clinical practice guideline the diagnosis and ......fewer antibiotic prescriptions written. despite...

38
CLINICAL PRACTICE GUIDELINE The Diagnosis and Management of Acute Otitis Media abstract This evidence-based clinical practice guideline is a revision of the 2004 acute otitis media (AOM) guideline from the American Academy of Pe- diatrics (AAP) and American Academy of Family Physicians. It provides recommendations to primary care clinicians for the management of children from 6 months through 12 years of age with uncomplicated AOM. In 2009, the AAP convened a committee composed of primary care physicians and experts in the elds of pediatrics, family practice, oto- laryngology, epidemiology, infectious disease, emergency medicine, and guideline methodology. The subcommittee partnered with the Agency for Healthcare Research and Quality and the Southern Califor- nia Evidence-Based Practice Center to develop a comprehensive review of the new literature related to AOM since the initial evidence report of 2000. The resulting evidence report and other sources of data were used to formulate the practice guideline recommendations. The focus of this practice guideline is the appropriate diagnosis and initial treatment of a child presenting with AOM. The guideline provides a specic, stringent denition of AOM. It addresses pain management, initial observation versus antibiotic treatment, appropriate choices of antibiotic agents, and preventive measures. It also addresses recur- rent AOM, which was not included in the 2004 guideline. Decisions were made on the basis of a systematic grading of the quality of evidence and benet-harm relationships. The practice guideline underwent comprehensive peer review before formal approval by the AAP. This clinical practice guideline is not intended as a sole source of guid- ance in the management of children with AOM. Rather, it is intended to assist primary care clinicians by providing a framework for clinical decision-making. It is not intended to replace clinical judgment or es- tablish a protocol for all children with this condition. These recommend- ations may not provide the only appropriate approach to the management of this problem. Pediatrics 2013;131:e964e999 Allan S. Lieberthal, MD, FAAP, Aaron E. Carroll, MD, MS, FAAP, Tasnee Chonmaitree, MD, FAAP, Theodore G. Ganiats, MD, Alejandro Hoberman, MD, FAAP, Mary Anne Jackson, MD, FAAP, Mark D. Joffe, MD, FAAP, Donald T. Miller, MD, MPH, FAAP, Richard M. Rosenfeld, MD, MPH, FAAP, Xavier D. Sevilla, MD, FAAP, Richard H. Schwartz, MD, FAAP, Pauline A. Thomas, MD, FAAP, and David E. Tunkel, MD, FAAP, FACS KEY WORDS acute otitis media, otitis media, otoscopy, otitis media with effusion, watchful waiting, antibiotics, antibiotic prophylaxis, tympanostomy tube insertion, immunization, breastfeeding ABBREVIATIONS AAFPAmerican Academy of Family Physicians AAPAmerican Academy of Pediatrics AHRQAgency for Healthcare Research and Quality AOMacute otitis media CIcondence interval FDAUS Food and Drug Administration LAIVlive-attenuated intranasal inuenza vaccine MEEmiddle ear effusion MICminimum inhibitory concentration NNTnumber needed to treat OMotitis media OMEotitis media with effusion ORodds ratio PCV7heptavalent pneumococcal conjugate vaccine PCV1313-valent pneumococcal conjugate vaccine RDrate difference SNAPsafety-net antibiotic prescription TIVtrivalent inactivated inuenza vaccine TMtympanic membrane WASPwait-and-see prescription This document is copyrighted and is property of the American Academy of Pediatrics and its Board of Directors. All authors have led conict of interest statements with the American Academy of Pediatrics. Any conicts have been resolved through a process approved by the Board of Directors. The American Academy of Pediatrics has neither solicited nor accepted any commercial involvement in the development of the content of this publication. The recommendations in this report do not indicate an exclusive course of treatment or serve as a standard of medical care. Variations, taking into account individual circumstances, may be appropriate. (Continued on last page) e964 FROM THE AMERICAN ACADEMY OF PEDIATRICS Organizational Principles to Guide and Dene the Child Health Care System and/or Improve the Health of all Children by guest on April 9, 2020 www.aappublications.org/news Downloaded from

Upload: others

Post on 02-Apr-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

CLINICAL PRACTICE GUIDELINE

The Diagnosis and Management of Acute Otitis Media

abstractThis evidence-based clinical practice guideline is a revision of the 2004acute otitis media (AOM) guideline from the American Academy of Pe-diatrics (AAP) and American Academy of Family Physicians. It providesrecommendations to primary care clinicians for the management ofchildren from 6 months through 12 years of age with uncomplicatedAOM.

In 2009, the AAP convened a committee composed of primary carephysicians and experts in the fields of pediatrics, family practice, oto-laryngology, epidemiology, infectious disease, emergency medicine,and guideline methodology. The subcommittee partnered with theAgency for Healthcare Research and Quality and the Southern Califor-nia Evidence-Based Practice Center to develop a comprehensive reviewof the new literature related to AOM since the initial evidence report of2000. The resulting evidence report and other sources of data wereused to formulate the practice guideline recommendations.

The focus of this practice guideline is the appropriate diagnosis andinitial treatment of a child presenting with AOM. The guideline providesa specific, stringent definition of AOM. It addresses pain management,initial observation versus antibiotic treatment, appropriate choices ofantibiotic agents, and preventive measures. It also addresses recur-rent AOM, which was not included in the 2004 guideline. Decisions weremade on the basis of a systematic grading of the quality of evidenceand benefit-harm relationships.

The practice guideline underwent comprehensive peer review beforeformal approval by the AAP.

This clinical practice guideline is not intended as a sole source of guid-ance in the management of children with AOM. Rather, it is intended toassist primary care clinicians by providing a framework for clinicaldecision-making. It is not intended to replace clinical judgment or es-tablish a protocol for all children with this condition. These recommend-ations may not provide the only appropriate approach to themanagement of this problem. Pediatrics 2013;131:e964–e999

Allan S. Lieberthal, MD, FAAP, Aaron E. Carroll, MD, MS,FAAP, Tasnee Chonmaitree, MD, FAAP, Theodore G. Ganiats,MD, Alejandro Hoberman, MD, FAAP, Mary Anne Jackson,MD, FAAP, Mark D. Joffe, MD, FAAP, Donald T. Miller, MD,MPH, FAAP, Richard M. Rosenfeld, MD, MPH, FAAP, Xavier D.Sevilla, MD, FAAP, Richard H. Schwartz, MD, FAAP, Pauline A.Thomas, MD, FAAP, and David E. Tunkel, MD, FAAP, FACS

KEY WORDSacute otitis media, otitis media, otoscopy, otitis media witheffusion, watchful waiting, antibiotics, antibiotic prophylaxis,tympanostomy tube insertion, immunization, breastfeeding

ABBREVIATIONSAAFP—American Academy of Family PhysiciansAAP—American Academy of PediatricsAHRQ—Agency for Healthcare Research and QualityAOM—acute otitis mediaCI—confidence intervalFDA—US Food and Drug AdministrationLAIV—live-attenuated intranasal influenza vaccineMEE—middle ear effusionMIC—minimum inhibitory concentrationNNT—number needed to treatOM—otitis mediaOME—otitis media with effusionOR—odds ratioPCV7—heptavalent pneumococcal conjugate vaccinePCV13—13-valent pneumococcal conjugate vaccineRD—rate differenceSNAP—safety-net antibiotic prescriptionTIV—trivalent inactivated influenza vaccineTM—tympanic membraneWASP—wait-and-see prescription

This document is copyrighted and is property of the AmericanAcademy of Pediatrics and its Board of Directors. All authorshave filed conflict of interest statements with the AmericanAcademy of Pediatrics. Any conflicts have been resolved througha process approved by the Board of Directors. The AmericanAcademy of Pediatrics has neither solicited nor accepted anycommercial involvement in the development of the content ofthis publication.

The recommendations in this report do not indicate an exclusivecourse of treatment or serve as a standard of medical care.Variations, taking into account individual circumstances, may beappropriate.

(Continued on last page)

e964 FROM THE AMERICAN ACADEMY OF PEDIATRICS

Organizational Principles to Guide and Define the ChildHealth Care System and/or Improve the Health of all Children

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 2: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Key Action Statement 1A: Cliniciansshould diagnose acute otitis media(AOM) in children who present withmoderate to severe bulging of thetympanic membrane (TM) or newonset of otorrhea not due to acuteotitis externa. Evidence Quality:Grade B. Strength: Recommendation.

Key Action Statement 1B: Cliniciansshould diagnose AOM in childrenwho present with mild bulging of theTM and recent (less than 48 hours)onset of ear pain (holding, tugging,rubbing of the ear in a nonverbalchild) or intense erythema ofthe TM. Evidence Quality: Grade C.Strength: Recommendation.

Key Action Statement 1C: Cliniciansshould not diagnose AOM in chil-dren who do not have middle eareffusion (MEE) (based on pneu-matic otoscopy and/or tympanometry).Evidence Quality: Grade B. Strength:Recommendation.

Key Action Statement 2: The man-agement of AOM should include anassessment of pain. If pain ispresent, the clinician should rec-ommend treatment to reduce pain.Evidence Quality: Grade B. Strength:Strong Recommendation.

Key Action Statement 3A: SevereAOM: The clinician should prescribeantibiotic therapy for AOM (bilateralor unilateral) in children 6 monthsand older with severe signs orsymptoms (ie, moderate or severeotalgia or otalgia for at least 48hours or temperature 39°C [102.2°F]or higher). Evidence Quality: Grade B.Strength: Strong Recommendation.

Key Action Statement 3B: Non-severe bilateral AOM in youngchildren: The clinician should pre-scribe antibiotic therapy for bi-lateral AOM in children 6 monthsthrough 23 months of age withoutsevere signs or symptoms (ie, mildotalgia for less than 48 hours and

temperature less than 39°C [102.2°F]).Evidence Quality: Grade B. Strength:Recommendation.

Key Action Statement 3C: Non-severe unilateral AOM in youngchildren: The clinician should ei-ther prescribe antibiotic therapyor offer observation with closefollow-up based on joint decision-making with the parent(s)/caregiverfor unilateral AOM in children 6months to 23 months of age withoutsevere signs or symptoms (ie, mildotalgia for less than 48 hoursand temperature less than 39°C[102.2°F]). When observation isused, a mechanism must be in placeto ensure follow-up and begin anti-biotic therapy if the child worsensor fails to improve within 48 to72 hours of onset of symptoms.Evidence Quality: Grade B. Strength:Recommendation.

Key Action Statement 3D: NonsevereAOM in older children: The clinicianshould either prescribe antibiotictherapy or offer observation withclose follow-up based on jointdecision-making with the parent(s)/caregiver for AOM (bilateral or uni-lateral) in children 24 months orolder without severe signs orsymptoms (ie, mild otalgia for lessthan 48 hours and temperature lessthan 39°C [102.2°F]). When obser-vation is used, a mechanism mustbe in place to ensure follow-up andbegin antibiotic therapy if the childworsens or fails to improve within48 to 72 hours of onset of symptoms.Evidence Quality: Grade B. Strength:Recommendation.

Key Action Statement 4A: Cliniciansshould prescribe amoxicillin forAOM when a decision to treat withantibiotics has been made and thechild has not received amoxicillin inthe past 30 days or the child doesnot have concurrent purulent con-junctivitis or the child is not allergic

to penicillin. Evidence Quality: GradeB. Strength: Recommendation.

Key Action Statement 4B: Cliniciansshould prescribe an antibiotic withadditional β-lactamase coveragefor AOM when a decision to treatwith antibiotics has been made,and the child has received amoxi-cillin in the last 30 days or hasconcurrent purulent conjunctivitis,or has a history of recurrent AOMunresponsive to amoxicillin. Evi-dence Quality: Grade C. Strength:Recommendation.

Key Action Statement 4C: Cliniciansshould reassess the patient if thecaregiver reports that the child’ssymptoms have worsened or failedto respond to the initial antibiotictreatment within 48 to 72 hoursand determine whether a changein therapy is needed. EvidenceQuality: Grade B. Strength: Recom-mendation.

Key Action Statement 5A: Cliniciansshould not prescribe prophylacticantibiotics to reduce the frequencyof episodes of AOM in children withrecurrent AOM. Evidence Quality:Grade B. Strength: Recommendation.

Key Action Statement 5B: Cliniciansmay offer tympanostomy tubes forrecurrent AOM (3 episodes in 6months or 4 episodes in 1 yearwith 1 episode in the preceding6 months). Evidence Quality: GradeB. Strength: Option.

Key Action Statement 6A: Cliniciansshould recommend pneumococcalconjugate vaccine to all childrenaccording to the schedule of theAdvisory Committee on Immuniza-tion Practices of the Centers forDisease Control and Prevention,American Academy of Pediatrics(AAP), and American Academy ofFamily Physicians (AAFP). EvidenceQuality: Grade B. Strength: StrongRecommendation.

PEDIATRICS Volume 131, Number 3, March 2013 e965

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 3: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Key Action Statement 6B: Cliniciansshould recommend annual influenzavaccine to all children according tothe schedule of the Advisory Com-mittee on Immunization Practices,AAP, and AAFP. Evidence Quality:Grade B. Strength: Recommendation.

2Key Action Statement 6C: Cliniciansshould encourage exclusive breast-feeding for at least 6 months. Evi-dence Quality: Grade B. Strength:Recommendation.

Key Action Statement 6D: Cliniciansshould encourage avoidance of to-bacco smoke exposure. EvidenceQuality: Grade C. Strength: Recom-mendation.

INTRODUCTION

In May 2004, the AAP and AAFP pub-lished the “Clinical Practice Guideline:Diagnosis and Management of AcuteOtitis Media”.1 The guideline offered8 recommendations ranked accord-ing to level of evidence and benefit-harm relationship. Three of therecommendations—diagnostic criteria,observation, and choice of antibiotics—led to significant discussion, especiallyamong experts in the field of otitis me-dia (OM). Also, at the time the guidelinewas written, information regarding theheptavalent pneumococcal conjugatevaccine (PCV7) was not yet published.Since completion of the guideline inNovember 2003 and its publication inMay 2004, there has been a significantbody of additional literature on AOM.

Although OM remains the most commoncondition for which antibacterial agentsare prescribed for children in the UnitedStates2,3 clinician visits for OM de-creased from 950 per 1000 children in1995–1996 to 634 per 1000 children in2005–2006. There has been a pro-portional decrease in antibiotic pre-scriptions for OM from 760 per 1000in 1995–1996 to 484 per 1000 in2005–2006. The percentage of OM visits

resulting in antibiotic prescriptionsremained relatively stable (80% in 1995–1996; 76% in 2005–2006).2 Many factorsmay have contributed to the decreasein visits for OM, including financialissues relating to insurance, such ascopayments, that may limit doctor visits,public education campaigns regardingthe viral nature of most infectious dis-eases, use of the PCV7 pneumococcalvaccine, and increased use of theinfluenza vaccine. Clinicians may also bemore attentive to differentiating AOMfrom OM with effusion (OME), resultingin fewer visits coded for AOM andfewer antibiotic prescriptions written.

Despite significant publicity andawareness of the 2004 AOM guideline,evidence shows that clinicians arehesitant to follow the guideline recom-mendations. Vernacchio et al4 surveyed489 primary care physicians as to theirmanagement of 4 AOM scenariosaddressed in the 2004 guideline. Nosignificant changes in practice werenoted on this survey, compared witha survey administered before the 2004AOM guideline. Coco5 used the NationalAmbulatory Medical Care Survey from2002 through 2006 to determine thefrequency of AOM visits without anti-biotics before and after publication ofthe 2004 guideline. There was no dif-ference in prescribing rates. A similarresponse to otitis guidelines was foundin Italy as in the United States.6,7

These findings parallel results of otherinvestigations regarding clinician aware-ness and adherence to guidelinerecommendations in all specialties,including pediatrics.8 Clearly, for clin-ical practice guidelines to be effective,more must be done to improve theirdissemination and implementation.

This revision and update of the AAP/AAFP2004 AOM guideline1 will evaluate pub-lished evidence on the diagnosis andmanagement of uncomplicated AOMand make recommendations based onthat evidence. The guideline is intended

for primary care clinicians includingpediatricians and family physicians,emergency department physicians,otolaryngologists, physician assistants,and nurse practitioners. The scopeof the guideline is the diagnosisand management of AOM, includingrecurrent AOM, in children 6 monthsthrough 12 years of age. It applies onlyto an otherwise healthy child withoutunderlying conditions that may alterthe natural course of AOM, includingbut not limited to the presence oftympanostomy tubes; anatomic abnor-malities, including cleft palate; geneticconditions with craniofacial abnormali-ties, such as Down syndrome; immunedeficiencies; and the presence of co-chlear implants. Children with OMEwithout AOM are also excluded.

Glossary of Terms

AOM—the rapid onset of signs andsymptoms of inflammation in themiddle ear9,10

Uncomplicated AOM—AOM withoutotorrhea1

Severe AOM—AOM with the presenceof moderate to severe otalgia or feverequal to or higher than 39°C9,10

Nonsevere AOM—AOM with thepresence of mild otalgia and a tem-perature below 39°C9,10

Recurrent AOM—3 or more well-documented and separate AOM epi-sodes in the preceding 6 months or4 or more episodes in the preceding12 months with at least 1 episode inthe past 6 months11,12

OME—inflammation of the middle earwith liquid collected in the middle ear;the signs and symptoms of acute in-fection are absent9

MEE—liquid in the middle ear withoutreference to etiology, pathogenesis,pathology, or duration9

Otorrhea—discharge from the ear,originating at 1 or more of the follow-ing sites: the external auditory canal,

e966 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 4: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

middle ear, mastoid, inner ear, or in-tracranial cavity

Otitis externa—an infection of theexternal auditory canal

Tympanometry—measuring acousticimmittance (transfer of acoustic en-ergy) of the ear as a function of earcanal air pressure13,14

Number needed to treat (NNT)—thenumber of patients who need to betreated to prevent 1 additional badoutcome15

Initial antibiotic therapy—treatmentof AOM with antibiotics that are pre-scribed at the time of diagnosis with theintent of starting antibiotic therapy assoon as possible after the encounter

Initial observation—initial manage-ment of AOM limited to symptomaticrelief, with commencement of antibiotictherapy only if the child’s conditionworsens at any time or does not showclinical improvement within 48 to 72hours of diagnosis; a mechanism mustbe in place to ensure follow-up andinitiation of antibiotics if the child failsobservation

METHODS

Guideline development using anevidence-based approach requiresthat all evidence related to theguideline is gathered in a systematicfashion, objectively assessed, and thendescribed so readers can easily seethe links between the evidence andrecommendations made. An evidence-based approach leads to recom-mendations that are guided by boththe quality of the available evidenceand the benefit-to-harm ratio thatresults from following the recom-mendation. Figure 1 shows the re-lationship of evidence quality andbenefit-harm balance in determiningthe level of recommendation. Table 1presents the AAP definitions andimplications of different levels ofevidence-based recommendations.16

In preparing for the 2004 AAP guide-lines, the Agency for Healthcare Re-search and Quality (AHRQ) funded andconducted an exhaustive review of theliterature on diagnosis and manage-ment of AOM.17–19 In 2008, the AHRQ andthe Southern California Evidence-BasedPractice Center began a similar pro-cess of reviewing the literature pub-lished since the 2001 AHRQ report. TheAAP again partnered with AHRQ andthe Southern California Evidence-BasedPractice Center to develop the evi-dence report, which served as a majorsource of data for these practiceguideline recommendations.20,21 Newkey questions were determined bya technical expert panel. The scope ofthe new report went beyond the 2001AHRQ report to include recurrent AOM.

The key questions addressed by AHRQin the 2010 report were as follows:

1. Diagnosis of AOM: What are the op-erating characteristics (sensitivity,specificity, and likelihood ratios) ofclinical symptoms and otoscopicfindings (such as bulging TM) todiagnose uncomplicated AOM andto distinguish it from OME?

2. What has been the effect of the useof heptavalent PCV7 on AOM micro-bial epidemiology, what organisms(bacterial and viral) are associatedwith AOM since the introduction ofPCV7, and what are the patterns

of antimicrobial resistance in AOMsince the introduction of PCV7?

3. What is the comparative effective-ness of various treatment optionsfor treating uncomplicated AOM inaverage risk children?

4. What is the comparative effectivenessof different management options forrecurrent OM (uncomplicated) andpersistent OM or relapse of AOM?

5. Do treatment outcomes in Ques-tions 3 and 4 differ by character-istics of the condition (AOM), patient,environment, and/or health care de-livery system?

6. What adverse effects have been ob-served for treatments for whichoutcomes are addressed in Ques-tions 3 and 4?

For the 2010 review, searches of PubMedand the Cochrane Database of System-atic Reviews, Cochrane Central Registerof Controlled Trials, and EducationResources Information Center wereconducted by using the same searchstrategies used for the 2001 report forpublications from 1998 through June2010. Additional terms or conditions notconsidered in the 2001 review (recurrentOM, new drugs, and heptavalent pneu-mococcal vaccine) were also included.The Web of Science was also used tosearch for citations of the 2001 reportand its peer-reviewed publications. Titleswere screened independently by 2

FIGURE 1Relationship of evidence quality and benefit-harm balance in determining the level of recommen-dation. RCT, randomized controlled trial.

PEDIATRICS Volume 131, Number 3, March 2013 e967

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 5: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

pediatricians with experience in con-ducting systematic reviews.

For the question pertaining to diagnosis,efficacy, and safety, the search wasprimarily for clinical trials. For thequestion pertaining to the effect of PCV7on epidemiology and microbiology, thegroup searched for trials that comparedmicrobiology in the same populationsbefore and after introduction of thevaccine or observational studies thatcompared microbiology across vacci-nated and unvaccinated populations.

In total, the reviewers examined 7646titles, of which 686 titles were identifiedfor further review. Of those, 72 articlesthat met the predetermined inclusionand exclusion criteria were reviewed indetail. Investigators abstracted datainto standard evidence tables, withaccuracy checked by a second in-vestigator. Studies were quality-ratedby 2 investigators by using estab-lished criteria. For randomized con-trolled trials, the Jadad criteria wereused.22 QUADAS criteria23 were used toevaluate the studies that pertained todiagnosis. GRADE criteria were appliedto pooled analyses.24 Data abstracted

included parameters necessary to de-fine study groups, inclusion/exclusioncriteria, influencing factors, and out-come measures. Some of the data foranalysis were abstracted by a bio-statistician and checked by a physicianreviewer. A sequential resolution strat-egy was used to match and resolve thescreening and review results of the2 pediatrician reviewers.

For the assessment of treatment effi-cacy, pooled analyses were performedfor comparisons for which 3 or moretrials could be identified. Studies eligi-ble for analyses of questions pertainingto treatment efficacy were grouped forcomparisons by treatment options. Eachcomparison consisted of studies thatwere considered homogeneous acrossclinical practice. Because some of thekey questions were addressed in the2001 evidence report,17 studies identi-fied in that report were included withnewly identified articles in the 2010evidence report.20

Decisions were made on the basis ofa systematic grading of the quality of ev-idence and strength of recommendationsas well as expert consensus when

definitive data were not available.Results of the literature review werepresented in evidence tables and pub-lished in the final evidence report.20

In June 2009, the AAP convened a newsubcommittee to review and revise theMay 2004 AOM guideline.1 The sub-committee comprised primary carephysicians and experts in the fields ofpediatrics, family practice, otolaryn-gology, epidemiology, infectious dis-ease, emergency medicine, andguideline methodology. All panelmembers reviewed the AAP policy onconflict of interest and voluntary dis-closure and were given an opportu-nity to present any potential conflictswith the subcommittee’s work. All po-tential conflicts of interest are listedat the end of this document. The projectwas funded by the AAP. New literatureon OM is continually being published.Although the systematic review per-formed by AHRQ could not be repli-cated with new literature, membersof the Subcommittee on Diagnosisand Management of Acute Otitis Mediareviewed additional articles. PubMedwas searched by using the singlesearch term “acute otitis media,”

TABLE 1 Guideline Definitions for Evidence-Based Statements

Statement Definition Implication

Strong Recommendation A strong recommendation in favor of a particular action is madewhen the anticipated benefits of the recommendedintervention clearly exceed the harms (as a strongrecommendation against an action is made when theanticipated harms clearly exceed the benefits) and the qualityof the supporting evidence is excellent. In some clearlyidentified circumstances, strong recommendations may bemade when high-quality evidence is impossible to obtain andthe anticipated benefits strongly outweigh the harms.

