n)

Defi

nit

ion

of

dia

rrhea

or

AA

DM

anuf

actu

rer

Sp

onso

r

La

cto

ba

cill

us

aci

do

ph

ilu

san

dL

act

oba

cill

us

rha

mno

sus

and

La

cto

ba

cill

us

bu

lga

ricu

sa

nd

La

cto

ba

cill

us

case

ian

dStr

ther

mophil

us

and

Bifi

do

ba

cter

ium

infa

nti

san

dB

ifid

oba

cter

ium

bre

ve

Kh

od

adad

etal

(38

)N¼

66

,in

pat

ien

ts

(3–

14

y)

1�

10

9C

FU

/day

Fo

rth

ed

ura

tio

no

f

Hel

ico

ba

cter

pyl

ori

erad

icat

ion

ther

apy

(4w

k?;

no

tcl

earl

yst

ated

)

No

tst

ated

Am

oxic

illi

n,

fura

zoli

done

No

tst

ated

Pro

tex

inC

o,U

KN

ot

repo

rted

Kefi

r

Mer

enst

ein

etal

(36

)N¼

12

5,

ou

tpat

ien

ts

(1–

5y

)

At

leas

th

alf

of

a

15

0m

Ld

rin

k

con

tain

ing

7to

10

9C

FU

bac

teri

a

and

yea

st

Fo

rth

ed

ura

tio

n

of

anti

bio

tic

trea

tmen

t

(10

day

s)

14

day

sA

nti

bio

tics

for

up

per

resp

irat

ory

trac

t

infe

ctio

ns

(no

t

spec

ified

oth

erw

ise)

No

tre

port

edP

robu

gs

(Lif

eway

Fo

ods,

Inc,

Chi

cag

o,

IL)

Lif

eway

Fo

od

s,

Inc

AA

anti

bio

tic-

asso

ciat

edd

iarr

hea

;B

SS¼

Bri

sto

lS

too

lS

cale

;C

FU¼

colo

ny

-fo

rmin

gu

nit

s;S

Sb

ou

lard

ii.

�A

sca

lcu

late

db

yth

eau

tho

rs.

Arvola 1999

Ran

dom

seq

uenc

e ge

nera

tion

(sel

ectio

n bi

as)

Allo

catio

n co

ncea

lmen

t (se

lect

ion

bias

)

Blin

ding

of p

artic

ipan

ts a

nd p

erso

nnel

(pe

rfor

man

ce b

ias)

Blin

ding

of o

utco

me

asse

ssm

ent (

dete

ctio

n bi

as)

Inco

mpl

ete

outc

ome

data

(at

triti

on b

ias)

Sel

ectiv

e re

port

ing

(rep

ortin

g bi

as)

Bin 2015

Casem 2013

Contardi 1991

Correa 2005

Destura (unpublished)

Erdeve 2004

Fox 2015

Jirapinyo 2002

Khodadad 2013

King 2010

Kotowska 2005

Merenstein 2009

Ruszczynski 2008

Shan 2013

Szajewska 2009

Szymanski 2008

Tankanow 1990

Vaisanen 1998

Vanderhoof 1999

Zhao 2014

FIGURE 1. Methodological quality summary.

JPGN � Volume 62, Number 3, March 2016 Probiotics for the Prevention of AAD

www.jpgn.org

A 2015 systematic review with a meta-analysis (40) ident-ified 5 relevant RCTs (445 participants) (18–22). The methodo-logical quality of the trials varied (Fig. 1). Only 1 trial was at a lowrisk of bias. In the remaining trials, the limitations included unclearrandom sequence generation, unclear or no allocation concealment,

authorized reproduction of this article is prohibited.

and unclear or no blinding of participants and personnel. Intention-to-treat analysis was performed in only 1 trial. Using the GRADE,

501

Co

1.1.1 S boulardiiKotowska 2005Erdeve 2004Shan 2013Bin 2015Zhao 2014Casem 2013Subtotal (95% CI)Total eventsHeterogeneity Tau2 = 0.07; Chi2 = 8.26, df = 5(P = 0.14); I2 = 39%

Test for overall effect: Z = 4.87 (P < 0.00001)

Vanderhoof 1999Szajewska 2009Arvola 1999King 2010Vaisanen 1998

Total events

1.1.3 B clausiiDestura (unpublished)

Heterogeneity : Not applicable

Heterogeneity : Not applicable

Total events

Total events

Total events 13

Subtotal (95% CI)

Test for overall effect: Z = 1.25 (P = 0.21)

Test for overall effect: Z = 2.13 (P = 0.03)

Heterogeneity : Not applicableTest for overall effect: Z = 0.62 (P = 0.54)

Total eventsHeterogeneity : Not applicableTest for overall effect: Z = 0.19 (P = 0.85)

1.1.4 B lactis & Str themophilus

1.1.6 L acidophilus & bulgaricus

1.1.5 B longum PL03 & L rhamnosus KL53A & L plantrarum PL02

Correa 2005Subtotal (95% CI)

Subtotal (95% CI)Szymanski 2008

Tankanow 1990

Total eventsHeterogeneity : Not applicableTest for overall effect: Z = 0.57 (P = 0.12)

Jirapinyo 2002

1.1.7 L acidophilus & B infantis

Total eventsHeterogeneity : Not applicableTest for overall effect Not applicable

Contardi 1991

1.1.8 L acidophilus & B breve

Subtotal (95% CI)

Subtotal (95% CI)

Subtotal (95% CI)

4146

122711

72336

21

3

3

1

10

13

1

10

3

3

0

0

13224416710512069

837

9334618

23219

162162

8080

4040

1515

88

2020

224218264716

74 171

256948

52

7

7

24

24

2

2

16

16

8

8

0

0

137222166100120

71816

9530587

36226

161161

7777

7.4%7.4%

3838

2323

1010

2020

Not estimableNot estimable

4.0%7.6%4.8%7.1%9.7%6.5%

39.7%

0.19 (0.07, 0.53)0.30 (0.17, 0.54)0.33 (0.13, 0.81)0.44 (0.23, 0.82)0.57 (0.39, 0.86)0.71 (0.35, 1.41)0.43 (0.30, 0.60)

5.6%2.2%3.0%3.7%4.7%

19.3%

0.29 (0.13, 0.63)0.29 (0.06, 1.35)0.32 (0.09, 1.11)0.66 (0.22, 1.97)1.17 (0.47, 2.95)0.48 (0.26, 0.89)

2.8%2.8%

0.43 (0.11, 1.62)0.43 (0.11, 1.62)

0.52 (0.29, 0.95)0.52 (0.29, 0.95)

1.0%1.0%

0.47 (0.04, 5.03)0.47 (0.04, 5.03)

9.1%9.1%

0.96 (0.61, 1.50)0.96 (0.61, 1.50)

4.5%4.5%

0.47 (0.18, 1.21)0.47 (0.18, 1.21)

Test for overall effect: Z =2.33(P = 0.02)Heterogeneity Tau2 = 0.07; Chi2 = 8.26, df = 5(P = 0.14); I2 = 40%

Subtotal (95% CI)

1.1.2 Lactobacillus GG

Study or subgroup Events Total EventsProbiotics

Total WeightControl

M-H, Random, 95% CIRisk ratio

M-H, Random, 95% CIRisk ratio

A B C D E FRisk of bias

0.01 0.1Favours probiotics Favours control

1 10 100

FIGURE 2. Effect of individual probiotic strains and probiotics as a group for preventing antibiotic-associated diarrhea.

(Continued on next page )

Szajewska et al JPGN � Volume 62, Number 3, March 2016

the overall quality of evidence was rated as moderate (Table S1,http://links.lww.com/MPG/A587).