Clinicians should follow a strong recommendation unlessa clear and compelling rationale for an alternative approachis present.

Recommendation A recommendation in favor of a particular action is made whenthe anticipated benefits exceed the harms, but the quality ofevidence is not as strong. Again, in some clearly identifiedcircumstances, recommendations may be made when high-quality evidence is impossible to obtain but the anticipatedbenefits outweigh the harms.

Clinicians would be prudent to follow a recommendation butshould remain alert to new information and sensitive topatient preferences.

Option Options define courses that may be taken when either thequality of evidence is suspect or carefully performed studieshave shown little clear advantage to 1 approach over another.

Clinicians should consider the option in their decision-making,and patient preference may have a substantial role.

No Recommendation No recommendation indicates that there is a lack of pertinentpublished evidence and that the anticipated balance ofbenefits and harms is presently unclear.

Clinicians should be alert to new published evidence thatclarifies the balance of benefit versus harm.

e968 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 6: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

approximately every 6 months fromJune 2009 through October 2011 toobtain new articles. Subcommitteemembers evaluated pertinent articlesfor quality of methodology and im-portance of results. Selected articlesused in the AHRQ review were alsoreevaluated for their quality. Con-clusions were based on the consensusof the subcommittee after the reviewof newer literature and reevaluation ofthe AHRQ evidence. Key action state-ments were generated using BRIDGE-Wiz(Building Recommendations in a Devel-opers Guideline Editor), an interactivesoftware tool that leads guideline de-velopment through a series of questionsthat are intended to create a more ac-tionable set of key action statements.25

BRIDGE-Wiz also incorporates the qualityof available evidence into the final de-termination of the strength of eachrecommendation.

After thorough review by the sub-committee for this guideline, a draftwas reviewed by other AAP committeesand sections, selected outside organ-izations, and individuals identifiedby the subcommittee as experts inthe field. Additionally, members ofthe subcommittee were encouraged todistribute the draft to interested par-ties in their respective specialties. Allcomments were reviewed by the writ-ing group and incorporated into thefinal guideline when appropriate.

This clinical practice guideline is notintended as a sole source of guidancein the management of children withAOM. Rather, it is intended to assistclinicians in decision-making. It is notintended to replace clinical judgmentor establish a protocol for the careof all children with this condition.These recommendations may notprovide the only appropriate approachto the management of children withAOM.

It is AAP policy to review and updateevidence-based guidelines every 5 years.

KEY ACTION STATEMENTSKey Action Statement 1A

Clinicians should diagnose AOM inchildren who present with moderate

to severe bulging of the TM or newonset of otorrhea not due to acute

otitis externa. (Evidence Quality: Grade

B, Rec. Strength: Recommendation)

Key Action Statement 1B

Clinicians should diagnose AOM inchildren who present with mildbulging of the TM and recent (lessthan 48 hours) onset of ear pain

(holding, tugging, rubbing of theear in a nonverbal child) or intenseerythema of the TM. (EvidenceQuality: Grade C, Rec. Strength:Recommendation)

Key Action Statement Profile: KAS 1AAggregate evidence quality Grade B

Benefits • Identify a population of children most likely to benefit fromintervention.

• Avoid unnecessary treatment of those without highly certainAOM.

• Promote consistency in diagnosis.Risks, harms, cost May miss AOM that presents with a combination of mild bulging,

intense erythema, or otalgia that may not necessarilyrepresent less severe disease and may also benefit fromintervention.

Benefits-harms assessment Preponderance of benefit.Value judgments Identification of a population of children with highly certain AOM

is beneficial. Accurate, specific diagnosis is helpful to theindividual patient. Modification of current behavior ofoverdiagnosis is a goal. Increased specificity is preferredeven as sensitivity is lowered.

Intentional vagueness By using stringent diagnostic criteria, the TM appearance of lesssevere illness that might be early AOM has not beenaddressed.

Role of patient preferences NoneExclusions NoneStrength RecommendationNotes Tympanocentesis studies confirm that using these diagnostic

findings leads to high levels of isolation of pathogenicbacteria. Evidence is extrapolated from treatment studiesthat included tympanocentesis.

Key Action Statement Profile: KAS 1BAggregate evidence quality Grade C

Benefits Identify AOM in children when the diagnosis is not highlycertain.

Risks, harms, cost Overdiagnosis of AOM. Reduced precision in diagnosis.Benefits-harms assessment Benefits greater than harms.Value judgments None.Intentional vagueness Criteria may be more subjective.Role of patient preferences NoneExclusions NoneStrength RecommendationNotes Recent onset of ear pain means within the past 48 hours.

PEDIATRICS Volume 131, Number 3, March 2013 e969

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 7: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Key Action Statement 1C

Clinicians should not diagnose AOM inchildren who do not have MEE (based

on pneumatic otoscopy and/or tym-panometry). (Evidence Quality: Grade

B, Rec. Strength: Recommendation)

Purpose of This Section

There is no gold standard for the di-agnosis of AOM. In fact, AOM hasa spectrum of signs as the diseasedevelops.26 Therefore, the purpose ofthis section is to provide cliniciansand researchers with a working clin-ical definition of AOM and to differ-entiate AOM from OME. The criteriawere chosen to achieve high specific-ity recognizing that the resulting de-creased sensitivity may exclude lesssevere presentations of AOM.

Changes From AAP/AAFP 2004 AOMGuideline

Accurate diagnosis of AOM is critical tosound clinical decision-making andhigh-quality research. The 2004 “Clin-ical Practice Guideline: Diagnosis andManagement of AOM”1 used a 3-partdefinition for AOM: (1) acute onset ofsymptoms, (2) presence of MEE, and(3) signs of acute middle ear in-flammation. This definition generatedextensive discussion and reanalysis ofthe AOM diagnostic evidence. The 2004definition lacked precision to excludecases of OME, and diagnoses of AOM

could be made in children with acuteonset of symptoms, including severeotalgia and MEE, without other otoscopicfindings of inflammation.27 Further-more, the use of “uncertain dia-gnosis” in the 2004 AOM guideline mayhave permitted diagnoses of AOMwithout clear visualization of the TM.Earlier studies may have enrolledchildren who had OME rather thanAOM, resulting in the possible classi-fication of such children as improvedbecause their nonspecific symptomswould have abated regardless oftherapy.28–30 Two studies, published in2011, used stringent diagnostic crite-ria for diagnosing AOM with muchless risk of conclusions based on datafrom mixed patients.31,32

Since publication of the 2004 AOMguideline, a number of studies havebeen conducted evaluating scales forthe presence of symptoms. Thesestudies did not show a consistentcorrelation of symptoms with the ini-tial diagnosis of AOM, especially inpreverbal children.33–35

Recent research has used preciselystated stringent criteria of AOM for

purposes of the studies.31,32 The currentguideline endorses stringent otoscopicdiagnostic criteria as a basis for man-agement decisions (described later). Asclinicians use the proposed stringentcriteria to diagnose AOM, they shouldbe aware that children with AOM mayalso present with recent onset of earpain and intense erythema of the TMas the only otoscopic finding.

Symptoms

Older children with AOM usuallypresent with a history of rapid onset ofear pain. However, in young preverbalchildren, otalgia as suggested bytugging/rubbing/holding of the ear,excessive crying, fever, or changes inthe child’s sleep or behavior patternas noted by the parent are often rel-atively nonspecific symptoms. A num-ber of studies have attempted tocorrelate symptom scores with di-agnoses of AOM.

A systematic review36 identified 4articles that evaluated the accuracyof symptoms.37–40 Ear pain appeareduseful in diagnosing AOM (combinedpositive likelihood ratio 3.0–7.3, nega-tive likelihood ratio 0.4–0.6); however,it was only present in 50% to 60% ofchildren with AOM. Conclusions fromthese studies may be limited, becausethey (1) enrolled children seen byspecialists, not likely to represent thewhole spectrum of severity of illness;(2) used a clinical diagnosis of AOMbased more on symptomatology ratherthan on tympanocentesis; and (3) in-cluded relatively older children.37,40

Laine et al34 used a questionnaireadministered to 469 parents whosuspected their children, aged 6 to 35months, had AOM. Of the children, 237had AOM using strict otoscopic crite-ria, and 232 had upper respiratorytract infection without AOM. Restlesssleep, ear rubbing, fever, and non-specific respiratory or gastrointestinal

Key Action Statement Profile: KAS 1CAggregate evidence quality Grade B

Benefits Reduces overdiagnosis and unnecessary treatment. Increasescorrect diagnosis of other conditions with symptoms thatotherwise might be attributed to AOM. Promotes the use ofpneumatic otoscopy and tympanometry to improvediagnostic accuracy.

Risks, harms, cost Cost of tympanometry. Need to acquire or reacquire skills inpneumatic otoscopy and tympanometry for some clinicians.

Benefits-harms assessment Preponderance of benefit.Value judgments AOM is overdiagnosed, often without adequate visualization of

the TM. Early AOM without effusion occurs, but the risk ofoverdiagnosis supersedes that concern.

Intentional vagueness NoneRole of patient preferences NoneExclusions Early AOM evidenced by intense erythema of the TM.Strength Recommendation

e970 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 8: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

tract symptoms did not differentiatechildren with or without AOM.

McCormick et al30 used 2 symptomscores—a 3-item score (OM-3), con-sisting of symptoms of physical suffer-ing such as ear pain or fever, emotionaldistress (irritability, poor appetite), andlimitation in activity; and a 5-item score(Ear Treatment Group Symptom Ques-tionnaire, 5 Items [ETG-5]), includingfever, earache, irritability, decreasedappetite, and sleep disturbance—toassess AOM symptoms at the time ofdiagnosis and daily during the 10-daytreatment or observation period. Theyfound both to be a responsive measureof changes in clinical symptoms. Thesame group35 also tested a visual scale,Acute Otitis Media-Faces Scale (AOM-FS),with faces similar to the Wong-Bakerpain scale.41 None of the scales wereadequately sensitive for making the di-agnosis of AOM based on symptoms. TheAOM-FS combined with an otoscopy score,OS-8,30 were presented as a double-sidedpocket card. The combination of AOM-FSand OS-8 was more responsive to changethan either instrument alone.

Shaikh et al33,42 validated a 7-itemparent-reported symptom score (AcuteOtitis Media Severity of Symptom Scale[AOM-SOS]) for children with AOM, fol-lowing stringent guidance of the USFood and Drug Administration (FDA)on the development of patient-reportedoutcome scales. Symptoms includedear tugging/rubbing/holding, excessivecrying, irritability, difficulty sleeping,decreased activity or appetite, andfever. AOM-SOS was correlated withotoscopic diagnoses (AOM, OME, andnormal middle ear status). AOM-SOSchanged appropriately in response toclinical change. Its day-to-day re-sponsiveness supports its usefulness infollowing AOM symptoms over time.

Signs of AOM

Few studies have evaluated the re-lationship of otoscopic findings in AOM

and tympanocentesis. A study byKarma et al43 is often cited as the bestsingle study of otoscopic findings inAOM. However, the study uses onlya symptom-based diagnosis of AOMplus the presence of MEE. Thus, chil-dren with acute upper respiratorytract infection symptoms and OMEwould have been considered to haveAOM. There also were significant dif-ferences in findings at the 2 centersthat participated in the study.

The investigators correlated TM color,mobility, and position with the pres-ence of middle ear fluid obtained bytympanocentesis. At 2 sites in Finland(Tampere and Oulu), 2911 childrenwere followed from 6 months to 2.5years of age. A single otolaryngologistat Tampere and a single pediatrician atOulu examined subjects. Color, posi-tion, and mobility were recorded.Myringotomy and aspiration wereperformed if MEE was suspected.AOM was diagnosed if MEE was foundand the child had fever, earache, irri-tability, ear rubbing or tugging, si-multaneous other acute respiratorytract symptoms, vomiting, or di-arrhea. The presence or absence ofMEE was noted, but no analyses ofthe fluid, including culture, were per-formed. Pneumatic otoscopic findingswere classified as follows: color—hemorrhagic, strongly red, moderatelyred, cloudy or dull, slightly red, or nor-mal; position—bulging, retracted, ornormal; and mobility—distinctly im-paired, slightly impaired, or normal.

For this analysis, 11 804 visits wereavailable. For visits with acute symp-toms, MEE was found in 84.9% and81.8% at the 2 sites at which the studywas performed. There were signifi-cant differences among the results atthe 2 centers involved in the study.Table 2 shows specific data for eachfinding.

The combination of a “cloudy,” bulgingTM with impaired mobility was the

best predictor of AOM using thesymptom-based diagnosis in this study.Impaired mobility had the highest sen-sitivity and specificity (approximately95% and 85%, respectively). Cloudi-ness had the next best combination ofhigh sensitivity (∼74%) and highspecificity (∼93%) in this study. Bulg-ing had high specificity (∼97%) butlower sensitivity (∼51%). A TM thatwas hemorrhagic, strongly red, ormoderately red also correlated withthe presence of AOM, and a TM thatwas only “slightly red” was not helpfuldiagnostically.

McCormick et al reported that a bulg-ing TM was highly associated with thepresence of a bacterial pathogen, withor without a concomitant viral patho-gen.44 In a small study, 31 children(40 ears) underwent myringotomy.45

Bulging TMs had positive bacterialcultures 75% of the time. Thepercentage of positive cultures fora pathogen increased to 80% if thecolor of the TM was yellow. The con-clusion is that moderate to severebulging of the TM represents the mostimportant characteristic in the di-agnosis of AOM—a finding that has

TABLE 2 Otoscopic Findings in Children WithAcute Symptoms and MEEa

TM Finding inAcute VisitsWith MEE

Group I(Tampere,Finland), %

Group II(Oulo,

Finland), %

ColorDistinctly red 69.8 65.6Hemorrhagic 81.3 62.9Strongly red 87.7 68.1Moderately red 59.8 66.0Slightly red 39.4 16.7Cloudy 95.7 80.0Normal 1.7 4.9

PositionBulging 96.0 89Retracted 46.8 48.6Normal 32.1 22.2

MobilityDistinctly impaired 94.0 78.5Slightly impaired 59.7 32.8Normal 2.7 4.8

a Totals are greater than 100%, because each ear mayhave had different findings.43

PEDIATRICS Volume 131, Number 3, March 2013 e971

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 9: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

implications for clinical care, re-search, and education.

The committee recognized that there isa progression from the presence ofMEE to the bulging of the TM, and itis often difficult to differentiate thisequivocal appearance from the highlycertain AOM criteria advocated in thisguideline.26 As such, there is a role forindividualized diagnosis and manage-ment decisions. Examples of normal,mild bulging, moderate bulging, andsevere bulging can be seen in Fig 2.

Distinguishing AOM From OME

OME may occur either as the aftermathof an episode of AOM or as a conse-quence of eustachian tube dysfunctionattributable to an upper respiratorytract infection.46 However, OME mayalso precede and predispose to thedevelopment of AOM. These 2 forms ofOM may be considered segments ofa disease continuum.47 However, be-cause OME does not represent anacute infectious process that benefitsfrom antibiotics, it is of utmost im-portance for clinicians to becomeproficient in distinguishing normalmiddle ear status from OME or AOM.Doing so will avoid unnecessary useof antibiotics, which leads to in-creased adverse effects of medicationand facilitates the development ofantimicrobial resistance.

Examination of the TM

Accurate diagnosis of AOM in infantsand young children may be difficult.

Symptoms may be mild or overlap withthose of an upper respiratory tractillness. The TM may be obscured bycerumen, and subtle changes in the TMmay be difficult to discern. Additionalfactors complicating diagnosis mayinclude lack of cooperation from thechild; less than optimal diagnosticequipment, including lack of a pneu-matic bulb; inadequate instrumentsfor clearing cerumen from the externalauditory canal; inadequate assistancefor restraining the child; and lack ofexperience in removing cerumen andperforming pneumatic otoscopy.

The pneumatic otoscope is the stan-dard tool used in diagnosing OM.Valuable also is a surgical head, whichgreatly facilitates cleaning cerumenfrom an infant’s external auditorycanal. Cerumen may be removed byusing a curette, gentle suction, or ir-rigation.48 The pneumatic otoscopeshould have a light source of suffi-cient brightness and an air-tight sealthat permits application of positiveand negative pressure. In general,nondisposable specula achieve a bet-ter seal with less pain because ofa thicker, smoother edge and betterlight transmission properties. Thespeculum size should be chosen togently seal at the outer portion of theexternal auditory canal.

Pneumatic otoscopy permits assess-ment of the contour of the TM (normal,retracted, full, bulging), its color(gray, yellow, pink, amber, white, red,blue), its translucency (translucent,

semiopaque, opaque), and its mobility(normal, increased, decreased, ab-sent). The normal TM is translucent,pearly gray, and has a ground-glassappearance (Fig 2A). Specific land-marks can be visualized. They includethe short process and the manubriumof the malleus and the pars flaccida,located superiorly. These are easilyobserved and help to identify the po-sition of the TM. Inward movement ofthe TM on positive pressure in theexternal canal and outward move-ment on negative pressure shouldoccur, especially in the superior pos-terior quadrant. When the TM isretracted, the short process of themalleus becomes more prominent,and the manubrium appears short-ened because of its change in positionwithin the middle ear. Inward motionoccurring with positive pressure isrestricted or absent, because theTM is frequently as far inward asits range of motion allows. However,outward mobility can be visualizedwhen negative pressure is applied. Ifthe TM does not move perceptibly withapplications of gentle positive ornegative pressure, MEE is likely.Sometimes, the application of pres-sure will make an air-fluid interfacebehind the TM (which is diagnostic ofMEE) more evident.49

Instruction in the proper evaluation ofthe child’s middle ear status shouldbegin with the first pediatric rotationin medical school and continuethroughout postgraduate training.50

FIGURE 2A, Normal TM. B, TM with mild bulging. C, TM with moderate bulging. D, TM with severe bulging. Courtesy of Alejandro Hoberman, MD.

e972 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 10: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Continuing medical education shouldreinforce the importance of, and re-train the clinician in, the use ofpneumatic otoscopy.51 Training toolsinclude the use of a video-otoscope inresidency programs, the use of Web-based educational resources,49,52 aswell as simultaneous or sequentialexamination of TMs with an expertotoscopist to validate findings by usinga double headed or video otoscope.Tools for learning the ear examinationcan be found in a CD distributed by theJohns Hopkins University School ofMedicine and the Institute for Johns

Hopkins Nursing,53 also available athttp://www2.aap.org/sections/infectdis/video.cfm,54 and through a Web-basedprogram, ePROM: Enhancing Proficiencyin Otitis Media.52

Key Action Statement 2

The management of AOM shouldinclude an assessment of pain. Ifpain is present, the clinicianshould recommend treatment toreduce pain. (Evidence Quality:Grade B, Rec. Strength: StrongRecommendation)

Purpose of This Section

Pain is the major symptom of AOM. Thissection addresses and updates theliterature on treating otalgia.

Changes From AAP/AAFP 2004 AOMGuideline

Only 2 new articles directly addressthe treatment of otalgia. Both addresstopical treatment. The 2 new articlesare consistent with the 2004 guidelinestatement. The text of the 2004 guidelineis, therefore, reproduced here, with theaddition of discussion of the 2 newarticles. Table 3 has been updated toinclude the new references.

Treatment of Otalgia

Many episodes of AOM are associatedwith pain.55 Some children with OMEalso have ear pain. Although pain is

a common symptom in these ill-nesses, clinicians often see otalgia asa peripheral concern not requiringdirect attention.56 Pain associated

with AOM can be substantial in thefirst few days of illness and oftenpersists longer in young children.57

Antibiotic therapy of AOM does notprovide symptomatic relief in the first24 hours58–61 and even after 3 to 7days, there may be persistent pain,fever, or both in 30% of childrenyounger than 2 years.62 In contrast,analgesics do relieve pain associatedwith AOM within 24 hours63 andshould be used whether antibiotictherapy is or is not prescribed; theyshould be continued as long asneeded. The AAP published the policystatement “The Assessment andManagement of Acute Pain in Infants,Children, and Adolescents”64 to assistthe clinician in addressing pain in thecontext of illness. The management ofpain, especially during the first 24hours of an episode of AOM, should beaddressed regardless of the use ofantibiotics.

Various treatments of otalgia havebeen used, but none has been wellstudied. The clinician should selecta treatment on the basis of a consid-eration of benefits and risks and,wherever possible, incorporateparent/caregiver and patient prefer-ence (Table 3).

Key Action Statement Profile: KAS 2Aggregate evidence quality Grade B

Benefits Relieves the major symptom of AOM.Risks, harms, cost Potential medication adverse effects. Variable efficacy of some

modes of treatment.Benefits-harms assessment Preponderance of benefit.Value judgments Treating pain is essential whether or not antibiotics are

prescribed.Intentional vagueness Choice of analgesic is not specified.Role of patient preferences Parents may assist in the decision as to what means of pain

relief they prefer.Exclusions Topical analgesics in the presence of a perforated TM.Strength Strong Recommendation

TABLE 3 Treatments for Otalgia in AOM

Treatment Modality Comments

Acetaminophen, ibuprofen63 Effective analgesia for mild to moderate pain.Readily available. Mainstay of pain managementfor AOM.

Home remedies (no controlled studiesthat directly address effectiveness)

May have limited effectiveness.

DistractionExternal application of heat or coldOil drops in external auditory canal

Topical agentsBenzocaine, procaine, lidocaine65,67,70 Additional, but brief, benefit over acetaminophen

in patients older than 5 y.Naturopathic agents68 Comparable to amethocaine/phenazone drops in

patients older than 6 y.Homeopathic agents71,72 No controlled studies that directly address pain.Narcotic analgesia with codeine

or analogsEffective for moderate or severe pain. Requires

prescription; risk of respiratory depression, alteredmental status, gastrointestinal tract upset, andconstipation.

Tympanostomy/myringotomy73 Requires skill and entails potential risk.

PEDIATRICS Volume 131, Number 3, March 2013 e973

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 11: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Since the 2004 guideline was pub-lished, there have been only 2 signifi-cant new articles.

Bolt et al reported in 2008 on a double-blind placebo-controlled trial at theAustralia Children’s Hospital emer-gency department conducted in2003–2004.65 They used a conveniencesample of children 3 to 17 years ofage diagnosed with AOM in the ED.They excluded children with perfora-tion of the TM, pressure-equalizingtube, allergy to local anesthetic orparacetamol, epilepsy, or liver, renal,or cardiac disease. Sixty-three eligiblechildren were randomized to receiveaqueous lidocaine or normal salineear drops up to 3 times in 24 hours.They demonstrated a statistically sig-nificant 50% reduction in reportedpain at 10 and 30 minutes but not at20 minutes after application of topicallidocaine, compared with normal sa-line. Complications were minimal: 3children reported some dizziness thenext day, and none reported tinnitus.A limitation was that some childrenhad received oral acetaminophen be-fore administration of ear drops.

A Cochrane review of topical analgesiafor AOM66 searched the Cochraneregister of controlled trials, random-ized controlled trials, or quasi-randomized controlled trials thatcompared otic preparations to pla-cebo or that compared 2 otic prepa-rations. It included studies of adultsand children, without TM perforation.