Compared with placebo or no treatment, LGG administrationin children reduced the risk of AAD, regardless of the reason forwhich probiotics were used (ie, as part of Helicobacter pylorieradication or for other reasons), from 23% to 9.6% (5 RCTs,n¼ 445, RR 0.48, 95% CI 0.26–0.89; number needed to treat,

pyright 2016 by ESPGHAN and NASPGHAN. Un

NNT, 8, 95% CI 6–40) (Fig. 2). No significant heterogeneity wasfound (x2¼ 6.61, P¼ 0.16, I2¼ 40%). Only 1 trial (19) evaluated the

502

effect of LGG on the risk of C difficile-associated diarrhea in childrenand found no effect (RR 0.95, 95% CI 0.06–14.85) (Fig. 3).

The optimal daily dose of LGG for preventing AAD remainsunclear (40). In children, the best effect (reduction in the risk ofAAD by 71%) was achieved with the highest dose (1–2� 1010

CFU) (18). A similar effect size was, however, not achieved inanother trial using the same dose (19), perhaps because of a lower

authorized reproduction of this article is prohibited.

baseline risk of AAD. In adults, there was no clear link between theeffect size and the LGG dose.

www.jpgn.org

Co

Total eventsHeterogeneity : Not applicableTest for overall effect: Z = 1.85 (P = 0.06)

Test for overall effect: Z = 2.10 (P = 0.04)

Test for overall effect: Z = 3.00 (P = 0.003)

1.1.11 L rhamnosus GG & Bb12 & L acidophilus

1.1.9 L acidophilus & L rhamnosus & L bulgaricus & L casei & Str therophilus & B infantis & B breve

Subtotal (95% CI)Khodadad 2013

Total eventsHeterogeneity : Not applicable

Total eventsHeterogeneity : Not applicable

Test for overall effect: Z = 0.53 (P = 0.60)

Test for overall effect: Z = 5.80 (P < 000001)

1.1.12 Kefir

Total events

Total events

Heterogeneity : Not applicable

Heterogeneity Tau2 = 0.13; Chi2 = 34.51, df = 19 (P = 0.02); I2 = 45%

Test for subgroup differences: Chi2 = 16.82, df = 10 (P = 0.08); I2 = 40.6%

1.1.10 L rhamnosus E/N, Oxy, PenRuszczynki 2008

Fox 2015

Subtotal (95% CI)

Subtotal (95% CI)

Subtotal (95% CI)

Total (95% CI)

Merenstein 2009

2

2

9

1

11

148

9

1

11

3333

120120

3434

5757

1625

8

8

20

20

21

21

14

14

343

3333

120120

3636

6060

1620

2.4%2.4%

0.25 (0.06, 1.09)0.25 (0.06, 1.09)

6.0%6.0%

0.45 (0.21, 0.95)0.45 (0.21, 0.95)

1.5%1.5%

0.05 (0.01, 0.35)0.05 (0.01, 0.35)

6.4%6.4%

0.83 (0.41, 1.67)0.83 (0.41, 1.67)

100% 0.48 (0.37, 0.61)

Risk of bias legend

Random sequence generation (selection bias)Allocation concealment (selection bias)Blinding of participants and personnel (performance bias)Blinding of outcome assessment (detection bias)Incomplete outcome data (attrition bias)Selective reporting (reporting bias)

(A)(B)(C)(D)(E)(F)

0.01 0.1Favours probiotics Favours control

1 10 100

FIGURE 2. (Continued )

JPGN � Volume 62, Number 3, March 2016 Probiotics for the Prevention of AAD

Saccharomyces boulardii

RECOMMENDATION. If the use of probiotics forpreventing AAD in children is considered, the WG recom-mends using S boulardii for preventing AAD in children.QUALITY OF EVIDENCE: Moderate.STRENGTH OF RECOMMENDATION: Strong.RECOMMENDATION. If the use of probiotics for pre-venting C difficile-associated diarrhea in children isconsidered, the WG suggests using S boulardii.QUALITY OF EVIDENCE: Low.STRENGTH OF RECOMMENDATION: Conditional.

A 2015 systematic review with a meta-analysis (39) ident-ified 6 relevant RCTs (1653 participants) (23–28). The methodo-logical quality of the trials varied. Only 1 trial was at a low risk ofbias. In the remaining trials, the limitations included unclearrandom sequence generation, unclear or no allocation concealment,and unclear or no blinding of participants and personnel. Intention-to-treat analysis was performed in only 2 trials. Using the GRADE,the overall quality of evidence for AAD and C difficile-associateddiarrhea was rated as moderate and low, respectively (Tables S2 andS3, http://links.lww.com/MPG/A587).

Compared with placebo or no treatment, S boulardii admin-istration in children reduced the risk of diarrhea, regardless of the

pyright 2016 by ESPGHAN and NASPGHAN. Un

reason for which probiotics were used (ie, as part of H pylorieradication or for other reasons), from 20.9% to 8.8% (6 RCTs,

www.jpgn.org

n¼ 1653, RR 0.43, 95% CI 0.30–0.60, NNT 9, 95% CI 7–12). Nosignificant heterogeneity was found (x2¼ 8.26, P¼ 0.14, I2¼ 39%)(Fig. 2).

The administration of S boulardii also reduced the risk of Cdifficile-associated diarrhea in children (2 RCTs, n¼ 579, RR 0.25,95% CI 0.08–0.73) (Fig. 3).

The optimal dose of S boulardii has not been established. A2015 meta-analysis showed that various doses of S boulardii wereused with no clear dose-dependent effect (39). Until more data onthe optimal dose of S boulardii become available, a daily dose of not<250 mg but not >500 mg in children and not >1000 mg in adultscould be used to match the doses used in RCTs.

PROBIOTICS WITH INSUFFICIENT EVIDENCETO MAKE A RECOMMENDATION

Single ProbioticsBacillus clausii

A 2011 Cochrane review (7) identified 1 unpublished RCT(29). Compared with no intervention, administration of Bacillusclausii (strain specification not given) had no effect on the risk ofAAD (n¼ 323, RR 0.43, 95% CI 0.11–1.62).

Mixtures of Probiotics

Bacillus lactis/Streptococcus thermophilusOne RCT (n¼ 157) conducted in inpatients who were chil-

dren (aged 6–36 months) showed that compared with the control

authorized reproduction of this article is prohibited.

formula, the administration of infant formula supplemented with Blactis Bb-12 and Streptococcus thermophilus significantly reduced

503

Co

Study or subgroup Events TotalTreatment

Events Total WeightControl

M-H, Random, 95% CIRisk ratio

M-H, Random, 95% CIRisk ratio

A B C D E FRisk of bias

Risk of bias legendRandom sequence generation (selection bias)Allocation concealment (selection bias)Blinding of participants and personnel (performance bias)Blinding of outcome assessment (detection bias)Incomplete outcome data (attrition bias)Selective reporting (reorting bias)

(A)(B)(C)(D)(E)(F)

Kotowska 20051.2.1 S boulardii

1.2.3 L rhamnosus E/N, Oxy, Pen

1.2.2 Lactobacillus GG

Shan 2013Subtotal (95% CI)

119167

108

286

127166293

40.1%15.0%55.1%

0.32 (0.09, 1.14)0.12 (0.02, 0.98)0.25 (0.08, 0.73)

Subtotal (95% CI) 61 58 8.5% 0.95 (0.06, 14.85)Arvola 1999 61 58 8.5% 0.95 (0.06, 14.85)

Subtotal (95% CI) 120 120 36.4% 0.43 (0.11, 1.62)

467 471 100.0% 0.34 (0.15, 0.76)

Ruszczynski 2008 120 120 36.4% 0.43 (0.11, 1.62)

Total eventsHeterogeneity: Tau2 = 0.00; Chi2 = 0.60, df = 1 (P =0.44); I2 = 0%Test for overall effect: Z = 2.54 (P = 0.01)

Total events

Total events

Total (95 % CI)

Heterogeneity: Tau2 = 0.00; Chi2 = 1.61, df = 3 (P = 0.66); I2 = 0%

Heterogeneity: Not applicable

Test for overall effect: Z = 2.65 (P = 0.008)

Test for overall effect: Z = 1.25 (P = 0.21)

Heterogeneity: Not applicableTest for overall effect: Z = 0.04 (P = 0.97)

Test for subgroup differences: Chi2 = 0.99, df = 2 (P = 0.61), I2 = 0%

3

73

73

Total events 11

11

268

1

4

0.01 0.1Favours treatment Favours control

1 10 100

oup

Szajewska et al JPGN � Volume 62, Number 3, March 2016

the risk of AAD (31.2% vs 16.3%, respectively; RR 0.52, 95% CI0.29–0.95, NNT 7, 95% CI 4–62) (30).