It identified 5 trials in children 3 to18 years of age. Two (including Boltet al,65 discussed above) comparedanesthetic drops and placebo at di-agnosis of AOM. In both studies, somechildren also received oral analgesics.Three studies compared anestheticear drops with naturopathic herbaldrops. Naturopathic drops were fa-vored 15 to 30 minutes afterinstallation, and 1 to 3 days afterdiagnosis, but the difference was notstatistically significant. The Cochranegroup concluded that there is limitedevidence that ear drops are effectiveat 30 minutes and unclear if resultsfrom these studies are a result of thenatural course of illness, placebo ef-fect of receiving treatment, soothingeffect of any liquid in the ear, or thedrops themselves. Three of the stud-ies included in this review were citedin the 2004 AAP guideline67–69 and the1 new paper by Bolt et al.65

Key Action Statement 3A

Severe AOM

The clinician should prescribe an-tibiotic therapy for AOM (bilateralor unilateral) in children 6 monthsand older with severe signs orsymptoms (ie, moderate or severeotalgia or otalgia for at least 48hours, or temperature 39°C[102.2°F] or higher). (EvidenceQuality: Grade B, Rec. Strength:Strong Recommendation)

Key Action Statement 3B

Nonsevere Bilateral AOM in YoungChildren

The clinician should prescribe an-tibiotic therapy for bilateral AOM inchildren younger than 24 monthswithout severe signs or symptoms(ie, mild otalgia for less than 48hours, temperature less than 39°C[102.2°F]). (Evidence Quality: GradeB, Rec. Strength: Recommendation)

Key Action Statement 3C

Nonsevere Unilateral AOM in YoungChildren

The clinician should either prescribeantibiotic therapy or offer obser-vation with close follow-up basedon joint decision-making with theparent(s)/caregiver for unilateralAOM in children 6 months to 23months of age without severesigns or symptoms (ie, mild otalgiafor less than 48 hours, tempera-ture less than 39°C [102.2°F]).When observation is used, a mech-anism must be in place to ensure

Key Action Statement Profile: KAS 3AAggregate evidence quality Grade B

Benefits Increased likelihood of more rapid resolution of symptoms.Increased likelihood of resolution of AOM.

Risks, harms, cost Adverse events attributable to antibiotics, such as diarrhea,diaper dermatitis, and allergic reactions. Overuse ofantibiotics leads to increased bacterial resistance. Cost ofantibiotics.

Benefits-harms assessment Preponderance of benefit over harm.Value judgments NoneRole of patient preference NoneIntentional vagueness NoneExclusions NoneStrength Strong Recommendation

Key Action Statement Profile: KAS3BAggregate evidencequality

Grade B

Benefits Increased likelihood of morerapid resolution of symptoms.Increased likelihood ofresolution of AOM.

Risks, harms,cost

Adverse events attributable toantibiotics, such as diarrhea,diaper dermatitis, andallergic reactions. Overuseof antibiotics leads toincreased bacterial resistance.Cost of antibiotics.

Benefits-harmsassessment

Preponderance of benefit overharm.

Value judgments NoneRole of patientpreference

None

Intentionalvagueness

None

Exclusions NoneStrength Recommendation

e974 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 12: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

follow-up and begin antibiotic ther-apy if the child worsens or fails toimprove within 48 to 72 hours of

onset of symptoms. (Evidence Qual-ity: Grade B, Rec. Strength: Recom-mendation)

Key Action Statement 3D

Nonsevere AOM in Older Children

The clinician should either pre-scribe antibiotic therapy or offerobservation with close follow-upbased on joint decision-making withthe parent(s)/caregiver for AOM(bilateral or unilateral) in children24 months or older without severesigns or symptoms (ie, mild otalgia

for less than 48 hours, tempera-ture less than 39°C [102.2°F]).When observation is used, a mecha-nism must be in place to ensurefollow-up and begin antibiotic ther-apy if the child worsens or failsto improve within 48 to 72 hoursof onset of symptoms. (EvidenceQuality: Grade B, Rec Strength:Recommendation)

Purpose of This Section

The purpose of this section is to offerguidance on the initial management ofAOM by helping clinicians choose be-tween the following 2 strategies:

1. Initial antibiotic therapy, defined astreatment of AOM with antibioticsthat are prescribed at the time ofdiagnosis with the intent of start-ing antibiotic therapy as soon aspossible after the encounter.

2. Initial observation, defined as ini-tial management of AOM limitedto symptomatic relief, with com-mencement of antibiotic therapyonly if the child’s condition wors-ens at any time or does not showclinical improvement within 48 to72 hours of diagnosis. A mecha-nism must be in place to ensurefollow-up and initiation of antibiot-ics if the child fails observation.

This section assumes that the clinicianhas made an accurate diagnosis ofAOM by using the criteria and strate-gies outlined earlier in this guideline.Another assumption is that a cleardistinction is made between the role ofanalgesics and antibiotics in providingsymptomatic relief for children withAOM.

Changes From Previous AOMGuideline

The AOM guideline published by theAAP and AAFP in 2004 proposed, for thefirst time in North America, an “ob-servation option” for selected childrenwith AOM, building on successfulimplementation of a similar policy inthe state of New York74 and the use ofa similar paradigm in many countriesin Europe. A common feature of bothapproaches was to prioritize initialantibiotic therapy according to di-agnostic certainty, with greaterreliance on observation when the di-agnosis was uncertain. In response tocriticism that allowing an “uncertain

Key Action Statement Profile: KAS 3CAggregate evidence quality Grade B

Benefits Moderately increased likelihood of more rapid resolution of symptomswith initial antibiotics. Moderately increased likelihood of resolutionof AOM with initial antibiotics.

Risks, harms, cost Adverse events attributable to antibiotics, such as diarrhea, diaperdermatitis, and allergic reactions. Overuse of antibiotics leads toincreased bacterial resistance. Cost of antibiotics.

Benefits-harms assessment Moderate degree of benefit over harm.Value judgments Observation becomes an alternative as the benefits and harms

approach balance.Role of patient preference Joint decision-making with the family is essential before choosing

observation.Intentional vagueness Joint decision-making is highly variable from family to familyExclusions NoneStrength RecommendationNote In the judgment of 1 Subcommittee member (AH), antimicrobial

treatment of these children is preferred because of a preponderanceof benefit over harm. AH did not endorse Key Action Statement 3C

Key Action Statement Profile: KAS 3DAggregate evidence quality Grade B

Benefits Initial antibiotic treatment: Slightly increased likelihood of morerapid resolution of symptoms; slightly increased likelihood ofresolution of AOM. Initial observation: Decreased use of antibiotics;decreased adverse effects of antibiotics; decreased potential fordevelopment of bacterial resistance.

Risks, harms, cost Initial antibiotic treatment: Adverse events attributable to antibioticssuch as diarrhea, rashes, and allergic reactions. Overuse ofantibiotics leads to increased bacterial resistance. Initialobservation: Possibility of needing to start antibiotics in 48 to 72 hif the patient continues to have symptoms. Minimal risk of adverseconsequences of delayed antibiotic treatment. Potential increasedphone calls and doctor visits.

Benefits-harms assessment Slight degree of benefit of initial antibiotics over harm.Value judgments Observation is an option as the benefits and harms approach balance.Role of patient preference Joint decision-making with the family is essential before choosing

observation.Intentional vagueness Joint decision-making is highly variable from family to family.Exclusions NoneStrength Recommendation.

PEDIATRICS Volume 131, Number 3, March 2013 e975

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 13: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

diagnosis” might condone incompletevisualization of the TM or allow in-appropriate antibiotic use, this cate-gory has been eliminated with greateremphasis now placed on maximizingdiagnostic accuracy for AOM.

Since the earlier AOM guideline waspublished, there has been substantialnew research on initial managementof AOM, including randomized con-trolled trials of antibiotic therapyversus placebo or no therapy,31,32,75

immediate versus delayed antibiotictherapy,30,76,77 or delayed antibioticwith or without a concurrent pre-scription.78 The Hoberman and Tähtinenarticles are especially important asthey used stringent criteria for di-agnosing AOM.31,32 Systematic reviewshave been published on delayed anti-biotic therapy,79 the natural history ofAOM in untreated children,57 pre-dictive factors for antibiotic benefits,62

and the effect of antibiotics onasymptomatic MEE after therapy.80

Observational studies provide addi-tional data on outcomes of initial ob-servation with delayed antibiotictherapy, if needed,81 and on the re-lationship of previous antibiotic ther-apy for AOM to subsequent acutemastoiditis.82,83

In contrast to the earlier AOM guide-line,1 which recommended antibiotictherapy for all children 6 months to 2years of age with a certain diagnosis,

the current guideline indicatesa choice between initial antibiotictherapy or initial observation in thisage group for children with unilat-eral AOM and mild symptoms butonly after joint decision-making withthe parent(s)/caregiver (Table 4).This change is supported by evidenceon the safety of observation ordelayed prescribing in young chil-dren.30,31,32,75,76,81 A mechanism mustbe in place to ensure follow-up andbegin antibiotics if the child failsobservation.

Importance of Accurate Diagnosis

The recommendations for manage-ment of AOM assume an accuratediagnosis on the basis of criteriaoutlined in the diagnosis section of thisguideline. Many of the studies sincethe 2004 AAP/AAFP AOM guideline1

used more stringent and well-definedAOM diagnostic definitions than werepreviously used. Bulging of the TMwas required for diagnosis of AOM formost of the children enrolled in themost recent studies.31,32 By using thecriteria in this guideline, clinicianswill more accurately distinguish AOMfrom OME. The management of OMEcan be found in guidelines written bythe AAP, AAFP, and American Academyof Otolaryngology-Head and NeckSurgery.84,85

Age, Severity of Symptoms,Otorrhea, and Laterality

Rovers et al62 performed a systematicsearch for AOM trials that (1) usedrandom allocation of children, (2) in-cluded children 0 to 12 years of agewith AOM, (3) compared antibioticswith placebo or no treatment, and (4)had pain or fever as an outcome. Theoriginal investigators were asked fortheir original data.

Primary outcome was pain and/orfever (>38°C) at 3 to 7 days. The ad-verse effects of antibiotics were alsoanalyzed. Baseline predictors wereage <2 years versus ≥2 years, bi-lateral AOM versus unilateral AOM,and the presence versus absence ofotorrhea. Statistical methods wereused to assess heterogeneity and toanalyze the data.

Of the 10 eligible studies, the inves-tigators of 6 studies30,75,86–89 providedthe original data requested, and 4 didnot. A total of 1642 patients were in-cluded in the 6 studies from whichdata were obtained. Of the casessubmitted, the average age was 3 to 4years, with 35% of children youngerthan 2 years. Bilateral AOM waspresent in 34% of children, and 42% ofchildren had a bulging TM. Otorrheawas present in 21% of children. Theantibiotic and control groups werecomparable for all characteristics.

The rate difference (RD) for pain, fever,or both between antibiotic and controlgroups was 13% (NNT = 8). For chil-dren younger than 2 years, the RD was15% (NNT = 7); for those ≥2 years, RDwas 11% (NNT = 10). For unilateralAOM, the RD was 6% (NNT = 17); forbilateral AOM, the RD was 20% (NNT =5). When unilateral AOM was brokeninto age groups, among those youngerthan 2 years, the RD was 5% (NNT =20), and among those ≥2 years, theRD was 7% (NNT = 15). For bilateralAOM in children younger than 2 years,the RD was 25% (NNT = 4); for

TABLE 4 Recommendations for Initial Management for Uncomplicated AOMa

Age OtorrheaWithAOMa

Unilateral orBilateral AOMa

With SevereSymptomsb

Bilateral AOMa

Without OtorrheaUnilateral AOMa

Without Otorrhea

6 mo to 2 y Antibiotictherapy

Antibiotictherapy

Antibiotic therapy Antibiotic therapy oradditional observation

≥2 y Antibiotictherapy

Antibiotictherapy

Antibiotic therapy oradditional observation

Antibiotic therapy oradditional observationc

a Applies only to children with well-documented AOM with high certainty of diagnosis (see Diagnosis section).b A toxic-appearing child, persistent otalgia more than 48 h, temperature ≥39°C (102.2°F) in the past 48 h, or if there isuncertain access to follow-up after the visit.c This plan of initial management provides an opportunity for shared decision-making with the child’s family for thosecategories appropriate for additional observation. If observation is offered, a mechanism must be in place to ensurefollow-up and begin antibiotics if the child worsens or fails to improve within 48 to 72 h of AOM onset.

e976 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 14: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

bilateral AOM in children ≥2 years,the RD was 12% (NNT = 9). Forotorrhea, the RD was 36% (NNT = 3).One child in the control group whodeveloped meningitis had receivedantibiotics beginning on day 2 be-cause of worsening status. Therewere no cases of mastoiditis.

In a Cochrane Review, Sanders et al59

identified 10 studies that met the fol-lowing criteria: (1) randomized con-trolled trial, (2) compared antibioticversus placebo or antibiotic versusobservation, (3) age 1 month to 15years, (4) reported severity and dura-tion of pain, (5) reported adverseevents, and (6) reported serious com-plications of AOM, recurrent attacks,and hearing problems. Studies wereanalyzed for risk of bias and assess-ment of heterogeneity. The studieswere the same as analyzed by Roverset al62 but included the 4 studies forwhich primary data were not availableto Rovers.60,61,90,91

The authors’ conclusions were thatantibiotics produced a small re-duction in the number of children withpain 2 to 7 days after diagnosis. Theyalso concluded that most casesspontaneously remitted with no com-plications (NNT = 16). Antibiotics weremost beneficial in children youngerthan 2 years with bilateral AOM and inchildren with otorrhea.

Two recent studies only includedchildren younger than 3 years32 oryounger than 2 years.31 Both includedonly subjects in whom the diagnosisof AOM was certain. Both studies usedimprovement of symptoms and im-provement in the appearance of theTM in their definitions of clinical suc-cess or failure.

Hoberman et al31 conducted a random-ized, double-blind, placebo-controlledstudy of the efficacy of antimicrobialtreatment on AOM. The criteria forAOM were acute symptoms witha score of at least 3 on the AOM-SOS,

a validated symptom scale33,92; MEE;and moderate or marked bulging ofthe TM or slight bulging accompaniedby either otalgia or marked erythemaof the TM. They chose to use high-dose amoxicillin-clavulanate (90 mg/kg/day) as active treatment, because ithas the best oral antibiotic coveragefor organisms causing AOM. Includedin the study were 291 patients 6 to 23months of age: 144 in the antibioticgroup and 147 in the placebo group.The primary outcome measures werethe time to resolution of symptomsand the symptom burden over time.The initial resolution of symptoms (ie,the first recording of an AOM-SOSscore of 0 or 1) was recordedamong the children who receivedamoxicillin-clavulanate in 35% by day2, 61% by day 4, and 80% by day 7.Among children who received placebo,an AOM-SOS score of 0 or 1 wasrecorded in 28% by day 2, 54% by day4, and 74% by day 7 (P = .14 for theoverall comparison). For sustainedresolution of symptoms (ie, the timeto the second of 2 successiverecordings of an AOM-SOS score of0 or 1), the corresponding valueswere 20% at day 2, 41% at day 4, and67% at day 7 with amoxicillin-clavulanate, compared with 14%,36%, and 53% with placebo (P = .04for the overall comparison). Thesymptom burden (ie, mean AOM-SOSscores) over the first 7 days werelower for the children treated withamoxicillin-clavulanate than for thosewho received placebo (P = .02). Clini-cal failure at or before the 4- to 5-dayvisit was defined as “either a lack ofsubstantial improvement in symp-toms, a worsening of signs on oto-scopic examination, or both,” andclinical failure at the 10- to 12-day visitwas defined as “the failure to achievecomplete or nearly complete resolu-tion of symptoms and of otoscopicsigns, without regard to the persis-tence or resolution of middle ear

effusion.” Treatment failure occurred byday 4 to 5 in 4% of the antimicrobialtreatment group versus 23% in theplacebo group (P < .001) and at day10 to 12 in 16% of the antimicrobialtreatment group versus 51% in theplacebo group (NNT = 2.9, P < .001). Ina comparison of outcome in unilateralversus bilateral AOM, clinical failurerates by day 10 to 12 in children withunilateral AOM were 9% in thosetreated with amoxicillin-clavulanateversus 41% in those treated withplacebo (RD, 32%; NNT = 3) and 23%vs 60% (RD, 37%; NNT = 3) in thosewith bilateral AOM. Most common ad-verse events were diarrhea (25% vs15% in the treatment versus placebogroups, respectively; P = .05) and di-aper dermatitis (51% vs 35% in thetreatment versus placebo groups,respectively; P = .008). One placeborecipient developed mastoiditis. Ac-cording to these results, antimicrobialtreatment of AOM was more beneficialthan in previous studies that usedless stringent diagnostic criteria.

Tähtinen et al32 conducted a random-ized, double-blind, placebo-controlled,intention-to-treat study of amoxicillin-clavulanate (40 mg/kg/day) versusplacebo. Three hundred nineteenpatients from 6 to 35 months of agewere studied: 161 in the antibioticgroup and 158 in the placebo group.AOM definition was the presence ofMEE, distinct erythema over a bulgingor yellow TM, and acute symptomssuch as ear pain, fever, or respiratorysymptoms. Compliance was measuredby using daily patient diaries andnumber of capsules remaining at theend of the study. Primary outcomewas time to treatment failure de-fined as a composite of 6 indepen-dent components: no improvement inoverall condition by day 3, worseningof the child’s condition at any time, noimprovement in otoscopic signs byday 8, perforation of the TM,

PEDIATRICS Volume 131, Number 3, March 2013 e977

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 15: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

development of severe infection (eg,pneumonia, mastoiditis), and any otherreason for stopping the study drug/placebo.

Groups were comparable on multipleparameters. In the treatment group,135 of 161 patients (84%) were youn-ger than 24 months, and in the placebogroup, 124 of 158 patients (78%) wereyounger than 24 months. Treatmentfailure occurred in 18.6% of thetreatment group and 44.9% in theplacebo group (NNT = 3.8, P < .001).Rescue treatment was needed in 6.8%of the treatment group and 33.5% ofplacebo patients (P < .001). Contra-lateral AOM developed in 8.2% and18.6% of treatment and placebogroups, respectively (P = .007). Therewas no significant difference in use ofanalgesic or antipyretic medicine,which was used in 84.2% of theamoxicillin-clavulanate group and85.9% of the placebo group.

Parents of child care attendees onplacebo missed more days of work(P = .005). Clinical failure ratesin children with unilateral AOMwere 17.2% in those treated withamoxicillin-clavulanate versus 42.7%in those treated with placebo; for bi-lateral AOM, clinical failure rateswere 21.7% for those treated withamoxicillin-clavulanate versus 46.3%in the placebo group. Reported ratesof treatment failure by day 8 were17.2% in the amoxicillin-clavulanategroup versus 42.7% in the placebogroup in children with unilateral AOMand 21.7% vs 46.3% among those withbilateral disease.

Adverse events, primarily diarrheaand/or rash, occurred in 52.8% of thetreatment group and 36.1% of theplacebo group (P = .003). Overallcondition as evaluated by the parentsand otoscopic appearance of the TMshowed a benefit of antibiotics overplacebo at the end of treatment visit(P < .001). Two placebo recipients

developed a severe infection; 1 de-veloped pneumococcal bacteremia, and1 developed radiographically confirmedpneumonia.

Most studies have excluded childrenwith severe illness and all excludethose with bacterial disease otherthan AOM (pneumonia, mastoiditis,meningitis, streptococcal pharyngitis).Kaleida et al91 compared myringotomyalone with myringotomy plus anti-biotics. Severe AOM was defined astemperature >39°C (102.2°F) or thepresence of severe otalgia. Patientswith severe AOM in the group thatreceived only myringotomy (withoutinitial antibiotics) had much worseoutcomes.

Initial Antibiotic Therapy

The rationale for antibiotic therapy inchildren with AOM is based on a highprevalence of bacteria in the accom-panying MEE.93 Bacterial and viralcultures of middle ear fluid collectedby tympanocentesis from childrenwith AOM showed 55% with bacteriaonly and 15% with bacteria and viru-ses. A beneficial effect of antibioticson AOM was first demonstrated in1968,94 followed by additional ran-domized trials and a meta-analysis95

showing a 14% increase in absoluterates of clinical improvement. Sys-tematic reviews of the literature pub-lished before 201121,59,62 revealedincreases of clinical improvementwith initial antibiotics of 6% to 12%.

Randomized clinical trials usingstringent diagnostic criteria for AOM inyoung children31,32 show differences inclinical improvement of 26% to 35%favoring initial antibiotic treatment ascompared with placebo. Greater ben-efit of immediate antibiotic therapywas observed for bilateral AOM62,96 orAOM associated with otorrhea.62 Inmost randomized trials,30,75,77,88,89 an-tibiotic therapy also decreased theduration of pain, analgesic use, or

school absence and parent daysmissed from work.

Children younger than 2 years withAOM may take longer to improveclinically than older children,57 andalthough they are more likely to ben-efit from antibiotics,31,32 AOM in manychildren will resolve without anti-biotics.62 A clinically significant benefitof immediate antibiotic therapy isobserved for bilateral AOM,62,96 Strep-tococcus pneumoniae infection, orAOM associated with otorrhea.62

Initial Observation for AOM

In systematic reviews of studies thatcompare antibiotic therapy for AOMwith placebo, a consistent finding hasbeen the overall favorable naturalhistory in control groups (NNT = 8–16).12,59,62,95 However, randomized tri-als in these reviews had varyingdiagnostic criteria that would havepermitted inclusion of some childrenwith OME, viral upper respiratoryinfections, or myringitis, therebylimiting the ability to apply thesefindings to children with a highlycertain AOM diagnosis. In more re-cent AOM studies31,32 using stringentdiagnostic criteria, approximatelyhalf of young children (younger than2–3 years) experienced clinical suc-cess when given placebo, but theeffect of antibiotic therapy was sub-stantially greater than suggested bystudies without precise diagnosis(NNT = 3–4).

Observation as initial management forAOM in properly selected childrendoes not increase suppurative com-plications, provided that follow-up isensured and a rescue antibiotic isgiven for persistent or worseningsymptoms.17 In contrast, withholdingof antibiotics in all children withAOM, regardless of clinical course,would risk a return to the suppu-rative complications observed in the

e978 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 16: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

preantibiotic era. At the populationlevel, antibiotics halve the risk ofmastoiditis after AOM, but the highNNT of approximately 4800 patients toprevent 1 case of mastoiditis pre-cludes a strategy of universal antibiotictherapy as a means to prevent mas-toiditis.83

The favorable natural history of AOMmakes it difficult to demonstrate sig-nificant differences in efficacy betweenantibiotic and placebo when a suc-cessful outcome is defined by relief orimprovement of presenting signs andsymptoms. In contrast, when otoscopicimprovement (resolution of TM bulg-ing, intense erythema, or both) is alsorequired for a positive outcome,31,32

the NNT is 3 to 4, compared with 8 to16 for symptom improvement alone inolder studies that used less precisediagnostic criteria. MEE, however, maypersist for weeks or months after anAOM episode and is not a criterion forotoscopic failure.

National guidelines for initial obser-vation of AOM in select children werefirst implemented in the Netherlands97

and subsequently in Sweden,98 Scot-land,99 the United States,1 the UnitedKingdom,100 and Italy.101 All includedobservation as an initial treatmentoption under specified circumstances.

In numerous studies, only approximatelyone-third of children initially observedreceived a rescue antibiotic for persis-tent or worsening AOM,30,32,76,81,89,102

suggesting that antibiotic use couldpotentially be reduced by 65% in eligiblechildren. Given the high incidence ofAOM, this reduction could help sub-stantially in curtailing antibiotic-relatedadverse events.

McCormick et al30 reported on 233patients randomly assigned to receiveimmediate antibiotics (amoxicillin, 90mg/kg/day) or to undergo watchfulwaiting. Criteria for inclusion weresymptoms of ear infection, otoscopicevidence of AOM, and nonsevere AOM

based on a 3-item symptom score(OM-3) and TM appearance based onan 8-item scale (OS-8). Primary out-comes were parent satisfaction withAOM care, resolution of AOM symptomsafter initial treatment, AOM failure andrecurrence, and nasopharyngeal car-riage of S pneumoniae strains resistantto antibiotics after treatment. The studywas confounded by including patientswho had received antibiotics in theprevious 30 days.