L acidophilus/L bulgaricus

One small RCT (n¼ 38) showed that compared with placebo(lactose), administration of L acidophilus/L bulgaricus (strainspecification not given) had no effect on the risk of AAD (RR0.96, 95% CI 0.61–1.5) (31).

L acidophilus/Bifidobacterium infantis

One small RCT (n¼ 18) showed that compared with placebo(sugar), administration of L acidophilus/B infantis (strain specifica-tion not given) had no effect on the risk of AAD (8/10 vs 3/8,respectively; RR 0.47, 95% 0.18–1.21) (32).

L acidophilus/Bifidobacterium breve

One small RCT (n¼ 40) showed no cases of AAD in eitherthe L acidophilus/B infantis (strain specification not given) groupor the placebo (sugar) group (0/20 vs 0/20, respectively). Thus,the efficacy of this probiotic combination could not be evaluated(37).

L rhamnosus GG/Bb-12/L acidophilus La-5

In a multisite, double-blind, placebo-controlled RCT, chil-

FIGURE 3. Effect of individual probiotic strains and probiotics as a gr

pyright 2016 by ESPGHAN and NASPGHAN. Un

dren (n¼ 70), age 1 to 12 years, who were prescribed antibioticswere randomized to receive 200 g/day of either a yogurt containing

504

L rhamnosus GG, Bb-12 and L acidophilus La-5 or a pasteurizedplacebo yogurt (containing Streptococcus thermophilus plus Lbulgaricus) for the same duration as their antibiotic treatment.Compared with the placebo group, children in the probiotic groupexperienced a significant reduction in the risk of diarrhea (RR 0.05,95% CI 0.01–0.35) (33).

B longum PL03/L rhamnosus KL53A/Lplantarum PL02

One RCT (n¼ 78) showed that compared with placebo, theadministration of B longum, L rhamnosus, and L plantarum had noeffect on the risk of AAD (RR 0.47, 95% CI 0.04–5.03) (34).

L rhamnosus E/N, Oxy, Pen

One RCT involving 240 children showed that compared withplacebo, the administration of L rhamnosus (strains E/N, Oxy andPen) reduced the risk of any diarrhea (RR 0.45, 95% CI 0.21–0.95),but it did not have an effect on the risk of C difficile-associateddiarrhea (RR 0.43, 95% CI 0.11–1.62) (35).

L acidophilus/L rhamnosus/L bulgaricus/Lcasei/Str thermophilus/B infantis/B breve

One RCT involving 66 children showed that comparedwith placebo, the administration of L acidophilus/L rhamnosus/Lbulgaricus/L casei/Str thermophilus/B infantis/B breve (strain

for preventing Clostridium difficile-associated diarrhea.

authorized reproduction of this article is prohibited.

specification not given) reduced the risk of diarrhea (RR 0.25,95% CI 0.06–1.09) (38).

www.jpgn.org

Co

Kefir

One RCT evaluated the effect of kefir (ie, a fermented milkcontaining Lactococcus lactis, Lactococcus plantarum, Lacto-coccus rhamnosus, Lactococcus casei, Lactococcus lactis sub-species diacetylactis, Leuconostoc cremoris, B longum, B breve,Lactobacillus acidophilus, and 1 yeast, Saccharomyces florentinus)on the risk of AAD. There was no significant difference between thekefir group and the group receiving heat-treated kefir (RR 0.83,95% CI 0.41–1.67) (36).

Yogurt

Yogurt is a form of fermented milk that contains symbioticcultures of Streptococcus thermophilus and L delbrueckii subsp.bulgaricus. A 2015 systematic review with a meta-analysis ident-ified 2 relevant RCTs, both low in methodological quality. Com-pared with no intervention, yogurt consumption had no effect on therisk of AAD (2 RCTs, n¼ 314, RR 0.45; 95% CI 0.11–1.75) (41).