In the watchful waiting group, 66% ofchildren completed the study withoutantibiotics. There was no difference inparent satisfaction scores at day 12.A 5-item symptom score (ETG-5) wasassessed at days 0 to 10 by usingpatient diaries. Subjects receivingimmediate antibiotics resolved theirsymptoms faster than did subjectswho underwent watchful waiting (P =.004). For children younger than 2years, the difference was greater (P =.008). Otoscopic and tympanogramscores were also lower in the antibi-otic group as opposed to the watchfulwaiting group (P = .02 for otoscopicscore, P = .004 for tympanogram).Combining all ages, failure and re-currence rates were lower for theantibiotic group (5%) than for thewatchful waiting group (21%) at 12days. By day 30, there was no differ-ence in failure or recurrence for theantibiotic and watchful waiting groups(23% and 24%, respectively). The as-sociation between clinical outcomeand intervention group was not signifi-cantly different between age groups.Immediate antibiotics resulted in erad-ication of S pneumoniae carriage in themajority of children, but S pneumoniaestrains cultured from children in theantibiotic group at day 12 were morelikely to be multidrug resistant thanwere strains cultured from children inthe watchful waiting group.

The decision not to give initial antibi-otic treatment and observe should be

a joint decision of the clinician and theparents. In such cases, a system forclose follow-up and a means of be-ginning antibiotics must be in place ifsymptoms worsen or no improvementis seen in 48 to 72 hours.

Initial observation of AOM should bepart of a larger management strategythat includes analgesics, parent in-formation, and provisions for a rescueantibiotic. Education of parents shouldinclude an explanation about the self-limited nature of most episodes ofAOM, especially in children 2 years andolder; the importance of pain man-agement early in the course; and thepotential adverse effects of antibiotics.Such an approach can substantiallyreduce prescription fill rates for res-cue antibiotics.103

A critical component of any strategyinvolving initial observation for AOM isthe ability to provide a rescue antibi-otic if needed. This is often done byusing a “safety net” or a “wait-and-seeprescription,”76,102 in which theparent/caregiver is given an antibioticprescription during the clinical en-counter but is instructed to fill theprescription only if the child fails toimprove within 2 to 3 days or ifsymptoms worsen at any time. An al-ternative approach is not to providea written prescription but to instructthe parent/caregiver to call or returnif the child fails to improve within 2 to3 days or if symptoms worsen.

In one of the first major studies of ob-servation with a safety-net antibioticprescription (SNAP), Siegel et al102 en-rolled 194 patients with protocol de-fined AOM, of whom 175 completed thestudy. Eligible patients were givena SNAP with instructions to fill theprescription only if symptoms wors-ened or did not improve in 48 hours.The SNAP was valid for 5 days. Painmedicine was recommended to betaken as needed. A phone interview wasconducted 5 to 10 days after diagnosis.

PEDIATRICS Volume 131, Number 3, March 2013 e979

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 17: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

One hundred twenty of 175 families didnot fill the prescription. Reasons forfilling the prescription (more than 1reason per patient was acceptable)were as follows: continued pain, 23%;continued fever, 11%; sleep disruption,6%; missed days of work, 3%; misseddays of child care, 3%; and no reasongiven, 5%. One 16-month-old boy com-pleted observation successfully but 6weeks later developed AOM in the op-posite ear, was treated with antibiotics,and developed postauricular cellulitis.

In a similar study of a “wait-and-seeprescription” (WASP) in the emer-gency department, Spiro et al76 ran-domly assigned 283 patients to eithera WASP or standard prescription.Clinicians were educated on the 2004AAP diagnostic criteria and initialtreatment options for AOM; however,diagnosis was made at the discretionof the clinician. Patients were ex-cluded if they did not qualify for ob-servation per the 2004 guidelines. Theprimary outcome was whether theprescription was filled within 3 daysof diagnosis. Prescriptions were notfilled for 62% and 13% of the WASPand standard prescription patients,respectively (P < .001). Reasons forfilling the prescription in the WASPgroup were fever (60%), ear pain(34%), or fussy behavior (6%). No se-rious adverse events were reported.

Strategies to observe children with AOMwho are likely to improve on their ownwithout initial antibiotic therapyreduces common adverse effects ofantibiotics, such as diarrhea and di-aper dermatitis. In 2 trials, antibiotictherapy significantly increased the ab-solute rates of diarrhea by 10% to 20%and of diaper rash or dermatitis by 6%to 16%.31,32 Reduced antibiotic use mayalso reduce the prevalence of resis-tant bacterial pathogens. Multidrug-resistant S pneumoniae continues tobe a significant concern for AOM,despite universal immunization of

children in the United States withheptavalent pneumococcal conjugatevaccine.104,105 In contrast, countrieswith low antibiotic use for AOM havea low prevalence of resistant naso-pharyngeal pathogens in children.106

Key Action Statement 4A

Clinicians should prescribe amoxi-cillin for AOM when a decision

to treat with antibiotics has beenmade and the child has not re-

ceived amoxicillin in the past 30

days or the child does not have

concurrent purulent conjunctivitis

or the child is not allergic to

penicillin. (Evidence Quality:

Grade B, Rec. Strength: Recom-

mendation)

Key Action Statement 4B

Clinicians should prescribe an an-tibiotic with additional β-lactamasecoverage for AOM when a decisionto treat with antibiotics has beenmade and the child has received

amoxicillin in the past 30 days orhas concurrent purulent conjunc-tivitis or has a history of recurrentAOM unresponsive to amoxicillin.(Evidence Quality: Grade C, Rec.Strength: Recommendation)

Key Action Statement 4C

Clinicians should reassess the pa-tient if the caregiver reports thatthe child’s symptoms have wors-ened or failed to respond to the

initial antibiotic treatment within48 to 72 hours and determinewhether a change in therapy isneeded. (Evidence Quality: Grade B,Rec. Strength: Recommendation)

Key Action Statement Profile: KAS 4AAggregate evidence quality Grade B

Benefits Effective antibiotic for most children with AOM. Inexpensive, safe,acceptable taste, narrow antimicrobial spectrum.

Risks, harms, cost Ineffective against β-lactamase–producing organisms. Adverseeffects of amoxicillin.

Benefits-harms assessment Preponderance of benefit.Value judgments Better to use a drug that has reasonable cost, has an acceptable

taste, and has a narrow antibacterial spectrum.Intentional vagueness The clinician must determine whether the patient is truly

penicillin allergic.Role of patient preferences Should be considered if previous bad experience with

amoxicillin.Exclusions Patients with known penicillin allergy.Strength Recommendation.

Key Action Statement Profile: KAS 4BAggregate evidence quality Grade C

Benefits Successful treatment of β-lactamase–producing organisms.Risks, harms, cost Cost of antibiotic. Increased adverse effects.Benefits-harms assessment Preponderance of benefit.Value judgments Efficacy is more important than taste.Intentional vagueness None.Role of patient preferences Concern regarding side effects and taste.Exclusions Patients with known penicillin allergy.Strength Recommendation

e980 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 18: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Purpose of This Section

If an antibiotic will be used for treatmentof a child with AOM, whether as initialmanagement or after a period of ob-servation, the clinician must choose anantibiotic that will have a high likelihoodof being effective against the most likelyetiologic bacterial pathogens with con-siderations of cost, taste, convenience,and adverse effects. This section pro-poses first- and second-line antibioticsthat best meet these criteria whilebalancing potential benefits and harms.

Changes From AAP/AAFP 2004 AOMGuideline

Despite new data on the effect of PCV7and updated data on the in vitrosusceptibility of bacterial pathogensmost likely to cause AOM, the recom-mendations for the first-line antibioticremains unchanged from 2004. Thecurrent guideline contains revisedrecommendations regarding penicillinallergy based on new data. The in-crease of multidrug-resistant strainsof pneumococci is noted.

Microbiology

Microorganisms detected in the mid-dle ear during AOM include pathogenicbacteria, as well as respiratory viru-ses.107–110 AOM occurs most frequentlyas a consequence of viral upper re-spiratory tract infection,111–113 whichleads to eustachian tube inflammation/

dysfunction, negative middle ear pres-sure, and movement of secretionscontaining the upper respiratory tractinfection causative virus and patho-genic bacteria in the nasopharynx intothe middle ear cleft. By using com-prehensive and sensitive microbiologictesting, bacteria and/or viruses can bedetected in the middle ear fluid in upto 96% of AOM cases (eg, 66% bacteriaand viruses together, 27% bacteriaalone, and 4% virus alone).114 Studiesusing less sensitive or less compre-hensive microbiologic assays haveyielded less positive results for bacte-ria and much less positive results forviruses.115–117 The 3 most commonbacterial pathogens in AOM are Spneumoniae, nontypeable Haemophilusinfluenzae, and Moraxella catarrhalis.111

Streptococcus pyogenes (group Aβ-hemolytic streptococci) accountsfor less than 5% of AOM cases. Theproportion of AOM cases with patho-genic bacteria isolated from themiddle ear fluids varies dependingon bacteriologic techniques, trans-port issues, and stringency of AOMdefinition. In series of reports fromthe United States and Europe from1952–1981 and 1985–1992, the meanpercentage of cases with bacterialpathogens isolated from the middleear fluids was 69% and 72%, respec-tively.118 A large series from the Uni-versity of Pittsburgh Otitis MediaStudy Group reported bacterial path-ogens in 84% of the middle ear fluids

from 2807 cases of AOM.118 Studies thatapplied more stringent otoscopic cri-teria and/or use of bedside specimenplating on solid agar in addition toliquid transport media have a reportedrate of recovery of pathogenic bacteriafrom middle ear exudates rangingfrom 85% to 90%.119–121 When usingappropriate stringent diagnostic crite-ria, careful specimen handling, andsensitive microbiologic techniques, thevast majority of cases of AOM will in-volve pathogenic bacteria either aloneor in concert with viral pathogens.

Among AOM bacterial pathogens,S pneumoniae was the most frequentlycultured in earlier reports. Since thedebut and routine use of PCV7 in 2000,the ordinal frequency of these 3 majormiddle ear pathogens has evolved.105

In the first few years after PCV7 in-troduction, H influenzae became themost frequently isolated middle earpathogen, replacing S pneumoniae.122,123

Shortly thereafter, a shift to non-PCV7serotypes of S pneumoniae was de-scribed.124 Pichichero et al104 laterreported that 44% of 212 AOM casesseen in 2003–2006 were caused by Hinfluenzae, and 28% were caused by Spneumoniae, with a high proportion ofhighly resistant S pneumoniae. In thatstudy, a majority (77%) of cases in-volved recurrent disease or initialtreatment failure. A later report125 withdata from 2007 to 2009, 6 to 8 yearsafter the introduction of PCV7 in theUnited States, showed that PCV7 strainsof S pneumoniae virtually disappearedfrom the middle ear fluid of childrenwith AOM who had been vaccinated.However, the frequency of isolation ofnon-PCV7 serotypes of S pneumoniaefrom the middle ear fluid overall wasincreased; this has made isolation of Spneumoniae and H influenzae of chil-dren with AOM nearly equal.

In a study of tympanocentesis over 4respiratory tract illness seasons ina private practice, the percentage of

Key Action Statement Profile: KAS 4CAggregate evidence quality Grade B

Benefits Identify children who may have AOM caused by pathogensresistant to previous antibiotics.

Risks, harms, cost Cost. Time for patient and clinician to make change. Potentialneed for parenteral medication.

Benefit-harm assessment Preponderance of benefit.Value judgments None.Intentional vagueness “Reassess” is not defined. The clinician may determine the

method of assessment.Role of patient preferences Limited.Exclusions Appearance of TM improved.Strength Recommendation

PEDIATRICS Volume 131, Number 3, March 2013 e981

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 19: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

S pneumoniae initially decreased rel-ative to H influenzae. In 2005–2006(N = 33), 48% of bacteria were Spneumoniae, and 42% were H influ-enzae. For 2006–2007 (N = 37), thepercentages were equal at 41%. In2007–2008 (N = 34), 35% were S pneu-moniae, and 59% were H influenzae. In2008–2009 (N = 24), the percentageswere 54% and 38%, respectively, withan increase in intermediate and non-susceptible S pneumoniae.126 Data onnasopharyngeal colonization fromPCV7-immunized children with AOMhave shown continued presence of Spneumoniae colonization. Revai et al127

showed no difference in S pneumoniaecolonization rate among children withAOM who have been unimmunized,underimmunized, or fully immunizedwith PCV7. In a study during a viralupper respiratory tract infection, in-cluding mostly PCV7-immunized chil-dren (6 months to 3 years of age), Spneumoniae was detected in 45.5% of968 nasopharyngeal swabs, H influen-zae was detected in 32.4%, and Mcatarrhalis was detected in 63.1%.128

Data show that nasopharyngeal colo-nization of children vaccinated withPCV7 increasingly is caused by Spneumoniae serotypes not containedin the vaccine.129–132 With the use of therecently licensed 13-valent pneumo-coccal conjugate vaccine (PCV13),133

the patterns of nasopharyngeal colo-nization and infection with these com-mon AOM bacterial pathogens willcontinue to evolve.

Investigators have attempted to pre-dict the type of AOM pathogenic bac-teria on the basis of clinical severity,but results have not been promising.S pyogenes has been shown to occurmore commonly in older children134

and to cause a greater degree of in-flammation of the middle ear and TM,a greater frequency of spontaneousrupture of the TM, and more frequentprogression to acute mastoiditis

compared with other bacterialpathogens.134–136 As for clinical find-ings in cases with S pneumoniae andnontypeable H influenzae, some stud-ies suggest that signs and symptomsof AOM caused by S pneumoniae maybe more severe (fever, severe ear-ache, bulging TM) than those causedby other pathogens.44,121,137 Thesefindings were refuted by results of thestudies that found AOM caused bynontypeable H influenzae to be asso-ciated with bilateral AOM and moresevere inflammation of the TM.96,138

Leibovitz et al139 concluded, in a studyof 372 children with AOM caused byH influenzae (N = 138), S pneumoniae(N = 64), and mixed H influenzae andS pneumoniae (N = 64), that clinical/otologic scores could not discriminateamong various bacterial etiologies ofAOM. However, there were significantlydifferent clinical/otologic scores be-tween bacterial culture negative andculture positive cases. A study ofmiddle ear exudates of 82 cases ofbullous myringitis has shown a 97%bacteria positive rate, primarily Spneumoniae. In contrast to the pre-vious belief, mycoplasma is rarely thecausative agent in this condition.140

Accurate prediction of the bacterialcause of AOM on the basis of clinicalpresentation, without bacterial cul-ture of the middle ear exudates, is notpossible, but specific etiologies maybe predicted in some situations. Pub-lished evidence has suggested thatAOM associated with conjunctivitis(otitis-conjunctivitis syndrome) is morelikely caused by nontypeable H influ-enzae than by other bacteria.141–143

Bacterial Susceptibility toAntibiotics

Selection of antibiotic to treat AOM isbased on the suspected type of bac-teria and antibiotic susceptibility pat-tern, although clinical pharmacology

and clinical and microbiologic resultsand predicted compliance with thedrug are also taken into account. Earlystudies of AOM patients show that 19%of children with S pneumoniae and48% with H influenzae cultured oninitial tympanocentesis who were nottreated with antibiotic cleared thebacteria at the time of a second tym-panocentesis 2 to 7 days later.144 Ap-proximately 75% of children infectedwith M catarrhalis experienced bac-teriologic cure even after treatmentwith amoxicillin, an antibiotic to whichit is not susceptible.145,146

Antibiotic susceptibility of major AOMbacterial pathogens continues tochange, but data on middle earpathogens have become scanty be-cause tympanocentesis is not gener-ally performed in studies of childrenwith uncomplicated AOM. Most avail-able data come from cases of per-sistent or recurrent AOM. Current USdata from a number of centers indi-cates that approximately 83% and 87%of isolates of S pneumoniae from allage groups are susceptible to regular(40 mg/kg/day) and high-dose amoxi-cillin (80–90 mg/kg/day divided twicedaily), respectively.130,147–150 Pediatricisolates are smaller in number andinclude mostly ear isolates collec-ted from recurrent and persistentAOM cases with a high percentage ofmultidrug-resistant S pneumoniae,most frequently nonvaccine serotypesthat have recently increased in fre-quency and importance.104

High-dose amoxicillin will yield middleear fluid levels that exceed the mini-mum inhibitory concentration (MIC) ofall S pneumoniae serotypes that areintermediately resistant to penicillin(penicillin MICs, 0.12–1.0 μg/mL), andmany but not all highly resistantserotypes (penicillin MICs, ≥2 μg/mL)for a longer period of the dosing in-terval and has been shown to improvebacteriologic and clinical efficacy

e982 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 20: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

compared with the regular dose.151–153

Hoberman et al154 reported superior

efficacy of high-dose amoxicillin-

clavulanate in eradication of S pneu-

moniae (96%) from the middle ear at

days 4 to 6 of therapy compared with

azithromycin.

The antibiotic susceptibility pattern for

S pneumoniae is expected to continue

to evolve with the use of PCV13,

a conjugate vaccine containing 13serotypes of S pneumoniae.133,155,156

Widespread use of PCV13 could po-tentially reduce diseases caused bymultidrug-resistant pneumococcalserotypes and diminish the need forthe use of higher dose of amoxicillinor amoxicillin-clavulanate for AOM.

Some H influenzae isolates produceβ-lactamase enzyme, causing the iso-late to become resistant to penicillins.Current data from different studieswith non-AOM sources and geographiclocations that may not be comparableshow that 58% to 82% of H influenzaeisolates are susceptible to regular-and high-dose amoxicillin.130,147,148,157,158

These data represented a significantdecrease in β-lactamase–producing H

influenzae, compared with data repor-ted in the 2004 AOM guideline.

Nationwide data suggest that 100% of Mcatarrhalis derived from the upper re-spiratory tract are β-lactamase–positivebut remain susceptible to amoxicillin-clavulanate.159 However, the high rate ofspontaneous clinical resolution occur-ring in children with AOM attributableto M catarrhalis treated with amoxicil-lin reduces the concern for the first-linecoverage for this microorganism.145,146

AOM attributable to M catarrhalis rarelyprogresses to acute mastoiditis or in-tracranial infections.102,160,161

Antibiotic Therapy

High-dose amoxicillin is recommendedas the first-line treatment in mostpatients, although there are a numberof medications that are clinically ef-fective (Table 5). The justification forthe use of amoxicillin relates to itseffectiveness against common AOMbacterial pathogens as well as itssafety, low cost, acceptable taste, andnarrow microbiologic spectrum.145,151

In children who have taken amoxicillinin the previous 30 days, those withconcurrent conjunctivitis, or those

for whom coverage for β-lactamase–positive H influenzae and M catarrhalisis desired, therapy should be initiatedwith high-dose amoxicillin-clavulanate(90 mg/kg/day of amoxicillin, with 6.4mg/kg/day of clavulanate, a ratio ofamoxicillin to clavulanate of 14:1, givenin 2 divided doses, which is less likely tocause diarrhea than other amoxicillin-clavulanate preparations).162

Alternative initial antibiotics includecefdinir (14 mg/kg per day in 1 or 2doses), cefuroxime (30 mg/kg per dayin 2 divided doses), cefpodoxime (10mg/kg per day in 2 divided doses), orceftriaxone (50 mg/kg, administeredintramuscularly). It is important tonote that alternative antibiotics vary intheir efficacy against AOM pathogens.For example, recent US data on in vitrosusceptibility of S pneumoniae to cef-dinir and cefuroxime are 70% to 80%,compared with 84% to 92% amoxicillinefficacy.130,147–149 In vitro efficacy ofcefdinir and cefuroxime against Hinfluenzae is approximately 98%, com-pared with 58% efficacy of amoxicillinand nearly 100% efficacy of amoxicillin-clavulanate.158 A multicenter doubletympanocentesis open-label study of

TABLE 5 Recommended Antibiotics for (Initial or Delayed) Treatment and for Patients Who Have Failed Initial Antibiotic Treatment

Initial Immediate or Delayed Antibiotic Treatment Antibiotic Treatment After 48–72 h of Failure of Initial Antibiotic Treatment

Recommended First-lineTreatment

Alternative Treatment(if Penicillin Allergy)

RecommendedFirst-line Treatment

AlternativeTreatment

Amoxicillin (80–90 mg/ kg perday in 2 divided doses)

Cefdinir (14 mg/kg per dayin 1 or 2 doses)

Amoxicillin-clavulanatea (90 mg/kg perday of amoxicillin, with 6.4 mg/kgper day of clavulanate in 2divided doses)

Ceftriaxone, 3 d Clindamycin(30–40 mg/kg per day in 3divided doses), with or withoutthird-generation cephalosporin

or Cefuroxime (30 mg/kg perday in 2 divided doses)

or Failure of second antibiotic

Amoxicillin-clavulanatea (90 mg/kgper day of amoxicillin, with 6.4 mg/kgper day of clavulanate [amoxicillin toclavulanate ratio, 14:1] in 2divided doses)

Cefpodoxime (10 mg/kg perday in 2 divided doses)

Ceftriaxone (50 mg IM or IV for 3 d) Clindamycin (30–40 mg/kg per dayin 3 divided doses) plusthird-generation cephalosporin

Tympanocentesisb

Ceftriaxone (50 mg IM or IVper day for 1 or 3 d)

Consult specialistb

IM, intramuscular; IV, intravenous.a May be considered in patients who have received amoxicillin in the previous 30 d or who have the otitis-conjunctivitis syndrome.b Perform tympanocentesis/drainage if skilled in the procedure, or seek a consultation from an otolaryngologist for tympanocentesis/drainage. If the tympanocentesis revealsmultidrug-resistant bacteria, seek an infectious disease specialist consultation.

c Cefdinir, cefuroxime, cefpodoxime, and ceftriaxone are highly unlikely to be associated with cross-reactivity with penicillin allergy on the basis of their distinct chemical structures.See text for more information.

PEDIATRICS Volume 131, Number 3, March 2013 e983

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 21: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

cefdinir in recurrent AOM attributableto H influenzae showed eradication ofthe organism in 72% of patients.163

For penicillin-allergic children, recentdata suggest that cross-reactivityamong penicillins and cephalo-sporins is lower than historicallyreported.164–167 The previously citedrate of cross-sensitivity to cepha-losporins among penicillin-allergicpatients (approximately 10%) is likelyan overestimate. The rate was basedon data collected and reviewed duringthe 1960s and 1970s. A study analyzingpooled data of 23 studies, including2400 patients with reported history ofpenicillin allergy and 39 000 with nopenicillin allergic history concludedthat many patients who present witha history of penicillin allergy do nothave an immunologic reaction topenicillin.166 The chemical structureof the cephalosporin determines therisk of cross-reactivity between spe-cific agents.165,168 The degree ofcross-reactivity is higher betweenpenicillins and first-generation ceph-alosporins but is negligible with thesecond- and third-generation cepha-losporins. Because of the differencesin the chemical structures, cefdinir,cefuroxime, cefpodoxime, and cef-triaxone are highly unlikely to beassociated with cross-reactivity withpenicillin.165 Despite this, the JointTask Force on Practice Parameters;American Academy of Allergy, Asthmaand Immunology; American College ofAllergy, Asthma and Immunology; andJoint Council of Allergy, Asthma andImmunology169 stated that “cephalo-sporin treatment of patients witha history of penicillin allergy, selectingout those with severe reaction histo-ries, show a reaction rate of 0.1%.”They recommend a cephalosporin incases without severe and/or recentpenicillin allergy reaction historywhen skin test is not available.