SAFETYThe WG abstained from evaluating the safety of probiotics,

as this was thoroughly reviewed in 2011 by the US Agency forHealthcare Research and Quality (for review, (42)). Althoughprobiotics are safe for use in otherwise healthy populations, cautionshould be taken in specific patient groups. Risk factors for adverseevents include immunosuppression, prematurity, critical illness,presence of structural heart disease, presence of a central venouscatheter, and the potential for translocation of probiotics across thebowel wall. There is a lack of data that specifically address thesafety of probiotics for preventing AAD in these vulnerable popu-lations. The risk of side effects is, however, greater in people whohave severe underlying health conditions.

JPGN � Volume 62, Number 3, March 2016

pyrig

www

SUMMARY

The WG questions pooling different probiotic strains togetherin a meta-analysis. Probiotic effects against AAD are strainspecific; thus, the efficacy and safety of each should be

e stablished and recommendations for using these strainsshould be made accordingly.

� T

he safety and clinical effects of 1 probiotic microorganismshould not be extrapolated to other probiotic microorganisms.A lack of evidence regarding the efficacy of a certain �p robiotic(s) does not mean that future studies will notestablish efficacy in preventing AAD.There is a lack of data that specifically address the safety of �p robiotics for preventing AAD in children who have severeunderlying health conditions.The WG recommends choosing a probiotic, the efficacyof which has been confirmed in well-conducted RCTs,from a manufacturer who has a regulated quality control of f actors including the composition and content of theprobiotic agent.Risk factors for the occurrence of AAD or C difficile-associated diarrhea such as class of antibiotic(s), duration ofantibiotic treatment, age, need for hospitalization, comorbid-ities, and previous episodes of AAD or C difficile-associated d iarrhea should be considered when making decisions on theuse of probiotics in children for preventing AAD.If the use of probiotics for preventing AAD is considered, the

ht 2016 by ESPGHAN and NASPGHAN. Un

WG recommends using L rhamnosus GG or S boulardii(moderate quality of evidence; strong recommendation).

.jpgn.org

auth

prca

If the use of probiotics for preventing C difficile-associated

Probiotics for the Prevention of AAD

�diarrhea is considered, the WG suggests using S boulardii(low quality of evidence; conditional recommendation).

REFERENCES1. Barbut F, Meynard JL. Managing antibiotic associated diarrhoea. BMJ

2002;324:1345–6.2. McFarland LV. Epidemiology, risk factors and treatments for antibiotic-

associated diarrhea. Dig Dis 1998;16:292–307.3. McFarland LV, Surawicz CM, Stamm WE. Risk factors for Clostridium

difficile carriage and C. difficile-associated diarrhea in a cohort ofhospitalized patients. J Infect Dis 1990;162:678–84.

4. McFarland LV. Meta-analysis of probiotics for the prevention of anti-biotic associated diarrhea and the treatment of Clostridium difficiledisease. Am J Gastroenterol 2006;101:812–22.

5. Hill C, Guarner F, Reid G, et al. Expert consensus document. TheInternational Scientific Association for Probiotics and Prebiotics con-sensus statement on the scope and appropriate use of the term probiotic.Nat Rev Gastroenterol Hepatol 2014;11:506–14.

6. Szajewska H, Guarino A, Hojsak I, et al., European Society for PediatricGastroenterology, Hepatology, and Nutrition. Use of probiotics formanagement of acute gastroenteritis: a position paper by the ESPGHANWorking Group for Probiotics and Prebiotics. J Pediatr GastroenterolNutr 2014;58:531–9.

7. Johnston BC, Goldenberg JZ, Vandvik PO, et al. Probiotics for theprevention of pediatric antibiotic-associated diarrhea. Cochrane Data-base Syst Rev 2011CD004827.

8. Guarner F, Khan AG, Garisch J, et al., World Gastroenterology Orga-nization. World Gastroenterology Organisation Global Guidelines:probiotics and prebiotics October 2011. J Clin Gastroenterol 2012;46:468–81.