Macrolides, such as erythromycin andazithromycin, have limited efficacyagainst both H influenzae and Spneumoniae.130,147–149 Clindamycinlacks efficacy against H influenzae.Clindamycin alone (30–40 mg/kg perday in 3 divided doses) may be usedfor suspected penicillin-resistant Spneumoniae; however, the drugwill likely not be effective for themultidrug-resistant serotypes.130,158,166

Several of these choices of antibioticsuspensions are barely palatable orfrankly offensive and may lead toavoidance behaviors or active rejectionby spitting out the suspension. Palat-ability of antibiotic suspensions hasbeen compared in many studies.170–172

Specific antibiotic suspensions such ascefuroxime, cefpodoxime, and clinda-mycin may benefit from adding taste-masking products, such as chocolateor strawberry flavoring agents, to ob-scure the initial bitter taste and theunpleasant aftertaste.172,173 In the pa-tient who is persistently vomiting orcannot otherwise tolerate oral medi-cation, even when the taste is masked,ceftriaxone (50 mg/kg, administeredintramuscularly in 1 or 2 sites in theanterior thigh, or intravenously) hasbeen demonstrated to be effective forthe initial or repeat antibiotic treat-ment of AOM.174,175 Although a singleinjection of ceftriaxone is approved bythe US FDA for the treatment of AOM,results of a double tympanocentesisstudy (before and 3 days after singledose ceftriaxone) by Leibovitz et al175

suggest that more than 1 ceftriaxonedose may be required to preventrecurrence of the middle ear infec-tion within 5 to 7 days after the initialdose.

Initial Antibiotic Treatment Failure

When antibiotics are prescribed forAOM, clinical improvement should benoted within 48 to 72 hours. During the24 hours after the diagnosis of AOM,

the child’s symptoms may worsenslightly. In the next 24 hours, thepatient’s symptoms should begin toimprove. If initially febrile, the tem-perature should decline within 48 to72 hours. Irritability and fussinessshould lessen or disappear, andsleeping and drinking patterns shouldnormalize.176,177 If the patient is notimproved by 48 to 72 hours, anotherdisease or concomitant viral infectionmay be present, or the causativebacteria may be resistant to the cho-sen therapy.

Some children with AOM and persis-tent symptoms after 48 to 72 hours ofinitial antibacterial treatment mayhave combined bacterial and viral in-fection, which would explain the per-sistence of ongoing symptoms despiteappropriate antibiotic therapy.109,178,179

Literature is conflicting on the corre-lation between clinical and bacterio-logic outcomes. Some studies reportgood correlation ranging from 86% to91%,180,181 suggesting continued pres-ence of bacteria in the middle ear ina high proportion of cases with per-sistent symptoms. Others report thatmiddle ear fluid from children withAOM in whom symptoms are persis-tent is sterile in 42% to 49% ofcases.123,182 A change in antibiotic maynot be required in some children withmild persistent symptoms.

In children with persistent, severesymptoms of AOM and unimprovedotologic findings after initial treat-ment, the clinician may considerchanging the antibiotic (Table 5). If thechild was initially treated with amoxicillinand failed to improve, amoxicillin-clavulanate should be used. Patientswho were given amoxicillin-clavulanateor oral third-generation cephalosporinsmay receive intramuscular ceftriaxone(50 mg/kg). In the treatment of AOMunresponsive to initial antibiotics, a 3-daycourse of ceftriaxone has been shown tobe better than a 1-day regimen.175

e984 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 22: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Although trimethoprim-sulfamethoxazoleand erythromycin-sulfisoxazole hadbeen useful as therapy for patientswith AOM, pneumococcal surveillancestudies have indicated that resis-tance to these 2 combination agentsis substantial.130,149,183 Therefore, whenpatients fail to improve while receiv-ing amoxicillin, neither trimethoprim-sulfamethoxazole184 nor erythromycin-sulfisoxazole is appropriate therapy.

Tympanocentesis should be consid-ered, and culture of middle ear fluidshould be performed for bacteriologicdiagnosis and susceptibility testingwhen a series of antibiotic drugs havefailed to improve the clinical condition.If tympanocentesis is not available,a course of clindamycin may be used,with or without an antibiotic that cov-ers nontypeable H influenzae and Mcatarrhalis, such as cefdinir, cefixime,or cefuroxime.

Because S pneumoniae serotype 19A isusually multidrug-resistant and maynot be responsive to clindamycin,104,149

newer antibiotics that are not ap-proved by the FDA for treatment ofAOM, such as levofloxacin or linezolid,may be indicated.185–187 Levofloxacin isa quinolone antibiotic that is not ap-proved by the FDA for use in children.Linezolid is effective against resistantGram-positive bacteria. It is not ap-proved by the FDA for AOM treatmentand is expensive. In children with re-peated treatment failures, every effortshould be made for bacteriologic di-agnosis by tympanocentesis withGram stain, culture, and antibioticsusceptibility testing of the organism(s) present. The clinician may con-sider consulting with pediatric medi-cal subspecialists, such as anotolaryngologist for possible tympano-centesis, drainage, and culture and aninfectious disease expert, before use ofunconventional drugs such as levo-floxacin or linezolid.

When tympanocentesis is not available, 1possible way to obtain information onthe middle ear pathogens and theirantimicrobial susceptibility is to obtaina nasopharyngeal specimen for bacterialculture. Almost all middle ear pathogensderive from the pathogens colonizing thenasopharynx, but not all nasopharyngealpathogens enter the middle ear to causeAOM. The positive predictive value ofnasopharyngeal culture during AOM(likelihood that bacteria cultured fromthe nasopharynx is the middle earpathogen) ranges from 22% to 44% forS pneumoniae, 50% to 71% for non-typeable H influenzae, and 17% to 19%for M catarrhalis. The negative pre-dictive value (likelihood that bacteria notfound in the nasopharynx are not AOMpathogens) ranges from 95% to 99% forall 3 bacteria.188,189 Therefore, if naso-pharyngeal culture is negative for spe-cific bacteria, that organism is likely notthe AOM pathogen. A negative culturefor S pneumoniae, for example, will helpeliminate the concern for multidrug-resistant bacteria and the need for un-conventional therapies, such as levo-floxacin or linezolid. On the other hand,if S pneumoniae is cultured from thenasopharynx, the antimicrobial suscep-tibility pattern can help guide treatment.

Duration of Therapy

The optimal duration of therapy forpatients with AOM is uncertain; theusual 10-day course of therapy wasderived from the duration of treatmentof streptococcal pharyngotonsillitis.Several studies favor standard 10-daytherapy over shorter courses for chil-dren younger than 2 years.162,190–194

Thus, for children younger than 2years and children with severe symp-toms, a standard 10-day course isrecommended. A 7-day course of oralantibiotic appears to be equally effec-tive in children 2 to 5 years of age withmild or moderate AOM. For children 6years and older with mild to moderate

symptoms, a 5- to 7-day course is ad-equate treatment.

Follow-up of the Patient With AOM

Once the child has shown clinical im-provement, follow-up is based on theusual clinical course of AOM. There islittle scientific evidence for a routine10- to 14-day reevaluation visit for allchildren with an episode of AOM. Thephysician may choose to reassesssome children, such as young childrenwith severe symptoms or recurrentAOM or when specifically requested bythe child’s parent.

Persistent MEE is common and can bedetected by pneumatic otoscopy (with orwithout verification by tympanometry)after resolution of acute symptoms. Twoweeks after successful antibiotic treat-ment of AOM, 60% to 70% of childrenhave MEE, decreasing to 40% at 1 monthand 10% to 25% at 3 months aftersuccessful antibiotic treatment.177,195

The presence of MEE without clinicalsymptoms is defined as OME. OME mustbe differentiated clinically from AOMand requires infrequent additionalmonitoring but not antibiotic therapy.Assurance that OME resolves is partic-ularly important for parents of childrenwith cognitive or developmental delaysthat may be affected adversely bytransient hearing loss associated withMEE. Detailed recommendations for themanagement of the child with OMEcan be found in the evidence-basedguideline from the AAP/AAFP/AmericanAcademy of Otolaryngology-Head andNeck Surgery published in 2004.84,85

Key Action Statement 5A

Clinicians should NOT prescribeprophylactic antibiotics to reducethe frequency of episodes of AOMin children with recurrent AOM.(Evidence Quality: Grade B, Rec.Strength: Recommendation)

PEDIATRICS Volume 131, Number 3, March 2013 e985

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 23: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Key Action Statement 5B

Clinicians may offer tympanostomytubes for recurrent AOM (3 epi-sodes in 6 months or 4 episodes in

1 year, with 1 episode in thepreceding 6 months). (Evidence

Quality: Grade B, Rec. Strength:

Option)

Purpose of This Section

Recurrent AOM has been defined as theoccurrence of 3 ormore episodes of AOMin a 6-month period or the occurrence of4 or more episodes of AOM in a 12-monthperiod that includes at least 1 episode inthe preceding 6 months.20 These epi-sodes should be well documented andseparate acute infections.11

Winter season, male gender, and pas-sive exposure to smoking have beenassociated with an increased likelihoodof recurrence. Half of children youngerthan 2 years treated for AOM will ex-perience a recurrence within 6 months.Symptoms that last more than 10 daysmay also predict recurrence.196

Changes From AAP/AAFP 2004 AOMGuideline

Recurrent AOM was not addressed inthe 2004 AOM guideline. This section

addresses the literature on recurrentAOM.

Antibiotic Prophylaxis

Long-term, low-dose antibiotic use, re-ferred to as antibiotic prophylaxis orchemoprophylaxis, has been used totreat children with recurrent AOM toprevent subsequent episodes.85 A 2006Cochrane review analyzed 16 studies oflong-term antibiotic use for AOM andfound such use prevented 1.5 episodesof AOM per year, reducing in half thenumber of AOM episodes during theperiod of treatment.197 Randomizedplacebo-controlled trials of prophylaxisreported a decrease of 0.09 episodesper month in the frequency of AOMattributable to therapy (approximately0.5 to 1.5 AOM episodes per year for95% of children). An estimated 5 chil-dren would need to be treated for 1

year to prevent 1 episode of OM. Theeffect may be more substantial forchildren with 6 or more AOM episodesin the preceding year.12

This decrease in episodes of AOM oc-curred only while the prophylactic an-tibiotic was being given. The modestbenefit afforded by a 6-month course ofantibiotic prophylaxis does not havelonger-lasting benefit after cessation oftherapy. Teele showed no differencesbetween children who received pro-phylactic antibiotics compared withthose who received placebo in AOMrecurrences or persistence of OME.198

Antibiotic prophylaxis is not appropriatefor children with long-term MEE or forchildren with infrequent episodes of AOM.The small reduction in frequency of AOMwith long-term antibiotic prophylaxismust be weighed against the cost of suchtherapy; the potential adverse effects ofantibiotics, principally allergic reactionand gastrointestinal tract consequences,such as diarrhea; and their contributionto the emergence of bacterial resistance.

Surgery for Recurrent AOM

The use of tympanostomy tubes fortreatment of ear disease in general, andfor AOM in particular, has been con-troversial.199 Most published studies ofsurgical intervention for OM focus onchildren with persistent MEE with orwithout AOM. The literature on surgeryfor recurrent AOM as defined hereis scant. A lack of consensus amongotolaryngologists regarding the role ofsurgery for recurrent AOM was reportedin a survey of Canadian otolaryngolo-gists in which 40% reported they would“never,” 30% reported they would“sometimes,” and 30% reported theywould “often or always” place tympa-nostomy tubes for a hypothetical 2-year-old child with frequent OM without per-sistent MEE or hearing loss.200

Tympanostomy tubes, however, remainwidely used in clinical practice for bothOME and recurrent OM.201 Recurrent

Key Action Statement Profile: KAS 5AAggregate evidence quality Grade B

Benefits No adverse effects from antibiotic. Reduces potential for developmentof bacterial resistance. Reduced costs.

Risks, harms, cost Small increase in episodes of AOM.Benefit-harm assessment Preponderance of benefit.Value judgments Potential harm outweighs the potential benefit.Intentional vagueness None.Role of patient preferences Limited.Exclusions Young children whose only alternative would be tympanostomy tubes.Strength Recommendation

Key Action Statement Profile: KAS 5BAggregate evidence quality Grade B

Benefits Decreased frequency of AOM. Ability to treat AOM with topicalantibiotic therapy.

Risks, harms, cost Risks of anesthesia or surgery. Cost. Scarring of TM, chronicperforation, cholesteatoma. Otorrhea.

Benefits-harms assessment Equilibrium of benefit and harm.Value judgments None.Intentional vagueness Option based on limited evidence.Role of patient preferences Joint decision of parent and clinician.Exclusions Any contraindication to anesthesia and surgery.Strength Option

e986 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 24: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

AOM remains a common indication forreferral to an otolaryngologist.

Three randomized controlled trials havecompared the number of episodes ofAOM after tympanostomy tube place-ment or no surgery.202 Two found sig-nificant improvement in mean numberof AOM episodes after tympanostomytubes during a 6-month follow-up pe-riod.203,204 One study randomly assignedchildren with recurrent AOM to groupsreceiving placebo, amoxicillin pro-phylaxis, or tympanostomy tubes andfollowed them for 2 years.205 Althoughprophylactic antibiotics reduced therate of AOM, no difference in numberof episodes of AOM was noted be-tween the tympanostomy tube groupand the placebo group over 2 years. ACochrane review of studies of tympa-nostomy tubes for recurrent AOM an-alyzed 2 studies204,206 that metinclusion criteria and found thattympanostomy tubes reduced thenumber of episodes of AOM by 1.5episodes in the 6 months after sur-gery.207 Tympanostomy tube insertionhas been shown to improve disease-specific quality-of-life measures inchildren with OM.208 One multicenter,nonrandomized observational studyshowed large improvements in adisease-specific quality-of-life instru-ment that measured psychosocialdomains of physical suffering, hearingloss, speech impairment, emotionaldistress, activity limitations, and care-giver concerns that are associated withear infections.209 These benefits oftympanostomy tubes have been dem-onstrated in mixed populations of chil-dren that include children with OME aswell as recurrent AOM.

Beyond the cost, insertion of tympa-nostomy tubes is associated witha small but finite surgical and anestheticrisk. A recent review looking at proto-cols to minimize operative risk reportedno major complications, such as sen-sorineural hearing loss, vascular injury,

or ossicular chain disruption, in 10 000tube insertions performed primarily byresidents, although minor complica-tions such as TM tears or displacedtubes in the middle ear were seen in0.016% of ears.210 Long-term sequelaeof tympanostomy tubes include TMstructural changes including focal at-rophy, tympanosclerosis, retractionpockets, and chronic perforation. Onemeta-analysis found tympanosclerosisin 32% of patients after placement oftympanostomy tubes and chronic per-forations in 2.2% of patients who hadshort-term tubes and 16.6% of patientswith long-term tubes.211

Adenoidectomy, without myringotomyand/or tympanostomy tubes, did notreduce the number of episodes of AOM

when compared with chemoprophylaxisor placebo.212 Adenoidectomy aloneshould not be used for prevention ofAOM but may have benefit when per-formed with placement of tympanos-tomy tubes or in children with previoustympanostomy tube placement in OME.213

Prevention of AOM: Key ActionStatement 6A

Pneumococcal Vaccine

Clinicians should recommend pneu-mococcal conjugate vaccine to allchildren according to the scheduleof the Advisory Committee on Im-munization Practices, AAP, and AAFP.(Evidence Quality: Grade B, Rec.Strength: Strong Recommendation)

Key Action Statement 6B

Influenza Vaccine: Cliniciansshould recommend annual in-fluenza vaccine to all childrenaccording to the schedule of

the Advisory Committee on Im-munization Practices, AAP, andAAFP. (Evidence Quality: Grade B,Rec. Strength: Recommenda-tion)

Key Action Statement Profile: KAS 6AAggregate evidence quality Grade B

Benefits Reduced frequency of AOM attributable to vaccine serotypes.Reduced risk of serious pneumococcal systemic disease.

Risks, harms, cost Potential vaccine side effects. Cost of vaccine.Benefits-harms assessment Preponderance of benefit.Value judgments Potential vaccine adverse effects are minimal.Intentional vagueness None.Role of patient preferences Some parents may choose to refuse the vaccine.Exclusions Severe allergic reaction (eg, anaphylaxis) to any component of

pneumococcal vaccine or any diphtheria toxoid-containingvaccine.

Strength Strong Recommendation

Key Action Statement Profile: KAS 6BAggregate evidence quality Grade B

Benefits Reduced risk of influenza infection. Reduction in frequency of AOMassociated with influenza.

Risks, harms, cost Potential vaccine adverse effects. Cost of vaccine. Requires annualimmunization.

Benefits-harms assessment Preponderance of benefit.Value judgments Potential vaccine adverse effects are minimal.Intentional vagueness NoneRole of patient preferences Some parents may choose to refuse the vaccine.Exclusions See CDC guideline on contraindications (http://www.cdc.gov/flu/

professionals/acip/shouldnot.htm).Strength Recommendation

PEDIATRICS Volume 131, Number 3, March 2013 e987

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 25: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Key Action Statement 6C

Breastfeeding: Clinicians shouldencourage exclusive breastfeeding

for at least 6 months. (EvidenceQuality: Grade B, Rec. Strength:

Recommendation)

Key Action Statement 6D

Clinicians should encourageavoidance of tobacco smoke ex-

posure. (Evidence Quality: Grade

C, Rec. Strength: Recommenda-

tion)

Purpose of This Section

The 2004 AOM guideline noted data onimmunizations, breastfeeding, andlifestyle changes that would reduce therisk of acquiring AOM. This sectionaddresses new data published since2004.

Changes From AAP/AAFP 2004 AOMGuideline

PCV7 has been in use in the UnitedStates since 2000. PCV13 was introducedin the United States in 2010. The 10-valent pneumococcal nontypeable Hinfluenzae protein D-conjugate vaccinewas recently licensed in Europe for

prevention of diseases attributable toS pneumoniae and nontypeable H influ-enzae. Annual influenza immunization isnow recommended for all children 6months of age and older in the UnitedStates.214,215 Updated information re-garding these vaccines and their effecton the incidence of AOM is reviewed.

The AAP issued a new breastfeedingpolicy statement in February 2012.216

This guideline also includes a recom-mendation regarding tobacco smokeexposure. Bottle propping, pacifieruse, and child care are discussed, butno recommendations are made be-cause of limited evidence. The use of

xylitol, a possible adjunct to AOMprevention, is discussed; however, norecommendations are made.

Pneumococcal Vaccine

Pneumococcal conjugate vaccineshave proven effective in preventing OMcaused by pneumococcal serotypescontained in the vaccines. A meta-analysis of 5 studies with AOM as anoutcome determined that there isa 29% reduction in AOM caused by allpneumococcal serotypes among chil-dren who received PCV7 before 24months of age.217 Although the overallbenefit seen in clinical trials for allcauses of AOM is small (6%–7%),218–221

observational studies have shown thatmedical office visits for otitis werereduced by up to 40% comparingyears before and after introduction ofPCV7.222–224 Grijvala223 reported noeffect, however, among children firstvaccinated at older ages. Poehlinget al225 reported reductions of fre-quent AOM and PE tube use after in-troduction of PCV7. The observationsby some of greater benefit observedin the community than in clinical tri-als is not fully understood but may berelated to effects of herd immunity ormay be attributed to secular trends orchanges in AOM diagnosis patternsover time.223,226–229 In a 2009 Cochranereview,221 Jansen et al found that theoverall reduction in AOM incidencemay only be 6% to 7% but noted thateven that small rate may have publichealth relevance. O’Brien et al con-curred and noted in addition the po-tential for cost savings.230 There isevidence that serotype replacementmay reduce the long-term efficacy ofpneumococcal conjugate vaccinesagainst AOM,231 but it is possible thatnew pneumococcal conjugate vac-cines may demonstrate an increasedeffect on reduction in AOM.232–234 Dataon AOM reduction secondary to thePCV13 licensed in the United States in2010 are not yet available.

Key Action Statement Profile: KAS 6CAggregate evidence quality Grade B

Benefits May reduce the risk of early AOM. Multiple benefits of breastfeedingunrelated to AOM.

Risk, harm, cost NoneBenefit-harm assessment Preponderance of benefit.Value judgments The intervention has value unrelated to AOM prevention.Intentional vagueness NoneRole of patient preferences Some parents choose to feed formula.Exclusions NoneStrength Recommendation

Key Action Statement Profile: KAS 6DAggregate evidence quality Grade C

Benefits May reduce the risk of AOM.Risks, harms, cost NoneBenefits-harms assessment Preponderance of benefit.Value judgments Avoidance of tobacco exposure has inherent value unrelated

to AOM.Intentional vagueness NoneRole of patient preferences Many parents/caregivers choose not to stop smoking. Some

also remain addicted, and are unable to quit smoking.Exclusions NoneStrength Recommendation

e988 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 26: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

The H influenzae protein D-conjugatevaccine recently licensed in Europehas potential benefit of protectionagainst 10 serotypes of S pneumoniaeand nontypeable H influenzae.221,234

Influenza Vaccine

Most cases of AOM follow upper re-spiratory tract infections caused byviruses, including influenza viruses. Asmany as two-thirds of young childrenwith influenza may have AOM.235

Investigators have studied the efficacyof trivalent inactivated influenza vac-cine (TIV) and live-attenuated in-tranasal influenza vaccine (LAIV) inpreventing AOM. Many studies havedemonstrated 30% to 55% efficacy ofinfluenza vaccine in prevention ofAOM during the respiratory illnessseason.6,235–239 One study reported nobenefit of TIV in reducing AOM burden;however, 1 of the 2 respiratory illnessseasons during which this study wasconducted had a relatively low in-fluenza activity. A pooled analysis240 of8 studies comparing LAIV versus TIVor placebo241–248 showed a higher ef-ficacy of LAIV compared with bothplacebo and with TIV. Influenza vacci-nation is now recommended for allchildren 6 months of age and older inthe United States.214,215

Breastfeeding

Multiple studies provide evidence thatbreastfeeding for at least 4 to 6months reduces episodes of AOM andrecurrent AOM.249–253 Two cohortstudies, 1 retrospective study250 and 1prospective study,253 suggest a doseresponse, with some protection frompartial breastfeeding and the greatestprotection from exclusive breastfeed-ing through 6 months of age. In mul-tivariate analysis controlling forexposure to child care settings, therisk of nonrecurrent otitis is 0.61(95% confidence interval [CI]: 0.4–0.92)comparing exclusive breastfeeding

through 6 months of age with nobreastfeeding or breastfeeding lessthan 4 months. In a prospective co-hort, Scariatti253 found a significantdose-response effect. In this study, OMwas self-reported by parents. In asystematic review, McNiel et al254

found that when exclusive breast-feeding was set as the normativestandard, the recalculated odds ratios(ORs) revealed the risks of any for-mula use. For example, any formulause in the first 6 months of age wassignificantly associated with in-creased incidence of OM (OR: 1.78;95% CI: 1.19–2.70; OR: 4.55; 95% CI:1.64–12.50 in the available studies;pooled OR for any formula in the first3 months of age, 2.00; 95% CI: 1.40–2.78). A number of studies255–259

addressed the association of AOM andother infectious illness in infants withduration and exclusivity of breast-feeding, but all had limitations andnone had a randomized controlleddesign. However, taken together, theycontinue to show a protective effect ofexclusive breastfeeding. In all studies,there has been a predominance ofwhite subjects, and child care atten-dance and smoking exposure may nothave been completely controlled. Also,feeding methods were self-reported.

The consistent finding of a lower in-cidence of AOM and recurrent AOMwith increased breastfeeding supportsthe AAP recommendation to encourageexclusive breastfeeding for the first 6months of life and to continue for atleast the first year and beyond for aslong as mutually desired by motherand child.216

Lifestyle Changes

In addition to its many other bene-fits,260 eliminating exposure to passivetobacco smoke has been postulatedto reduce the incidence of AOM in in-fancy.252,261–264 Bottles and pacifiershave been associated with AOM.