9. Grzeskowiak Ł, Isolauri E, Salminen S, et al. Manufacturing processinfluences properties of probiotic bacteria. Br J Nutr 2011;105:887–94.

10. Sanders ME, Guarner F, Guerrant R, et al. An update on the useand investigation of probiotics in health and disease. Gut 2013;62:787–96.

11. Higgins JPT, Altman DG, Sterne AC. Assessing risk of bias in includedstudies. In: Higgins JPT, Green S, eds. Cochrane Handbook for Sys-tematic Reviews of Interventions. Version 5.1.0. www.cochrane-handbook.org. Published 2011. Accessed July 2015.

12. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging consensuson rating quality of evidence and strength of recommendations. BMJ2008;336:924–6.

13. Andrews J, Guyatt G, Oxman AD, et al. GRADE guidelines: 14. Goingfrom evidence to recommendations: the significance and presentation ofrecommendations. J Clin Epidemiol 2013;66:719–25.

14. Center for Drug Evaluation and Research; Center for Biologics Eva-luation and Research. Guidance for industry: providing clinical evi-dence of effectiveness for human drug and biological products. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm078749.pdf. Published May 1998. AccessedJuly 2015.

15. Rijkers GT, Bengmark S, Enck P, et al. Guidance for substantiatingthe evidence for beneficial effects of probiotics: current status andrecommendations for future research. J Nutr 2010;140:671S–6S.

16. Hempel S, Newberry SJ, Maher AR, et al. Probiotics for the preventionand treatment of antibiotic-associated diarrhea: a systematic review andmeta-analysis. JAMA 2012;307:1959–69.

17. Goldenberg JZ, Ma SS, Saxton JD, et al. Probiotics for the preventionof Clostridium difficile-associated diarrhea in adults and children.Cochrane Database Syst Rev 2013;5:CD006095.

18. Vanderhoof JA, Whitney DB, Antonson DL, et al. Lactobacillus GG inthe prevention of antibiotic-associated diarrhea in children. J Pediatrics1999;135:564–8.

19. Arvola T, Laiho K, Torkkeli S, et al. Prophylactic Lactobacillus GGreduces antibiotic-associated diarrhea in children with respiratoryinfections: a randomized study. Pediatrics 1999;104:e64.

20. King SN, Chung AM, Vidal R. Randomized, double blind, placebocontrolled trial to assess the efficacy of Lactobacillus GG in the

orized reproduction of this article is prohibited.

evention of antibiotic-associated diarrhea in the pediatric internsivere unit (PICU). Pharmacotherapy 2010;30:457–8.

505

Co

21. Vaisanen LM, Leskinen M, Siitonen A, et al. Occurence of diarrhea inchildren receiving oral antibiotics with or without probiotic supple-mentation with Lactobacillus GG. Microb Ecol Health Dis 1998;10:199–200.

22. Szajewska H, Albrecht P, Topczewska-Cabanek A. Randomized, double-blind, placebo-controlled trial: effect of lactobacillus GG supplementationon Helicobacter pylori eradication rates and side effects during treatmentin children. J Pediatr Gastroenterol Nutr 2009;48:431–6.

23. Kotowska M, Albrecht P, Szajewska H. Saccharomyces boulardii in theprevention of antibiotic-associated diarrhea in children: a randomizeddouble-blind placebo-controlled trial. Aliment Pharmacol Ther 2005;21:583–90.

24. Casem RAO. Saccharomyces boulardii in the prevention of antibioticassociated diarrhea in children: a randomized controlled trial. PediatrInfect Dis Soc Philipp J 2013;14:70–6.

25. Erdeve O, Tiras U, Dallar Y. The probiotic effect of Saccharomycesboulardii in a pediatric age group. J Trop Pediatr 2004;50:234–6.

26. Shan LS, Hou P, Wang ZJ, et al. Prevention and treatment of diarrhoeawith Saccharomyces boulardii in children with acute lower respiratorytract infections. Benef Microbes 2013;4:329–34.