Avoiding supine bottle feeding (“bottlepropping”) and reducing or eliminat-ing pacifier use in the second 6months of life may reduce AOM in-cidence.265–267 In a recent cohortstudy, pacifier use was associatedwith AOM recurrence.268

During infancy and early childhood,reducing the incidence of upper re-spiratory tract infections by alteringchild care-center attendance patternscan reduce the incidence of recurrentAOM significantly.249,269

Xylitol

Xylitol, or birch sugar, is chemicallya pentitol or 5-carbon polyol sugaralcohol. It is available as chewing gum,syrup, or lozenges. A 2011 Cochranereview270 examined the evidence forthe use of xylitol in preventing re-current AOM. A statistically significant25% reduction in the risk of occur-rence of AOM among healthy childrenat child care centers in the xylitolgroup compared with the controlgroup (relative risk: 0.75; 95% CI: 0.65to 0.88; RD: –0.07; 95% CI: –0.12 to–0.03) in the 4 studies met criteria foranalysis.271–274 Chewing gum and loz-enges containing xylitol appeared tobe more effective than syrup. Childrenyounger than 2 years, those at thegreatest risk of having AOM, cannotsafely use lozenges or chewing gum.Also, xylitol needs to be given 3 to 5times a day to be effective. It is noteffective for treating AOM and it mustbe taken daily throughout the re-spiratory illness season to have aneffect. Sporadic or as-needed use isnot effective.

Future Research

Despite advances in research partiallystimulated by the 2004 AOM guideline,there are still many unansweredclinical questions in the field. Followingare possible clinical research ques-tions that still need to be resolved.

PEDIATRICS Volume 131, Number 3, March 2013 e989

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 27: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Diagnosis

There will probably never be a goldstandard for diagnosis of AOM becauseof the continuum from OME to AOM.Conceivably, new techniques that couldbe used on the small amount of fluidobtained during tympanocentesiscould identify inflammatory markersin addition to the presence of bacteriaor viruses. However, performing tym-panocentesis studies on children withuncomplicated otitis is likely not fea-sible because of ethical and otherconsiderations.

Devices that more accurately identifythe presence of MEE and bulging thatare easier to use than tympanometryduring office visits would be welcome,especially in the difficult-to-examineinfant. Additional development of in-expensive, easy-to-use video pneu-matic otoscopes is still a goal.

Initial Treatment

The recent studies of Hoberman31 andTähtinen32 have addressed clinicaland TM appearance by using stringentdiagnostic criteria of AOM. However,the outcomes for less stringent di-agnostic criteria, a combination ofsymptoms, MEE, and TM appearancenot completely consistent with OMEcan only be inferred from earlierstudies that used less stringent cri-teria but did not specify outcomes forvarious grades of findings. Random-ized controlled trials on these lesscertain TM appearances using scalessimilar to the OS-8 scale35 couldclarify the benefit of initial antibioticsand initial observation for these lesscertain diagnoses. Such studies mustalso specify severity of illness, later-ality, and otorrhea.

Appropriate end points must beestablished. Specifically is the ap-pearance of the TM in patients withoutclinical symptoms at the end of a studysignificant for relapse, recurrence, or

persistent MEE. Such a study wouldrequire randomization of patientswith unimproved TM appearance tocontinued observation and antibioticgroups.

The most efficient and acceptablemethods of initial observation shouldcontinue to be studied balancing theconvenience and benefits with thepotential risks to the patient.

Antibiotics

Amoxicillin-clavulanate has a broaderspectrum than amoxicillin and may bea better initial antibiotic. However,because of cost and adverse effects,the subcommittee has chosen amoxi-cillin as first-line AOM treatment.Randomized controlled trials com-paring the 2 with adequate power todifferentiate clinical efficacy wouldclarify this choice. Stringent diagnosticcriteria should be the standard forthese studies. Antibiotic comparisonsfor AOM should now include an ob-servation arm for patients with non-severe illness to ensure a clinicalbenefit over placebo. Studies shouldalso have enough patients to showsmall but meaningful differences.

Although there have been studies onthe likelihood of resistant S pneumo-niae or H influenzae in children inchild care settings and with siblingsyounger than 5 years, studies are stillneeded to determine whether theseand other risk factors would indicatea need for different initial treatmentthan noted in the guideline.

New antibiotics that are safe andeffective are needed for use inAOM because of the development ofmultidrug-resistant organisms. Suchnew antibiotics must be tested againstthe currently available medications.

Randomized controlled trials usingdifferent durations of antibiotic ther-apy in different age groups are neededto optimize therapy with the possibility

of decreasing duration of antibioticuse. These would need to be per-formed initially with amoxicillin andamoxicillin-clavulanate but should alsobe performed for any antibiotic used inAOM. Again, an observation arm shouldbe included in nonsevere illness.

Recurrent AOM

There have been adequate studiesregarding prophylactic antibiotic usein recurrent AOM. More and bettercontrolled studies of tympanostomytube placement would help determineits benefit versus harm.

Prevention

There should be additional de-velopment of vaccines targeted atcommon organisms associated withAOM.275 Focused epidemiologic studieson the benefit of breastfeeding, spe-cifically addressing AOM prevention,including duration of breastfeedingand partial versus exclusive breast-feeding, would clarify what is nowa more general database. Likewise,more focused studies of the effects oflifestyle changes would help clarifytheir effect on AOM.

Complementary and AlternativeMedicine

There are no well-designed random-ized controlled trials of the usefulnessof complementary and alternativemedicine in AOM, yet a large number offamilies turn to these methods. Al-though most alternative therapies arerelatively inexpensive, some may becostly. Such studies should comparethe alternative therapy to observationrather than antibiotics and only use anantibiotic arm if the alternative ther-apy is shown to be better than ob-servation. Such studies should focuson children with less stringent criteriaof AOM but using the same descriptivecriteria for the patients as notedabove.

e990 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 28: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

DISSEMINATION OF GUIDELINES

An Institute of Medicine Report notesthat “Effective multifaceted imple-mentation strategies targeting bothindividuals and healthcare systemsshould be employed by implementersto promote adherence to trustworthy[clinical practice guidelines].”230

Many studies of the effect of clinicalpractice guidelines have been per-formed. In general, the studies showlittle overt change in practice aftera guideline is published. However, aswas seen after the 2004 AOM guideline,the number of visits for AOM and thenumber of prescriptions for antibioticsfor AOM had decreased publication.Studies of educational and dissemi-nation methods both at the practic-ing physician level and especiallyat the resident level need to beexamined.

SUBCOMMITTEE ON DIAGNOSIS ANDMANAGEMENT OF ACUTE OTITISMEDIAAllan S. Lieberthal, MD, FAAP (Chair, generalpediatrician, no conflicts)

Aaron E. Carroll, MD, MS, FAAP (Partnershipfor Policy Implementation [PPI] Informatician,general academic pediatrician, no conflicts)

Tasnee Chonmaitree, MD, FAAP (pediatricinfectious disease physician, no financial con-flicts; published research related to AOM)

Theodore G. Ganiats, MD (family physician,American Academy of Family Physicians, noconflicts)

Alejandro Hoberman, MD, FAAP (general ac-ademic pediatrician, no financial conflicts;published research related to AOM)

Mary Anne Jackson, MD, FAAP (pediatric in-fectious disease physician, AAP Committee onInfectious Disease, no conflicts)

Mark D. Joffe, MD, FAAP (pediatric emer-gency medicine physician, AAP Committee/Section on Pediatric Emergency Medicine, noconflicts)

Donald T. Miller, MD, MPH, FAAP (generalpediatrician, no conflicts)

Richard M. Rosenfeld, MD, MPH, FAAP (oto-laryngologist, AAP Section on Otolaryngology,Head and Neck Surgery, American Academy ofOtolaryngology-Head and Neck Surgery, no finan-cial conflicts; published research related to AOM)Xavier D. Sevilla, MD, FAAP (general pediat-rics, Quality Improvement Innovation Network,no conflicts)Richard H. Schwartz, MD, FAAP (general pe-diatrician, no financial conflicts; published re-search related to AOM)Pauline A. Thomas, MD, FAAP (epidemiologist,general pediatrician, no conflicts)David E. Tunkel, MD, FAAP, FACS (otolaryn-gologist, AAP Section on Otolaryngology, Headand Neck Surgery, periodic consultant toMedtronic ENT)

CONSULTANTRichard N. Shiffman, MD, FAAP, FACMI(informatician, guideline methodologist, gen-eral academic pediatrician, no conflicts)

STAFFCaryn Davidson, MAOversight by the Steering Committee on QualityImprovement and Management, 2009–2012

REFERENCES

1. American Academy of Pediatrics Sub-committee on Management of Acute OtitisMedia. Diagnosis and management ofacute otitis media. Pediatrics. 2004;113(5):1451–1465

2. Grijalva CG, Nuorti JP, Griffin MR. Antibi-otic prescription rates for acute re-spiratory tract infections in USambulatory settings. JAMA. 2009;302(7):758–766

3. McCaig LF, Besser RE, Hughes JM. Trendsin antimicrobial prescribing rates forchildren and adolescents. JAMA. 2002;287(23):3096–3102

4. Vernacchio L, Vezina RM, Mitchell AA.Management of acute otitis media byprimary care physicians: trends since therelease of the 2004 American Academy ofPediatrics/American Academy of FamilyPhysicians clinical practice guideline. Pe-diatrics. 2007;120(2):281–287

5. Coco A, Vernacchio L, Horst M, AndersonA. Management of acute otitis media afterpublication of the 2004 AAP and AAFPclinical practice guideline. Pediatrics.2010;125(2):214–220

6. Marchisio P, Mira E, Klersy C, et al. Medi-cal education and attitudes about acute

otitis media guidelines: a survey ofItalian pediatricians and otolaryn-gologists. Pediatr Infect Dis J. 2009;28(1):1–4

7. Arkins ER, Koehler JM. Use of the obser-vation option and compliance withguidelines in treatment of acute otitismedia. Ann Pharmacother. 2008;42(5):726–727

8. Flores G, Lee M, Bauchner H, Kastner B.Pediatricians’ attitudes, beliefs, andpractices regarding clinical practiceguidelines: a national survey. Pediatrics.2000;105(3 pt 1):496–501

9. Bluestone CD. Definitions, terminology,and classification. In: Rosenfeld RM,Bluestone CD, eds. Evidence-Based OtitisMedia. Hamilton, Canada: BC Decker;2003:120–135

10. Bluestone CD, Klein JO. Definitions, ter-minology, and classification. In: BluestoneCD, Klein JO, eds. Otitis Media in Infantsand Children. 4th ed. Hamilton, Canada:BC Decker; 2007:1–19

11. Dowell SF, Marcy MS, Phillips WR, et al.Otitis media: principles of judicious use ofantimicrobial agents. Pediatrics. 1998;101(suppl):165–171

12. Rosenfeld RM. Clinical pathway for acuteotitis media. In: Rosenfeld RM, BluestoneCD, eds. Evidence-Based Otitis Media. 2nded. Hamilton, Canada: BC Decker; 2003:280–302

13. Carlson LH, Carlson RD. Diagnosis. In:Rosenfeld RM, Bluestone CD, eds.Evidence-Based Otitis Media. Hamilton,Canada: BC Decker; 2003: 136–146

14. Bluestone CD, Klein JO. Diagnosis. In: OtitisMedia in Infants and Children. 4th ed.Hamilton, Canada: BC Decker; 2007:147–212

15. University of Oxford, Centre for EvidenceBased Medicine. Available at: www.cebm.net/index.aspx?o=1044. Accessed July 17,2012

16. American Academy of Pediatrics SteeringCommittee on Quality Improvement andManagement. Classifying recommendationsfor clinical practice guidelines. Pediatrics.2004;114(3):874–877

17. Marcy M, Takata G, Shekelle P, et al.Management of Acute Otitis Media. Evi-dence Report/Technology Assessment No.15. Rockville, MD: Agency for HealthcareResearch and Quality; 2000

PEDIATRICS Volume 131, Number 3, March 2013 e991

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 29: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

18. Chan LS, Takata GS, Shekelle P, Morton SC,Mason W, Marcy SM. Evidence assess-ment of management of acute otitis me-dia: II. Research gaps and priorities forfuture research. Pediatrics. 2001;108(2):248–254

19. Takata GS, Chan LS, Shekelle P, Morton SC,Mason W, Marcy SM. Evidence assessmentof management of acute otitis media: I.The role of antibiotics in treatment ofuncomplicated acute otitis media. Pediat-rics. 2001;108(2):239–247

20. Shekelle PG, Takata G, Newberry SJ, et al.Management of Acute Otitis Media: Up-date. Evidence Report/Technology Assess-ment No. 198. Rockville, MD: Agency forHealthcare Research and Quality; 2010

21. Coker TR, Chan LS, Newberry SJ, et al.Diagnosis, microbial epidemiology, andantibiotic treatment of acute otitis mediain children: a systematic review. JAMA.2010;304(19):2161–2169

22. Jadad AR, Moore RA, Carroll D, et al.Assessing the quality of reports of ran-domized clinical trials: is blinding neces-sary? Control Clin Trials. 1996;17(1):1–12

23. Whiting P, Rutjes AW, Reitsma JB, BossuytPM, Kleijnen J. The development of QUADAS:a tool for the quality assessment of studiesof diagnostic accuracy included in sys-tematic reviews. BMC Med Res Methodol.2003;3:25

24. Guyatt GH, Oxman AD, Vist GE, et al; GRADEWorking Group. GRADE: an emerging con-sensus on rating quality of evidence andstrength of recommendations. BMJ. 2008;336(7650):924–926

25. Hoffman RN, Michel G, Rosenfeld RM,Davidson C. Building better guidelineswith BRIDGE-Wiz: development and evalu-ation of a software assistant to promoteclarity, transparency, and implement-ability. J Am Med Inform Assoc. 2012;19(1):94–101

26. Kalu SU, Ataya RS, McCormick DP, Patel JA,Revai K, Chonmaitree T. Clinical spectrumof acute otitis media complicating upperrespiratory tract viral infection. PediatrInfect Dis J. 2011;30(2):95–99

27. Block SL, Harrison CJ. Diagnosis andManagement of Acute Otitis Media. 3rded. Caddo, OK: Professional Communica-tions; 2005:48–50

28. Wald ER. Acute otitis media: more troublewith the evidence. Pediatr Infect Dis J.2003;22(2):103–104

29. Paradise JL, Rockette HE, Colborn DK,et al. Otitis media in 2253 Pittsburgh-areainfants: prevalence and risk factors dur-ing the first two years of life. Pediatrics.1997;99(3):318–333

30. McCormick DP, Chonmaitree T, Pittman C,et al. Nonsevere acute otitis media:a clinical trial comparing outcomes ofwatchful waiting versus immediate anti-biotic treatment. Pediatrics. 2005;115(6):1455–1465

31. Hoberman A, Paradise JL, Rockette HE,et al. Treatment of acute otitis media inchildren under 2 years of age. N Engl JMed. 2011;364(2):105–115

32. Tähtinen PA, Laine MK, Huovinen P, JalavaJ, Ruuskanen O, Ruohola A. A placebo-controlled trial of antimicrobial treat-ment for acute otitis media. N Engl J Med.2011;364(2):116–126

33. Shaikh N, Hoberman A, Paradise JL, et al.Development and preliminary evaluationof a parent-reported outcome instrumentfor clinical trials in acute otitis media.Pediatr Infect Dis J. 2009;28(1):5–8

34. Laine MK, Tähtinen PA, Ruuskanen O,Huovinen P, Ruohola A. Symptoms orsymptom-based scores cannot predictacute otitis media at otitis-prone age.Pediatrics. 2010;125(5). Available at: www.pediatrics.org/cgi/content/full/125/5/e1154

35. Friedman NR, McCormick DP, Pittman C,et al. Development of a practical tool forassessing the severity of acute otitis me-dia. Pediatr Infect Dis J. 2006;25(2):101–107

36. Rothman R, Owens T, Simel DL. Does thischild have acute otitis media? JAMA. 2003;290(12):1633–1640

37. Niemela M, Uhari M, Jounio-Ervasti K,Luotonen J, Alho OP, Vierimaa E. Lack ofspecific symptomatology in children withacute otitis media. Pediatr Infect Dis J.1994;13(9):765–768

38. Heikkinen T, Ruuskanen O. Signs andsymptoms predicting acute otitis media.Arch Pediatr Adolesc Med. 1995;149(1):26–29

39. Ingvarsson L. Acute otalgia in children—findings and diagnosis. Acta PaediatrScand. 1982;71(5):705–710

40. Kontiokari T, Koivunen P, Niemelä M, PokkaT, Uhari M. Symptoms of acute otitis me-dia. Pediatr Infect Dis J. 1998;17(8):676–679

41. Wong DL, Baker CM. Pain in children:comparison of assessment scales. PediatrNurs. 1988;14(1):9–17

42. Shaikh N, Hoberman A, Paradise JL, et al.Responsiveness and construct validity ofa symptom scale for acute otitis media.Pediatr Infect Dis J. 2009;28(1):9–12

43. Karma PH, Penttilä MA, Sipilä MM, KatajaMJ. Otoscopic diagnosis of middle eareffusion in acute and non-acute otitismedia. I. The value of different otoscopic

findings. Int J Pediatr Otorhinolaryngol.1989;17(1):37–49

44. McCormick DP, Lim-Melia E, Saeed K,Baldwin CD, Chonmaitree T. Otitis media:can clinical findings predict bacterial orviral etiology? Pediatr Infect Dis J. 2000;19(3):256–258

45. Schwartz RH, Stool SE, Rodriguez WJ,Grundfast KM. Acute otitis media: towarda more precise definition. Clin Pediatr(Phila). 1981;20(9):549–554

46. Rosenfeld RM. Antibiotic prophylaxis forrecurrent acute otitis media. In: Alper CM,Bluestone CD, eds. Advanced Therapy ofOtitis Media. Hamilton, Canada: BCDecker; 2004

47. Paradise J, Bernard B, Colborn D, Smith C,Rockette H; Pittsburgh-area Child De-velopment/Otitis Media Study Group. Otitismedia with effusion: highly prevalent andoften the forerunner of acute otitis mediaduring the first year of life [abstract].Pediatr Res. 1993;33:121A

48. Roland PS, Smith TL, Schwartz SR, et al.Clinical practice guideline: cerumen im-paction. Otolaryngol Head Neck Surg.2008;139(3 suppl 2):S1–S21

49. Shaikh N, Hoberman A, Kaleida PH, PloofDL, Paradise JL. Videos in clinical medi-cine. Diagnosing otitis media—otoscopyand cerumen removal. N Engl J Med. 2010;362(20):e62

50. Pichichero ME. Diagnostic accuracy, tym-panocentesis training performance, andantibiotic selection by pediatric residentsin management of otitis media. Pediatrics.2002;110(6):1064–1070

51. Kaleida PH, Ploof DL, Kurs-Lasky M, et al.Mastering diagnostic skills: EnhancingProficiency in Otitis Media, a model fordiagnostic skills training. Pediatrics. 2009;124(4). Available at: www.pediatrics.org/cgi/content/full/124/4/e714

52. Kaleida PH, Ploof D. ePROM: EnhancingProficiency in Otitis Media. Pittsburgh, PA:University of Pittsburgh School of Medi-cine. Available at: http://pedsed.pitt.edu.Accessed December 31, 2011

53. Innovative Medical Education. A ViewThrough the Otoscope: DistinguishingAcute Otitis Media from Otitis Media withEffusion. Paramus, NJ: Innovative MedicalEducation; 2000

54. American Academy of Pediatrics. Sectionon Infectious Diseases. A view through theotoscope: distinguishing acute otitis mediafrom otitis media with effusion [video].Available at: http://www2.aap.org/sections/infectdis/video.cfm. Accessed January 20, 2012

55. Hayden GF, Schwartz RH. Characteristicsof earache among children with acute

e992 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 30: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

otitis media. Am J Dis Child. 1985;139(7):721–723

56. Schechter NL. Management of pain asso-ciated with acute medical illness. In:Schechter NL, Berde CB, Yaster M, eds.Pain in Infants, Children, and Adolescents.Baltimore, MD: Williams & Wilkins; 1993:537–538

57. Rovers MM, Glasziou P, Appelman CL, et al.Predictors of pain and/or fever at 3 to 7days for children with acute otitis medianot treated initially with antibiotics:a meta-analysis of individual patient data.Pediatrics. 2007;119(3):579–585

58. Burke P, Bain J, Robinson D, Dunleavey J.Acute red ear in children: controlled trialof nonantibiotic treatment in children:controlled trial of nonantibiotic treatmentin general practice. BMJ. 1991;303(6802):558–562

59. Sanders S, Glasziou PP, DelMar C, RoversM. Antibiotics for acute otitis media inchildren [review]. Cochrane DatabaseSyst Rev. 2009;(2):1–43

60. van Buchem FL, Dunk JH, van’t Hof MA.Therapy of acute otitis media: myringotomy,antibiotics, or neither? A double-blindstudy in children. Lancet. 1981;2(8252):883–887

61. Thalin A, Densert O, Larsson A, et al. Ispenicillin necessary in the treatment ofacute otitis media? In: Proceedings of theInternational Conference on Acute andSecretory Otitis Media. Part 1. Amster-dam, Netherlands: Kugler Publications;1986:441–446

62. Rovers MM, Glasziou P, Appelman CL, et al.Antibiotics for acute otitis media: an in-dividual patient data meta-analysis. Lan-cet. 2006;368(9545):1429–1435

63. Bertin L, Pons G, d’Athis P, et al. Arandomized, double-blind, multicentrecontrolled trial of ibuprofen versus acet-aminophen and placebo for symptoms ofacute otitis media in children. FundamClin Pharmacol. 1996;10(4):387–392

64. American Academy of Pediatrics.Committee on Psychosocial Aspects ofChild and Family Health; Task Force onPain in Infants, Children, and Adolescents.The assessment and management ofacute pain in infants, children, andadolescents. Pediatrics. 2001;108(3):793–797

65. Bolt P, Barnett P, Babl FE, Sharwood LN.Topical lignocaine for pain relief in acuteotitis media: results of a double-blindplacebo-controlled randomised trial.Arch Dis Child. 2008;93(1):40–44

66. Foxlee R, Johansson AC, Wejfalk J, DawkinsJ, Dooley L, Del Mar C. Topical analgesia for

acute otitis media. Cochrane Database SystRev. 2006;(3):CD005657

67. Hoberman A, Paradise JL, Reynolds EA,Urkin J. Efficacy of Auralgan for treatingear pain in children with acute otitismedia. Arch Pediatr Adolesc Med. 1997;151(7):675–678

68. Sarrell EM, Mandelberg A, Cohen HA. Effi-cacy of naturopathic extracts in themanagement of ear pain associated withacute otitis media. Arch Pediatr AdolescMed. 2001;155(7):796–799

69. Sarrell EM, Cohen HA, Kahan E. Naturo-pathic treatment for ear pain in children.Pediatrics. 2003;111(5 pt 1):e574–e579

70. Adam D, Federspil P, Lukes M, Petrowicz O.Therapeutic properties and tolerance ofprocaine and phenazone containing eardrops in infants and very young children.Arzneimittelforschung. 2009;59(10):504–512

71. Barnett ED, Levatin JL, Chapman EH, et al.Challenges of evaluating homeopathictreatment of acute otitis media. PediatrInfect Dis J. 2000;19(4):273–275

72. Jacobs J, Springer DA, Crothers D. Ho-meopathic treatment of acute otitis mediain children: a preliminary randomizedplacebo-controlled trial. Pediatr Infect DisJ. 2001;20(2):177–183

73. Rosenfeld RM, Bluestone CD. Clinical effi-cacy of surgical therapy. In: Rosenfeld RM,Bluestone CD, eds. Evidence-Based OtitisMedia. 2003. Hamilton, Canada: BC Decker;2003:227–240

74. Rosenfeld RM. Observation option toolkitfor acute otitis media. Int J Pediatr Oto-rhinolaryngol. 2001;58(1):1–8

75. Le Saux N, Gaboury I, Baird M, et al.A randomized, double-blind, placebo-controlled noninferiority trial of amoxi-cillin for clinically diagnosed acute otitismedia in children 6 months to 5 years ofage. CMAJ. 2005;172(3):335–341

76. Spiro DM, Tay KY, Arnold DH, Dziura JD,Baker MD, Shapiro ED. Wait-and-see pre-scription for the treatment of acute otitismedia: a randomized controlled trial.JAMA. 2006;296(10):1235–1241

77. Neumark T, Mölstad S, Rosén C, et al.Evaluation of phenoxymethylpenicillintreatment of acute otitis media in chil-dren aged 2–16. Scand J Prim HealthCare. 2007;25(3):166–171

78. Chao JH, Kunkov S, Reyes LB, Lichten S,Crain EF. Comparison of two approachesto observation therapy for acute otitismedia in the emergency department.Pediatrics. 2008;121(5). Available at:www.pediatrics.org/cgi/content/full/121/5/e1352

79. Spurling GK, Del Mar CB, Dooley L, FoxleeR. Delayed antibiotics for respiratoryinfections. Cochrane Database Syst Rev.2007;(3):CD004417

80. Koopman L, Hoes AW, Glasziou PP, et al.Antibiotic therapy to prevent the de-velopment of asymptomatic middle eareffusion in children with acute otitismedia: a meta-analysis of individual pa-tient data. Arch Otolaryngol Head NeckSurg. 2008;134(2):128–132

81. Marchetti F, Ronfani L, Nibali SC, TamburliniG; Italian Study Group on Acute Otitis Me-dia. Delayed prescription may reduce theuse of antibiotics for acute otitis media:a prospective observational study in pri-mary care. Arch Pediatr Adolesc Med. 2005;159(7):679–684

82. Ho D, Rotenberg BW, Berkowitz RG. Therelationship between acute mastoiditisand antibiotic use for acute otitis mediain children. Arch Otolaryngol Head NeckSurg. 2008;34(1):45–48

83. Thompson PL, Gilbert RE, Long PF, SaxenaS, Sharland M, Wong IC. Effect of anti-biotics for otitis media on mastoiditis inchildren: a retrospective cohort studyusing the United Kingdom general prac-tice research database. Pediatrics. 2009;123(2):424–430

84. American Academy of Family Physicians;American Academy of Otolaryngology-Head and Neck Surgery; American Acad-emy of Pediatrics Subcommittee on OtitisMedia With Effusion. Otitis media with ef-fusion. Pediatrics. 2004;113(5):1412–1429

85. Rosenfeld RM, Culpepper L, Doyle KJ, et al;American Academy of Pediatrics Sub-committee on Otitis Media with Effusion;American Academy of Family Physicians;American Academy of Otolaryngology—Head and Neck Surgery. Clinical practiceguideline: otitis media with effusion. Oto-laryngol Head Neck Surg. 2004;130(suppl 5):S95–S118

86. Appelman CL, Claessen JQ, Touw-Otten FW,Hordijk GJ, de Melker RA. Co-amoxiclav inrecurrent acute otitis media: placebocontrolled study. BMJ. 1991;303(6815):1450–1452

87. Burke P, Bain J, Robinson D, Dunleavey J.Acute red ear in children: controlled trialof nonantibiotic treatment in children:controlled trial of nonantibiotic treatmentin general practice. BMJ. 1991;303(6802):558–562

88. van Balen FA, Hoes AW, Verheij TJ, deMelker RA. Primary care based random-ized, double blind trial of amoxicillin ver-sus placebo in children aged under 2years. BMJ. 2000;320(7231):350–354

PEDIATRICS Volume 131, Number 3, March 2013 e993

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 31: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

89. Little P, Gould C, Williamson I, Moore M,Warner G, Dunleavey J. Pragmatic rando-mised controlled trial of two prescribingstrategies for childhood acute otitis me-dia. BMJ. 2001;322(7282):336–342

90. Mygind N, Meistrup-Larsen K-I, Thomsen J,Thomsen VF, Josefsson K, Sørensen H. Pen-icillin in acute otitis media: a double-blindplacebo-controlled trial. Clin Otolaryngol Al-lied Sci. 1981;6(1):5–13

91. Kaleida PH, Casselbrant ML, Rockette HE,et al. Amoxicillin or myringotomy or bothfor acute otitis media: results of a ran-domized clinical trial. Pediatrics. 1991;87(4):466–474

92. Shaikh N, Hoberman A, Paradise JL, et al.Responsiveness and construct validity ofa symptom scale for acute otitis media.Pediatr Infect Dis J. 2009;28(1):9–12

93. Heikkinen T, Chonmaitree T. Importance ofrespiratory viruses in acute otitis media.Clin Microbiol Rev. 2003;16(2):230–241

94. Halsted C, Lepow ML, Balassanian N,Emmerich J, Wolinsky E. Otitis media.Clinical observations, microbiology, andevaluation of therapy. Am J Dis Child.1968;115(5):542–551

95. Rosenfeld RM, Vertrees J, Carr J, et al.Clinical efficacy of antimicrobials foracute otitis media: meta-analysis of 5,400children from 33 randomized trials. JPediatr. 1994;124(3):355–367

96. McCormick DP, Chandler SM, ChonmaitreeT. Laterality of acute otitis media: differentclinical and microbiologic characteristics.Pediatr Infect Dis J. 2007;26(7):583–588

97. Appelman CLM, Bossen PC, Dunk JHM,Lisdonk EH, de Melker RA, van WeertHCPM. NHG Standard Otitis Media Acuta(Guideline on acute otitis media of theDutch College of General Practitioners).Huisarts Wet. 1990;33:242–245

98. Swedish Medical Research Council.Treatment for acute inflammation of themiddle ear: consensus statement. Stock-holm, Sweden: Swedish Medical ResearchCouncil; 2000. Available at: http://soapimg.icecube.snowfall.se/strama/Konsensut_ora_eng.pdf. Accessed July 18, 2012

99. Scottish Intercollegiate Guideline Net-work. Diagnosis and management ofchildhood otitis media in primary care.Edinburgh, Scotland: Scottish Inter-collegiate Guideline Network; 2000.Available at: www.sign.ac.uk/guidelines/fulltext/66/index.html. Accessed July 18,2012

100. National Institute for Health and ClinicalExcellence, Centre for Clinical Practice.Respiratory tract infections—antibioticprescribing: prescribing of antibiotics for

self-limiting respiratory tract infections inadults and children in primary care. NICEClinical Guideline 69. London, United King-dom: National Institute for Health and Clini-cal Excellence; July 2008. Available at: www.nice.org.uk/CG069. Accessed July 18, 2012

101. Marchisio P, Bellussi L, Di Mauro G, et al.Acute otitis media: from diagnosis toprevention. Summary of the Italianguideline. Int J Pediatr Otorhinolaryngol.2010;74(11):1209–1216

102. Siegel RM, Kiely M, Bien JP, et al. Treat-ment of otitis media with observation anda safety-net antibiotic prescription. Pedi-atrics. 2003;112(3 pt 1):527–531

103. Pshetizky Y, Naimer S, Shvartzman P. Acuteotitis media—a brief explanation toparents and antibiotic use. Fam Pract.2003;20(4):417–419

104. Pichichero ME, Casey JR. Emergence ofa multiresistant serotype 19A pneumo-coccal strain not included in the 7-valentconjugate vaccine as an otopathogen inchildren. JAMA. 2007;298(15):1772–1778

105. Pichichero ME, Casey JR. Evolving micro-biology and molecular epidemiology ofacute otitis media in the pneumococcalconjugate vaccine era. Pediatr Infect Dis J.2007;26(suppl 10):S12–S16

106. Nielsen HUK, Konradsen HB, Lous J, Frimodt-Møller N. Nasopharyngeal pathogens inchildren with acute otitis media in a low-antibiotic use country. Int J Pediatr Oto-rhinolaryngol. 2004;68(9):1149–1155

107. Pitkäranta A, Virolainen A, Jero J, ArrudaE, Hayden FG. Detection of rhinovirus, re-spiratory syncytial virus, and coronavirusinfections in acute otitis media by reversetranscriptase polymerase chain reaction.Pediatrics. 1998;102(2 pt 1):291–295

108. Heikkinen T, Thint M, Chonmaitree T.Prevalence of various respiratory virusesin the middle ear during acute otitis me-dia. N Engl J Med. 1999;340(4):260–264

109. Chonmaitree T. Acute otitis media is nota pure bacterial disease. Clin Infect Dis.2006;43(11):1423–1425

110. Williams JV, Tollefson SJ, Nair S, ChonmaitreeT. Association of human metapneumoviruswith acute otitis media. Int J PediatrOtorhinolaryngol. 2006;70(7):1189–1193

111. Chonmaitree T, Heikkinen T. Role of viru-ses in middle-ear disease. Ann N Y AcadSci. 1997;830:143–157

112. Klein JO, Bluestone CD. Otitis media. In:Feigin RD, Cherry JD, Demmler-HarrisonGJ, Kaplan SL, eds. Textbook of PediatricInfectious Diseases. 6th ed. Philadelphia,PA: Saunders; 2009:216–237

113. Chonmaitree T, Revai K, Grady JJ, et al.Viral upper respiratory tract infection and

otitis media complication in young chil-dren. Clin Infect Dis. 2008;46(6):815–823

114. Ruohola A, Meurman O, Nikkari S, et al.Microbiology of acute otitis media in chil-dren with tympanostomy tubes: prevalencesof bacteria and viruses. Clin Infect Dis. 2006;43(11):1417–1422

115. Ruuskanen O, Arola M, Heikkinen T, ZieglerT. Viruses in acute otitis media: increasingevidence for clinical significance. PediatrInfect Dis J. 1991;10(6):425–427

116. Chonmaitree T. Viral and bacterial in-teraction in acute otitis media. PediatrInfect Dis J. 2000;19(suppl 5):S24–S30

117. Nokso-Koivisto J, Räty R, Blomqvist S, et al.Presence of specific viruses in the middleear fluids and respiratory secretions ofyoung children with acute otitis media. JMed Virol. 2004;72(2):241–248

118. Bluestone CD, Klein JO. Microbiology. In:Bluestone CD, Klein JO, eds. Otitis Media inInfants and Children. 4th ed. Hamilton,Canada: BC Decker; 2007:101–126

119. Del Beccaro MA, Mendelman PM, Inglis AF,et al. Bacteriology of acute otitis media:a new perspective. J Pediatr. 1992;120(1):81–84

120. Block SL, Harrison CJ, Hedrick JA, et al.Penicillin-resistant Streptococcus pneu-moniae in acute otitis media: risk factors,susceptibility patterns and antimicrobialmanagement. Pediatr Infect Dis J. 1995;14(9):751–759

121. Rodriguez WJ, Schwartz RH. Streptococ-cus pneumoniae causes otitis media withhigher fever and more redness of tym-panic membranes than Haemophilusinfluenzae or Moraxella catarrhalis.Pediatr Infect Dis J. 1999;18(10):942–944

122. Block SL, Hedrick J, Harrison CJ, et al.Community-wide vaccination with theheptavalent pneumococcal conjugate sig-nificantly alters the microbiology of acuteotitis media. Pediatr Infect Dis J. 2004;23(9):829–833

123. Casey JR, Pichichero ME. Changes in fre-quency and pathogens causing acute oti-tis media in 1995–2003. Pediatr Infect DisJ. 2004;23(9):824–828

124. McEllistrem MC, Adams JM, Patel K, et al.Acute otitis media due to penicillin-nonsusceptible Streptococcus pneumo-niae before and after the introduction ofthe pneumococcal conjugate vaccine. ClinInfect Dis. 2005;40(12):1738–1744

125. Casey JR, Adlowitz DG, Pichichero ME. Newpatterns in the otopathogens causingacute otitis media six to eight years afterintroduction of pneumococcal conjugatevaccine. Pediatr Infect Dis J. 2010;29(4):304–309

e994 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 32: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

126. Grubb MS, Spaugh DC. Microbiology ofacute otitis media, Puget Sound region,2005–2009. Clin Pediatr (Phila). 2010;49(8):727–730

127. Revai K, McCormick DP, Patel J, Grady JJ,Saeed K, Chonmaitree T. Effect of pneu-mococcal conjugate vaccine on nasopha-ryngeal bacterial colonization duringacute otitis media. Pediatrics. 2006;117(5):1823–1829

128. Pettigrew MM, Gent JF, Revai K, Patel JA,Chonmaitree T. Microbial interactionsduring upper respiratory tract infections.Emerg Infect Dis. 2008;14(10):1584–1591

129. O’Brien KL, Millar EV, Zell ER, et al. Effectof pneumococcal conjugate vaccine onnasopharyngeal colonization among im-munized and unimmunized children ina community-randomized trial. J Infect Dis.2007;196(8):1211–1220

130. Jacobs MR, Bajaksouzian S, Windau A,Good C. Continued emergence of non-vaccine serotypes of Streptococcus pneu-moniae in Cleveland. Proceedings of the49th Interscience Conference on Antimi-crobial Agents and Chemotherapy; 2009:G1-G1556

131. Hoberman A, Paradise JL, Shaikh N, et al.Pneumococcal resistance and serotype19A in Pittsburgh-area children with acuteotitis media before and after introductionof 7-valent pneumococcal polysaccharidevaccine. Clin Pediatr (Phila). 2011;50(2):114–120

132. Huang SS, Hinrichsen VL, Stevenson AE,et al. Continued impact of pneumococcalconjugate vaccine on carriage in youngchildren. Pediatrics. 2009;124(1). Availableat: www.pediatrics.org/cgi/content/full/124/1/e1

133. Centers for Disease Control and Pre-vention (CDC). Licensure of a 13-valentpneumococcal conjugate vaccine (PCV13)and recommendations for use amongchildren—Advisory Committee on Immu-nization Practices (ACIP), 2010. MMWRMorb Mortal Wkly Rep. 2010;59(9):258–261

134. Segal N, Givon-Lavi N, Leibovitz E, YagupskyP, Leiberman A, Dagan R. Acute otitis me-dia caused by Streptococcus pyogenes inchildren. Clin Infect Dis. 2005;41(1):35–41

135. Luntz M, Brodsky A, Nusem S, et al. Acutemastoiditis—the antibiotic era: a multi-center study. Int J Pediatr Otorhinolar-yngol. 2001;57(1):1–9

136. Nielsen JC. Studies on the Aetiology ofAcute Otitis Media. Copenhagen, Denmark:Ejnar Mundsgaard Forlag; 1945

137. Palmu AA, Herva E, Savolainen H, Karma P,Mäkelä PH, Kilpi TM. Association of clinicalsigns and symptoms with bacterial find-

ings in acute otitis media. Clin Infect Dis.2004;38(2):234–242

138. Leibovitz E, Asher E, Piglansky L, et al. Is bi-lateral acute otitis media clinically differentthan unilateral acute otitis media? PediatrInfect Dis J. 2007;26(7):589–592

139. Leibovitz E, Satran R, Piglansky L, et al.Can acute otitis media caused by Hae-mophilus influenzae be distinguishedfrom that caused by Streptococcus pneu-moniae? Pediatr Infect Dis J. 2003;22(6):509–515

140. Palmu AA, Kotikoski MJ, Kaijalainen TH,Puhakka HJ. Bacterial etiology of acutemyringitis in children less than two yearsof age. Pediatr Infect Dis J. 2001;20(6):607–611

141. Bodor FF. Systemic antibiotics for treat-ment of the conjunctivitis-otitis mediasyndrome. Pediatr Infect Dis J. 1989;8(5):287–290

142. Bingen E, Cohen R, Jourenkova N, GehannoP. Epidemiologic study of conjunctivitis-otitis syndrome. Pediatr Infect Dis J.2005;24(8):731–732

143. Barkai G, Leibovitz E, Givon-Lavi N, DaganR. Potential contribution by nontypableHaemophilus influenzae in protracted andrecurrent acute otitis media. Pediatr In-fect Dis J. 2009;28(6):466–471

144. Howie VM, Ploussard JH. Efficacy of fixedcombination antibiotics versus separatecomponents in otitis media. Effectivenessof erythromycin estrolate, triple sulfon-amide, ampicillin, erythromycin estolate-triple sulfonamide, and placebo in 280patients with acute otitis media under twoand one-half years of age. Clin Pediatr(Phila). 1972;11(4):205–214

145. Klein JO. Microbiologic efficacy ofantibacterial drugs for acute otitismedia. Pediatr Infect Dis J. 1993;12(12):973–975

146. Barnett ED, Klein JO. The problem of re-sistant bacteria for the management ofacute otitis media. Pediatr Clin North Am.1995;42(3):509–517

147. Tristram S, Jacobs MR, Appelbaum PC.Antimicrobial resistance in Haemophilusinfluenzae. Clin Microbiol Rev. 2007;20(2):368–389

148. Critchley IA, Jacobs MR, Brown SD, TraczewskiMM, Tillotson GS, Janjic N. Prevalence ofserotype 19A Streptococcus pneumoniaeamong isolates from U.S. children in2005≠2006 and activity of faropenem.Antimicrob Agents Chemother. 2008;52(7):2639–2643

149. Jacobs MR, Good CE, Windau AR, et al.Activity of ceftaroline against emergingserotypes of Streptococcus pneumoniae.

Antimicrob Agents Chemother. 2010;54(6):2716–2719

150. Jacobs MR. Antimicrobial-resistant Strep-tococcus pneumoniae: trends andmanagement. Expert Rev Anti Infect Ther.2008;6(5):619–635

151. Piglansky L, Leibovitz E, Raiz S, et al. Bac-teriologic and clinical efficacy of highdose amoxicillin for therapy of acute otitismedia in children. Pediatr Infect Dis J.2003;22(5):405–413

152. Dagan R, Johnson CE, McLinn S, et al.Bacteriologic and clinical efficacy ofamoxicillin/clavulanate vs. azithromycin inacute otitis media. Pediatr Infect Dis J.2000;19(2):95–104

153. Dagan R, Hoberman A, Johnson C, et al.Bacteriologic and clinical efficacy of highdose amoxicillin/clavulanate in childrenwith acute otitis media. Pediatr Infect DisJ. 2001;20(9):829–837

154. Hoberman A, Dagan R, Leibovitz E, et al.Large dosage amoxicillin/clavulanate,compared with azithromycin, for thetreatment of bacterial acute otitis mediain children. Pediatr Infect Dis J. 2005;24(6):525–532

155. De Wals P, Erickson L, Poirier B, Pépin J,Pichichero ME. How to compare the effi-cacy of conjugate vaccines to preventacute otitis media? Vaccine. 2009;27(21):2877–2883

156. Shouval DS, Greenberg D, Givon-Lavi N,Porat N, Dagan R. Serotype coverage ofinvasive and mucosal pneumococcal dis-ease in Israeli children younger than 3years by various pneumococcal conjugatevaccines. Pediatr Infect Dis J. 2009;28(4):277–282

157. Jones RN, Farrell DJ, Mendes RE, SaderHS. Comparative ceftaroline activity testedagainst pathogens associated withcommunity-acquired pneumonia: resultsfrom an international surveillance study. JAntimicrob Chemother. 2011;66(suppl 3):iii69–iii80

158. Harrison CJ, Woods C, Stout G, Martin B,Selvarangan R. Susceptibilities of Haemo-philus influenzae, Streptococcus pneumo-niae, including serotype 19A, and Moraxellacatarrhalis paediatric isolates from 2005to 2007 to commonly used antibiotics.J Antimicrob Chemother. 2009;63(3):511–519

159. Doern GV, Jones RN, Pfaller MA, Kugler K.Haemophilus influenzae and Moraxellacatarrhalis from patients with community-acquired respiratory tract infections: anti-microbial susceptibility patterns from theSENTRY antimicrobial Surveillance Pro-gram (United States and Canada, 1997).

PEDIATRICS Volume 131, Number 3, March 2013 e995

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 33: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

Antimicrob Agents Chemother. 1999;43(2):385–389

160. Nussinovitch M, Yoeli R, Elishkevitz K,Varsano I. Acute mastoiditis in children:epidemiologic, clinical, microbiologic, andtherapeutic aspects over past years. ClinPediatr (Phila). 2004;43(3):261–267

161. Roddy MG, Glazier SS, Agrawal D. Pediatricmastoiditis in the pneumococcal conju-gate vaccine era: symptom durationguides empiric antimicrobial therapy.Pediatr Emerg Care. 2007;23(11):779–784

162. Hoberman A, Paradise JL, Burch DJ, et al.Equivalent efficacy and reduced occur-rence of diarrhea from a new formulationof amoxicillin/clavulanate potassium (Au-gmentin) for treatment of acute otitismedia in children. Pediatr Infect Dis J.1997;16(5):463–470

163. Arguedas A, Dagan R, Leibovitz E, HobermanA, Pichichero M, Paris M. A multicenter, openlabel, double tympanocentesis study ofhigh dose cefdinir in children with acuteotitis media at high risk of persistent orrecurrent infection. Pediatr Infect Dis J.2006;25(3):211–218

164. Atanaskovi�c-Markovi�c M, Velickovi�c TC,Gavrovi�c-Jankulovi�c M, Vuckovi�c O,Nestorovi�c B. Immediate allergic reactionsto cephalosporins and penicillins andtheir cross-reactivity in children. PediatrAllergy Immunol. 2005;16(4):341–347

165. Pichichero ME. Use of selected cepha-losporins in penicillin-allergic patients:a paradigm shift. Diagn Microbiol InfectDis. 2007;57(suppl 3):13S–18S

166. Pichichero ME, Casey JR. Safe use of se-lected cephalosporins in penicillin-allergicpatients: a meta-analysis. OtolaryngolHead Neck Surg. 2007;136(3):340–347

167. DePestel DD, Benninger MS, Danziger L,et al. Cephalosporin use in treatment ofpatients with penicillin allergies. J AmPharm Assoc (2003). 2008;48(4):530–540

168. Fonacier L, Hirschberg R, Gerson S. Ad-verse drug reactions to a cephalosporinsin hospitalized patients with a history ofpenicillin allergy. Allergy Asthma Proc.2005;26(2):135–141

169. Joint Task Force on Practice Parameters;American Academy of Allergy, Asthma andImmunology; American College of Allergy,Asthma and Immunology; Joint Council ofAllergy, Asthma and Immunology. Drugallergy: an updated practice parameter.Ann Allergy Asthma Immunol. 2010;105(4):259–273

170. Powers JL, Gooch WM, III, Oddo LP. Compar-ison of the palatability of the oral suspen-sion of cefdinir vs. amoxicillin/clavulanatepotassium, cefprozil and azithromycin in

pediatric patients. Pediatr Infect Dis J. 2000;19(suppl 12):S174–S180

171. Steele RW, Thomas MP, Bégué RE. Compli-ance issues related to the selection ofantibiotic suspensions for children.Pediatr Infect Dis J. 2001;20(1):1–5

172. Steele RW, Russo TM, Thomas MP. Adher-ence issues related to the selection ofantistaphylococcal or antifungal antibioticsuspensions for children. Clin Pediatr(Phila). 2006;45(3):245–250

173. Schwartz RH. Enhancing children’s satis-faction with antibiotic therapy: a tastestudy of several antibiotic suspensions.Curr Ther Res. 2000;61(8):570–581

174. Green SM, Rothrock SG. Single-dose in-tramuscular ceftriaxone for acute otitismedia in children. Pediatrics. 1993;91(1):23–30

175. Leibovitz E, Piglansky L, Raiz S, Press J,Leiberman A, Dagan R. Bacteriologic andclinical efficacy of one day vs. three dayintramuscular ceftriaxone for treatmentof nonresponsive acute otitis media inchildren. Pediatr Infect Dis J. 2000;19(11):1040–1045

176. Rosenfeld RM, Kay D. Natural history ofuntreated otitis media. Laryngoscope.2003;113(10):1645–1657

177. Rosenfeld RM, Kay D. Natural history ofuntreated otitis media. In: Rosenfeld RM,Bluestone CD, eds. Evidence-Based OtitisMedia. 2nd ed. Hamilton, Canada: BCDecker; 2003:180–198

178. Arola M, Ziegler T, Ruuskanen O. Re-spiratory virus infection as a cause ofprolonged symptoms in acute otitis me-dia. J Pediatr. 1990;116(5):697–701

179. Chonmaitree T, Owen MJ, Howie VM. Re-spiratory viruses interfere with bacterio-logic response to antibiotic in childrenwith acute otitis media. J Infect Dis. 1990;162(2):546–549