27. Bin Z, Ya-Zheng X, Zhao-Hui D, et al. The efficacy of Saccharomycesboulardii CNCM I-745 in addition to standard Helicobacter pylorieradication treatment in children. Pediatr Gastroenterol Hepatol Nutr2015;18:17–22.

28. Zhao HM, Ou-Yang HJ, Duan BP, et al. Clinical effect of triple therapycombined with Saccharomyces boulardii in the treatment of Helico-bacter pylori infection in children. Zhongguo Dang Dai Er Ke Za Zhi2014;16:230–3.

29. Destura RV. Bacillus clausii in preventing antibiotic-associated diarrheaamong Filipino infants and children: a multi-center, randomized, open-label clinical trial of efficacy and safety. Unpublished.

30. Correa NBO, Peret Filho LA, Penna FJ, et al. A Randomized formulacontrolled trial of Bifidobacterium lactis and Streptococcus thermophi-lus for prevention of antibiotic-associated diarrhea in infants. J ClinGastroenterol 2005;39:385–9.

31. Tankanow RM, Ross MB, Ertel IJ, et al. A double-blind, placebo-

Szajewska et al

pyright 2016 by ESPGHAN and NASPGHAN. Un

amoxicillin-induced diarrhea. DICP 1990;24:382–4.

506

32. Jirapinyo P, Densupsoontorn N, Thamonsiri N, et al. Prevention ofantibiotic-associated diarrhea in infants by probiotics. J Med Assoc Thai2002;85(suppl 2):S739–42.

33. Fox MJ, Ahuja KDK, Robertson IK, et al. Can probiotics yogurt preventdiarrhea in children on antibiotics? A double-blind, randomized,placebo-controlled study. BMJ OPEN 2015;5:e006474.

34. Szymanski H, Armanska M, Kowalska-Duplaga K, et al. Bifidobacter-ium longum PL03, Lactobacillus rhamnosus KL53A, andLactobacillus plantarum PL02 in the prevention of antibiotic-associateddiarrhea in children: a randomized controlled pilot trial. Digestion2008;78:13–7.

35. Ruszczynski M, Radzikowski A, Szajewska H. Clinical trial: effective-ness of Lactobacillus rhamnosus (strains E/N, Oxy and Pen) in theprevention of antibiotic-associated diarrhoea in children. Aliment Phar-macol Ther 2008;28:154–61.

36. Merenstein DJ, Foster J, D’Amico F. A randomized clinical trialmeasuring the influence of kefir on antibiotic-associated diarrhea: themeasuring the influence of Kefir (MILK) Study. Arch Pediatr AdolescMed 2009;163:750–4.

37. Contardi I. Oral bacterial therapy in prevention of antibiotic-induceddiarrhea in childhood [Batterioterapia orale quale prevenzione delladiarrea da antibiotici in eta pediatrica]. Clinica Terapeutica 1991;136:409–13.

38. Khodadad A, Farahmand F, Najafi M, et al. Probiotics for the treatmentof pediatric Helicobacter pylori infection: a randomized double blindclinical trial. Iran J Pediatr 2013;23:79–84.

39. Szajewska H, Kołodziej M. Systematic review with meta-analysis:Saccharomyces boulardii in the prevention of antibiotic-associateddiarrhoea. Aliment Pharmacol Ther 2015;42:793–801.

40. Szajewska H, Kołodziej M. Systematic review with meta-analysis:Lactobacillus GG in the prevention of antibiotic-associated diarrhoea.Aliment Pharmacol Ther 2015;42:1149–57.

41. Patro-Gołab B, Sharmi R, Szajewska H. Yogurt for treating antibiotic-associated diarrhea: systematic review and meta-analysis. Nutrition2015;31:796–800.

42. Hempel S, Newberry S, Ruelaz A, et al. Safety of probiotics used

JPGN � Volume 62, Number 3, March 2016

to reduce risk and prevent or treat disease. Evid Rep Technol Assess

controlled study of the efficacy of Lactinex in the prophylaxis of

authorized reproduction of this article is prohibited.

(Full Rep) 2011;200:1–645.

www.jpgn.org