180. Dagan R, Leibovitz E, Greenberg D,Yagupsky P, Fliss DM, Leiberman A. Earlyeradication of pathogens from middle earfluid during antibiotic treatment of acuteotitis media is associated with improvedclinical outcome. Pediatr Infect Dis J.1998;17(9):776–782

181. Carlin SA, Marchant CD, Shurin PA, JohnsonCE, Super DM, Rehmus JM. Host factorsand early therapeutic response in acuteotitis media. J Pediatr. 1991;118(2):178–183

182. Teele DW, Pelton SI, Klein JO. Bacteriologyof acute otitis media unresponsive to ini-tial antimicrobial therapy. J Pediatr. 1981;98(4):537–539

183. Doern GV, Pfaller MA, Kugler K, Freeman J,Jones RN. Prevalence of antimicrobial re-

sistance among respiratory tract isolatesof Streptococcus pneumoniae in NorthAmerica: 1997 results from the SENTRYantimicrobial surveillance program. ClinInfect Dis. 1998;27(4):764–770

184. Leiberman A, Leibovitz E, Piglansky L, et al.Bacteriologic and clinical efficacy oftrimethoprim-sulfamethoxazole for treat-ment of acute otitis media. Pediatr InfectDis J. 2001;20(3):260–264

185. Humphrey WR, Shattuck MH, Zielinski RJ,et al. Pharmacokinetics and efficacy oflinezolid in a gerbil model of Streptococ-cus pneumoniae-induced acute otitis me-dia. Antimicrob Agents Chemother. 2003;47(4):1355–1363

186. Arguedas A, Dagan R, Pichichero M, et al.An open-label, double tympanocentesisstudy of levofloxacin therapy in childrenwith, or at high risk for, recurrent orpersistent acute otitis media. Pediatr In-fect Dis J. 2006;25(12):1102–1109

187. Noel GJ, Blumer JL, Pichichero ME, et al.A randomized comparative study of levo-floxacin versus amoxicillin/clavulanate fortreatment of infants and young childrenwith recurrent or persistent acute otitismedia. Pediatr Infect Dis J. 2008;27(6):483–489

188. Howie VM, Ploussard JH. Simultaneousnasopharyngeal and middle ear exudateculture in otitis media. Pediatr Digest.1971;13:31–35

189. Gehanno P, Lenoir G, Barry B, Bons J,Boucot I, Berche P. Evaluation of naso-pharyngeal cultures for bacteriologic as-sessment of acute otitis media inchildren. Pediatr Infect Dis J. 1996;15(4):329–332

190. Cohen R, Levy C, Boucherat M, Langue J,de La Rocque F. A multicenter, randomized,double-blind trial of 5 versus 10 days ofantibiotic therapy for acute otitis media inyoung children. J Pediatr. 1998;133(5):634–639

191. Pessey JJ, Gehanno P, Thoroddsen E, et al.Short course therapy with cefuroximeaxetil for acute otitis media: results ofa randomized multicenter comparisonwith amoxicillin/clavulanate. Pediatr In-fect Dis J. 1999;18(10):854–859

192. Cohen R, Levy C, Boucherat M, et al. Fivevs. ten days of antibiotic therapy for acuteotitis media in young children. PediatrInfect Dis J. 2000;19(5):458–463

193. Pichichero ME, Marsocci SM, Murphy ML,Hoeger W, Francis AB, Green JL. A pro-spective observational study of 5-, 7-, and10-day antibiotic treatment for acute otitismedia. Otolaryngol Head Neck Surg. 2001;124(4):381–387

e996 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 34: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

194. Kozyrskyj AL, Klassen TP, Moffatt M, HarveyK. Short-course antibiotics for acute otitismedia. Cochrane Database Syst Rev. 2010;(9):CD001095

195. Shurin PA, Pelton SI, Donner A, Klein JO.Persistence of middle-ear effusion afteracute otitis media in children. N Engl JMed. 1979;300(20):1121–1123

196. Damoiseaux RA, Rovers MM, Van Balen FA,Hoes AW, de Melker RA. Long-term prog-nosis of acute otitis media in infancy:determinants of recurrent acute otitismedia and persistent middle ear effusion.Fam Pract. 2006;23(1):40–45

197. Leach AJ, Morris PS. Antibiotics for theprevention of acute and chronic suppu-rative otitis media in children. CochraneDatabase Syst Rev. 2006;(4):CD004401

198. Teele DW, Klein JO, Word BM, et al; GreaterBoston Otitis Media Study Group. Antimi-crobial prophylaxis for infants at risk forrecurrent acute otitis media. Vaccine.2000;19(suppl 1):S140–S143

199. Paradise JL. On tympanostomy tubes: ra-tionale, results, reservations, and recom-mendations. Pediatrics. 1977;60(1):86–90

200. McIsaac WJ, Coyte PC, Croxford R, AscheCV, Friedberg J, Feldman W. Otolaryngolo-gists’ perceptions of the indications fortympanostomy tube insertion in children.CMAJ. 2000;162(9):1285–1288

201. Casselbrandt ML. Ventilation tubes forrecurrent acute otitis media. In: Alper CM,Bluestone CD, eds. Advanced Therapy ofOtitis Media. Hamilton, Canada: BC Decker;2004:113–115

202. Shin JJ, Stinnett SS, Hartnick CJ. Pediatricrecurrent acute otitis media. In: Shin JJ,Hartnick CJ, Randolph GW, eds. Evidence-Based Otolaryngology. New York, NY:Springer; 2008:91–95

203. Gonzalez C, Arnold JE, Woody EA, et al.Prevention of recurrent acute otitis me-dia: chemoprophylaxis versus tympanos-tomy tubes. Laryngoscope. 1986;96(12):1330–1334

204. Gebhart DE. Tympanostomy tubes in theotitis media prone child. Laryngoscope.1981;91(6):849–866

205. Casselbrant ML, Kaleida PH, Rockette HE,et al. Efficacy of antimicrobial prophylaxisand of tympanostomy tube insertion forprevention of recurrent acute otitis me-dia: results of a randomized clinical trial.Pediatr Infect Dis J. 1992;11(4):278–286

206. El-Sayed Y. Treatment of recurrent acuteotitis media chemoprophylaxis versusventilation tubes. Aust J Otolaryngol. 1996;2(4):352–355

207. McDonald S, Langton Hewer CD, Nunez DA.Grommets (ventilation tubes) for re-

current acute otitis media in children.Cochrane Database Syst Rev. 2008;(4):CD004741

208. Rosenfeld RM, Bhaya MH, Bower CM, et al.Impact of tympanostomy tubes on childquality of life. Arch Otolaryngol Head NeckSurg. 2000;126(5):585–592

209. Witsell DL, Stewart MG, Monsell EM, et al.The Cooperative Outcomes Group for ENT:a multicenter prospective cohort study onthe outcomes of tympanostomy tubes forchildren with otitis media. OtolaryngolHead Neck Surg. 2005;132(2):180–188

210. Isaacson G. Six Sigma tympanostomy tubeinsertion: achieving the highest safetylevels during residency training. Otolar-yngol Head Neck Surg. 2008;139(3):353–357

211. Kay DJ, Nelson M, Rosenfeld RM. Meta-analysis of tympanostomy tube sequelae.Otolaryngol Head Neck Surg. 2001;124(4):374–380

212. Koivunen P, Uhari M, Luotonen J, et al.Adenoidectomy versus chemoprophylaxisand placebo for recurrent acute otitismedia in children aged under 2 years:randomised controlled trial. BMJ. 2004;328(7438):487

213. Rosenfeld RM. Surgical prevention of otitismedia. Vaccine. 2000;19(suppl 1):S134–S139

214. Centers for Disease Control and Pre-vention (CDC). Prevention and control ofinfluenza with vaccines: recommendationsof the Advisory Committee on Immuniza-tion Practices (ACIP), 2011. MMWR MorbMortal Wkly Rep. 2011;60(33):1128–1132

215. American Academy of Pediatrics Commit-tee on Infectious Diseases. Recommen-dations for prevention and control ofinfluenza in children, 2011–2012. Pediat-rics. 2011;128(4):813–825

216. Section on Breastfeeding. Breastfeedingand the use of human milk. Pediatrics.2012;129(3). Available at: www.pediatrics.org/cgi/content/full/129/3/e827

217. Pavia M, Bianco A, Nobile CG, Marinelli P,Angelillo IF. Efficacy of pneumococcalvaccination in children younger than 24months: a meta-analysis. Pediatrics. 2009;123(6). Available at: www.pediatrics.org/cgi/content/full/123/6/e1103

218. Eskola J, Kilpi T, Palmu A, et al; FinnishOtitis Media Study Group. Efficacy ofa pneumococcal conjugate vaccineagainst acute otitis media. N Engl J Med.2001;344(6):403–409

219. Black S, Shinefield H, Fireman B, et al;Northern California Kaiser PermanenteVaccine Study Center Group. Efficacy,safety and immunogenicity of heptavalent

pneumococcal conjugate vaccine inchildren. Pediatr Infect Dis J. 2000;19(3):187–195

220. Jacobs MR. Prevention of otitis media:role of pneumococcal conjugate vaccinesin reducing incidence and antibiotic re-sistance. J Pediatr. 2002;141(2):287–293

221. Jansen AG, Hak E, Veenhoven RH,Damoiseaux RA, Schilder AG, SandersEA. Pneumococcal conjugate vaccinesfor preventing otitis media. CochraneDatabase Syst Rev. 2009;(2):CD001480

222. Fireman B, Black SB, Shinefield HR, Lee J,Lewis E, Ray P. Impact of the pneumococ-cal conjugate vaccine on otitis media.Pediatr Infect Dis J. 2003;22(1):10–16

223. Grijalva CG, Poehling KA, Nuorti JP, et al.National impact of universal childhoodimmunization with pneumococcal conju-gate vaccine on otitis media. Pediatr InfectDis J. 2006;118(3):865–873

224. Zhou F, Shefer A, Kong Y, Nuorti JP. Trendsin acute otitis media-related health careutilization by privately insured youngchildren in the United States, 1997–2004.Pediatrics. 2008;121(2):253–260

225. Poehling KA, Szilagyi PG, Grijalva CG, et al.Reduction of frequent otitis media andpressure-equalizing tube insertions inchildren after introduction of pneumo-coccal conjugate vaccine. Pediatrics. 2007;119(4):707–715

226. Pelton SI. Prospects for prevention of oti-tis media. Pediatr Infect Dis J. 2007;26(suppl 10):S20–S22

227. Pelton SI, Leibovitz E. Recent advances inotitis media. Pediatr Infect Dis J. 2009;28(suppl 10):S133–S137

228. De Wals P, Erickson L, Poirier B, Pépin J,Pichichero ME. How to compare the effi-cacy of conjugate vaccines to preventacute otitis media? Vaccine. 2009;27(21):2877–2883

229. Plasschaert AI, Rovers MM, Schilder AG,Verheij TJ, Hak E. Trends in doctor con-sultations, antibiotic prescription, andspecialist referrals for otitis media inchildren: 1995–2003. Pediatrics. 2006;117(6):1879–1886

230. O’Brien MA, Prosser LA, Paradise JL, et al.New vaccines against otitis media: pro-jected benefits and cost-effectiveness. Pe-diatrics. 2009;123(6):1452–1463

231. Hanage WP, Auranen K, Syrjänen R, et al.Ability of pneumococcal serotypes andclones to cause acute otitis media: impli-cations for the prevention of otitis mediaby conjugate vaccines. Infect Immun. 2004;72(1):76–81

232. Prymula R, Peeters P, Chrobok V, et al.Pneumococcal capsular polysaccharides

PEDIATRICS Volume 131, Number 3, March 2013 e997

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 35: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

conjugated to protein D for prevention ofacute otitis media caused by both Strep-tococcus pneumoniae and non-typableHaemophilus influenzae: a randomiseddouble-blind efficacy study. Lancet. 2006;367(9512):740–748

233. Prymula R, Schuerman L. 10-valent pneu-mococcal nontypeable Haemophilus influ-enzae PD conjugate vaccine: Synflorix.Expert Rev Vaccines. 2009;8(11):1479–1500

234. Schuerman L, Borys D, Hoet B, Forsgren A,Prymula R. Prevention of otitis media: nowa reality? Vaccine. 2009;27(42):5748–5754

235. Heikkinen T, Ruuskanen O, Waris M, ZieglerT, Arola M, Halonen P. Influenza vaccinationin the prevention of acute otitis media inchildren. Am J Dis Child. 1991;145(4):445–448

236. Clements DA, Langdon L, Bland C, Walter E.Influenza A vaccine decreases the in-cidence of otitis media in 6- to 30-month-old children in day care. Arch PediatrAdolesc Med. 1995;149(10):1113–1117

237. Belshe RB, Gruber WC. Prevention of otitismedia in children with live attenu-ated influenza vaccine given intranasally.Pediatr Infect Dis J. 2000;19(suppl 5):S66–S71

238. Marchisio P, Cavagna R, Maspes B, et al.Efficacy of intranasal virosomal influenzavaccine in the prevention of recurrentacute otitis media in children. Clin InfectDis. 2002;35(2):168–174

239. Ozgur SK, Beyazova U, Kemaloglu YK, et al.Effectiveness of inactivated influenza vac-cine for prevention of otitis media inchildren. Pediatr Infect Dis J. 2006;25(5):401–404

240. Block SL, Heikkinen T, Toback SL, Zheng W,Ambrose CS. The efficacy of live attenu-ated influenza vaccine against influenza-associated acute otitis media in children.Pediatr Infect Dis J. 2011;30(3):203–207

241. Ashkenazi S, Vertruyen A, Arístegui J, et al;CAIV-T Study Group. Superior relative effi-cacy of live attenuated influenza vaccinecompared with inactivated influenza vac-cine in young children with recurrent re-spiratory tract infections. Pediatr InfectDis J. 2006;25(10):870–879

242. Belshe RB, Edwards KM, Vesikari T, et al;CAIV-T Comparative Efficacy Study Group.Live attenuated versus inactivated in-fluenza vaccine in infants and youngchildren [published correction appears inN Engl J Med. 2007;356(12):1283]. N EnglJ Med. 2007;356(7):685–696

243. Bracco Neto H, Farhat CK, Tregnaghi MW,et al; D153-P504 LAIV Study Group. Efficacyand safety of 1 and 2 doses of live atten-uated influenza vaccine in vaccine-naive

children. Pediatr Infect Dis J. 2009;28(5):365–371

244. Tam JS, Capeding MR, Lum LC, et al;Pan-Asian CAIV-T Pediatric Efficacy TrialNetwork. Efficacy and safety of a live atten-uated, cold-adapted influenza vaccine, tri-valent against culture-confirmed influenzain young children in Asia. Pediatr Infect DisJ. 2007;26(7):619–628

245. Vesikari T, Fleming DM, Aristegui JF, et al;CAIV-T Pediatric Day Care Clinical TrialNetwork. Safety, efficacy, and effective-ness of cold-adapted influenza vaccine-trivalent against community-acquired,culture-confirmed influenza in youngchildren attending day care. Pediatrics.2006;118(6):2298–2312

246. Forrest BD, Pride MW, Dunning AJ, et al.Correlation of cellular immune re-sponses with protection against culture-confirmed influenza virus in young chil-dren. Clin Vaccine Immunol. 2008;15(7):1042–1053

247. Lum LC, Borja-Tabora CF, Breiman RF, et al.Influenza vaccine concurrently adminis-tered with a combination measles,mumps, and rubella vaccine to youngchildren. Vaccine. 2010;28(6):1566–1574

248. Belshe RB, Mendelman PM, Treanor J, et al.The efficacy of live attenuated, cold-adapted,trivalent, intranasal influenzavirus vaccinein children. N Engl J Med. 1998;338(20):1405–1412

249. Daly KA, Giebink GS. Clinical epidemiologyof otitis media. Pediatr Infect Dis J. 2000;19(suppl 5):S31–S36

250. Duncan B, Ey J, Holberg CJ, Wright AL,Martinez FD, Taussig LM. Exclusive breast-feeding for at least 4 months protectsagainst otitis media. Pediatrics. 1993;91(5):867–872

251. Duffy LC, Faden H, Wasielewski R, Wolf J,Krystofik D. Exclusive breastfeeding pro-tects against bacterial colonization andday care exposure to otitis media. Pediatrics.1997;100(4). Available at: www.pediatrics.org/cgi/content/full/100/4/e7

252. Paradise JL. Short-course antimicrobialtreatment for acute otitis media: not bestfor infants and young children. JAMA.1997;278(20):1640–1642

253. Scariati PD, Grummer-Strawn LM, Fein SB.A longitudinal analysis of infant morbidityand the extent of breastfeeding in theUnited States. Pediatrics. 1997;99(6).Available at: www.pediatrics.org/cgi/content/full/99/6/e5

254. McNiel ME, Labbok MH, Abrahams SW.What are the risks associated with for-mula feeding? A re-analysis and review.Breastfeed Rev. 2010;18(2):25–32

255. Chantry CJ, Howard CR, Auinger P. Fullbreastfeeding duration and associateddecrease in respiratory tract infection inUS children. Pediatrics. 2006;117(2):425–432

256. Hatakka K, Piirainen L, Pohjavuori S,Poussa T, Savilahti E, Korpela R. Factorsassociated with acute respiratory illnessin day care children. Scand J Infect Dis.2010;42(9):704–711

257. Ladomenou F, Kafatos A, Tselentis Y,Galanakis E. Predisposing factors foracute otitis media in infancy. J Infect.2010;61(1):49–53

258. Ladomenou F, Moschandreas J, Kafatos A,Tselentis Y, Galanakis E. Protective effectof exclusive breastfeeding againstinfections during infancy: a prospectivestudy. Arch Dis Child. 2010;95(12):1004–1008

259. Duijts L, Jaddoe VW, Hofman A, Moll HA.Prolonged and exclusive breastfeedingreduces the risk of infectious diseases ininfancy. Pediatrics. 2010;126(1). Availableat: www.pediatrics.org/cgi/content/full/126/1/e18

260. Best D; Committee on EnvironmentalHealth; Committee on Native AmericanChild Health; Committee on Adolescence.From the American Academy of Pediatrics:technical report—secondhand and pre-natal tobacco smoke exposure. Pediatrics.2009;124(5). Available at: www.pediatrics.org/cgi/content/full/124/5/e1017

261. Etzel RA, Pattishall EN, Haley NJ, FletcherRH, Henderson FW. Passive smokingand middle ear effusion among childrenin day care. Pediatrics. 1992;90(2 pt 1):228–232

262. Ilicali OC, Keles N, De�ger K, Savas I. Re-lationship of passive cigarette smoking tootitis media. Arch Otolaryngol Head NeckSurg. 1999;125(7):758–762

263. Wellington M, Hall CB. Pacifier as a riskfactor for acute otitis media [letter]. Pe-diatrics. 2002;109(2):351–352, author re-ply 353

264. Kerstein R. Otitis media: prevention in-stead of prescription. Br J Gen Pract.2008;58(550):364–365

265. Brown CE, Magnuson B. On the physics ofthe infant feeding bottle and middle earsequela: ear disease in infants can beassociated with bottle feeding. Int JPediatr Otorhinolaryngol. 2000;54(1):13–20

266. Niemelä M, Pihakari O, Pokka T, Uhari M.Pacifier as a risk factor for acute otitismedia: a randomized, controlled trial ofparental counseling. Pediatrics. 2000;106(3):483–488

e998 FROM THE AMERICAN ACADEMY OF PEDIATRICS by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 36: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

267. Tully SB, Bar-Haim Y, Bradley RL. Abnormaltympanography after supine bottle feed-ing. J Pediatr. 1995;126(6):S105–S111

268. Rovers MM, Numans ME, Langenbach E,Grobbee DE, Verheij TJ, Schilder AG. Ispacifier use a risk factor for acute otitismedia? A dynamic cohort study. FamPract. 2008;25(4):233–236

269. Adderson EE. Preventing otitis media: medicalapproaches. Pediatr Ann. 1998;27(2):101–107

270. Azarpazhooh A, Limeback H, Lawrence HP,Shah PS. Xylitol for preventing acute otitismedia in children up to 12 years of age.

Cochrane Database Syst Rev. 2011;(11):CD007095

271. Hautalahti O, Renko M, Tapiainen T, Kontio-kari T, Pokka T, Uhari M. Failure of xylitolgiven three times a day for preventingacute otitis media. Pediatr Infect DisJ. 2007;26(5):423–427

272. Tapiainen T, Luotonen L, Kontiokari T,Renko M, Uhari M. Xylitol administeredonly during respiratory infections failed toprevent acute otitis media. Pediatrics.2002;109(2). Available at: www.pediatrics.org/cgi/content/full/109/2/e19

273. Uhari M, Kontiokari T, Koskela M, NiemeläM. Xylitol chewing gum in prevention ofacute otitis media: double blind rando-mised trial. BMJ. 1996;313(7066):1180–1184

274. Uhari M, Kontiokari T, Niemelä M. A noveluse of xylitol sugar in preventing acuteotitis media. Pediatrics. 1998;102(4 pt 1):879–884

275. O’Brien MA, Prosser LA, Paradise JL, et al.New vaccines against otitis media: pro-jected benefits and cost-effectiveness. Pe-diatrics. 2009;123(6):1452–1463

(Continued from first page)

All clinical practice guidelines from the American Academy of Pediatrics automatically expire 5 years after publication unless reaffirmed, revised, or retired at orbefore that time.

www.pediatrics.org/cgi/doi/10.1542/peds.2012-3488

doi:10.1542/peds.2012-3488

PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275).

Copyright © 2013 by the American Academy of Pediatrics

PEDIATRICS Volume 131, Number 3, March 2013 e999

FROM THE AMERICAN ACADEMY OF PEDIATRICS

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 37: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

DOI: 10.1542/peds.2012-3488 originally published online February 25, 2013; 2013;131;e964Pediatrics 

E. TunkelM. Rosenfeld, Xavier D. Sevilla, Richard H. Schwartz, Pauline A. Thomas and DavidAlejandro Hoberman, Mary Anne Jackson, Mark D. Joffe, Donald T. Miller, Richard

Allan S. Lieberthal, Aaron E. Carroll, Tasnee Chonmaitree, Theodore G. Ganiats,The Diagnosis and Management of Acute Otitis Media

ServicesUpdated Information &

http://pediatrics.aappublications.org/content/131/3/e964including high resolution figures, can be found at:

Referenceshttp://pediatrics.aappublications.org/content/131/3/e964#BIBLThis article cites 247 articles, 66 of which you can access for free at:

Permissions & Licensing

http://www.aappublications.org/site/misc/Permissions.xhtmlin its entirety can be found online at: Information about reproducing this article in parts (figures, tables) or

Reprintshttp://www.aappublications.org/site/misc/reprints.xhtmlInformation about ordering reprints can be found online:

by guest on April 9, 2020www.aappublications.org/newsDownloaded from

Page 38: CLINICAL PRACTICE GUIDELINE The Diagnosis and ......fewer antibiotic prescriptions written. Despite significant publicity and awareness of the 2004 AOM guideline, evidence shows that

DOI: 10.1542/peds.2012-3488 originally published online February 25, 2013; 2013;131;e964Pediatrics 

E. TunkelM. Rosenfeld, Xavier D. Sevilla, Richard H. Schwartz, Pauline A. Thomas and DavidAlejandro Hoberman, Mary Anne Jackson, Mark D. Joffe, Donald T. Miller, Richard

Allan S. Lieberthal, Aaron E. Carroll, Tasnee Chonmaitree, Theodore G. Ganiats,The Diagnosis and Management of Acute Otitis Media

http://pediatrics.aappublications.org/content/131/3/e964located on the World Wide Web at:

The online version of this article, along with updated information and services, is

ISSN: 1073-0397. 60007. Copyright © 2013 by the American Academy of Pediatrics. All rights reserved. Print the American Academy of Pediatrics, 141 Northwest Point Boulevard, Elk Grove Village, Illinois,has been published continuously since 1948. Pediatrics is owned, published, and trademarked by Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it

by guest on April 9, 2020www.aappublications.org/newsDownloaded from